10192393|t|A common human skin tumour is caused by activating mutations in beta-catenin. 10192393|a|WNT signalling orchestrates a number of developmental programs. In response to this stimulus, cytoplasmic beta-catenin (encoded by CTNNB1) is stabilized, enabling downstream transcriptional activation by members of the LEF/TCF family. One of the target genes for beta-catenin/TCF encodes c-MYC, explaining why constitutive activation of the WNT pathway can lead to cancer, particularly in the colon. Most colon cancers arise from mutations in the gene encoding adenomatous polyposis coli (APC), a protein required for ubiquitin-mediated degradation of beta-catenin, but a small percentage of colon and some other cancers harbour beta-catenin-stabilizing mutations. Recently, we discovered that transgenic mice expressing an activated beta-catenin are predisposed to developing skin tumours resembling pilomatricomas. Given that the skin of these adult mice also exhibits signs of de novo hair-follicle morphogenesis, we wondered whether human pilomatricomas might originate from hair matrix cells and whether they might possess beta-catenin-stabilizing mutations. Here, we explore the cell origin and aetiology of this common human skin tumour. We found nuclear LEF-1 in the dividing tumour cells, providing biochemical evidence that pilomatricomas are derived from hair matrix cells. At least 75% of these tumours possess mutations affecting the amino-terminal segment, normally involved in phosphorylation-dependent, ubiquitin-mediated degradation of the protein. This percentage of CTNNB1 mutations is greater than in all other human tumours examined thus far, and directly implicates beta-catenin/LEF misregulation as the major cause of hair matrix cell tumorigenesis in humans.. 10192393 15 26 skin tumour DiseaseClass D012878 10192393 443 449 cancer DiseaseClass D009369 10192393 483 496 colon cancers DiseaseClass D003110 10192393 539 565 adenomatous polyposis coli SpecificDisease D011125 10192393 567 570 APC SpecificDisease D011125 10192393 670 698 colon and some other cancers CompositeMention D003110|D009369 10192393 855 867 skin tumours DiseaseClass D012878 10192393 879 893 pilomatricomas SpecificDisease D018296 10192393 1021 1035 pilomatricomas SpecificDisease D018296 10192393 1210 1221 skin tumour DiseaseClass D012878 10192393 1262 1268 tumour Modifier D009369 10192393 1312 1326 pilomatricomas SpecificDisease D018296 10192393 1385 1392 tumours DiseaseClass D009369 10192393 1615 1622 tumours DiseaseClass D009369 10194428|t|HFE mutations analysis in 711 hemochromatosis probands: evidence for S65C implication in mild form of hemochromatosis. 10194428|a|Hereditary hemochromatosis (HH) is a common autosomal recessive genetic disorder of iron metabolism. The HFE candidate gene encoding an HLA class I-like protein involved in HH was identified in 1996. Two missense mutations have been described C282Y, accounting for 80% to 90% of HH chromosomes, and H63D, which is associated with a milder form of the disease representing 40% to 70% of non-C282Y HH chromosomes. We report here on the analysis of C282Y, H63D, and the 193A-- > T substitution leading to the S65C missense substitution in a large series of probands and controls. The results confirm that the C282Y substitution was the main mutation involved in hemochromatosis, accounting for 85% of carrier chromosomes, whereas the H63D substitution represented 39% of the HH chromosomes that did not carry the C282Y mutation. In addition, our screening showed that the S65C substitution was significantly enriched in probands with at least one chromosome without an assigned mutation. This substitution accounted for 7. 8% of HH chromosomes that were neither C282Y nor H63D. This enrichment of S65C among HH chromosomes suggests that the S65C substitution is associated with the mild form of hemochromatosis. 10194428 30 45 hemochromatosis Modifier D016399 10194428 102 117 hemochromatosis SpecificDisease D006432 10194428 119 145 Hereditary hemochromatosis SpecificDisease D006432 10194428 147 149 HH SpecificDisease D006432 10194428 163 199 autosomal recessive genetic disorder DiseaseClass D030342 10194428 292 294 HH SpecificDisease D006432 10194428 399 401 HH Modifier D006432 10194428 516 518 HH Modifier D006432 10194428 779 794 hemochromatosis SpecificDisease D006432 10194428 892 894 HH Modifier D006432 10194428 1146 1148 HH Modifier D006432 10194428 1225 1227 HH Modifier D006432 10194428 1312 1327 hemochromatosis SpecificDisease D006432 10196379|t|Germline BRCA1 alterations in a population-based series of ovarian cancer cases. 10196379|a|The objective of this study was to provide more accurate frequency estimates of breast cancer susceptibility gene 1 (BRCA1) germline alterations in the ovarian cancer population. To achieve this, we determined the prevalence of BRCA1 alterations in a population-based series of consecutive ovarian cancer cases. This is the first population-based ovarian cancer study reporting BRCA1 alterations derived from a comprehensive screen of the entire coding region. One hundred and seven ovarian cancer cases were analyzed for BRCA1 alterations using the RNase mismatch cleavage assay followed by direct sequencing. Two truncating mutations, 962del4 and 3600del11, were identified. Both patients had a family history of breast or ovarian cancer. Several novel as well as previously reported uncharacterized variants were also identified, some of which were associated with a family history of cancer. The frequency distribution of common polymorphisms was determined in the 91 Caucasian cancer cases in this series and 24 sister controls using allele-specific amplification. The rare form of the Q356R polymorphism was significantly (P = 0. 03) associated with a family history of ovarian cancer, suggesting that this polymorphism may influence ovarian cancer risk. In summary, our data suggest a role for some uncharacterized variants and rare forms of polymorphisms in determining ovarian cancer risk, and highlight the necessity to screen for missense alterations as well as truncating mutations in this population. 10196379 59 73 ovarian cancer Modifier D010051 10196379 161 174 breast cancer Modifier D001943 10196379 233 247 ovarian cancer Modifier D010051 10196379 371 385 ovarian cancer Modifier D010051 10196379 428 442 ovarian cancer Modifier D010051 10196379 564 578 ovarian cancer Modifier D010051 10196379 796 820 breast or ovarian cancer CompositeMention D001943|D010051 10196379 969 975 cancer DiseaseClass D009369 10196379 1063 1069 cancer Modifier D009369 10196379 1257 1271 ovarian cancer SpecificDisease D010051 10196379 1321 1335 ovarian cancer Modifier D010051 10196379 1459 1473 ovarian cancer Modifier D010051 10021369|t|Identification of APC2, a homologue of the adenomatous polyposis coli tumour suppressor. 10021369|a|The adenomatous polyposis coli (APC) tumour-suppressor protein controls the Wnt signalling pathway by forming a complex with glycogen synthase kinase 3beta (GSK-3beta), axin/conductin and betacatenin. Complex formation induces the rapid degradation of betacatenin. In colon carcinoma cells, loss of APC leads to the accumulation of betacatenin in the nucleus, where it binds to and activates the Tcf-4 transcription factor (reviewed in [1] [2]). Here, we report the identification and genomic structure of APC homologues. Mammalian APC2, which closely resembles APC in overall domain structure, was functionally analyzed and shown to contain two SAMP domains, both of which are required for binding to conductin. Like APC, APC2 regulates the formation of active betacatenin-Tcf complexes, as demonstrated using transient transcriptional activation assays in APC -/- colon carcinoma cells. Human APC2 maps to chromosome 19p13. 3. APC and APC2 may therefore have comparable functions in development and cancer. 10021369 43 76 adenomatous polyposis coli tumour Modifier D011125 10021369 93 132 adenomatous polyposis coli (APC) tumour Modifier D011125 10021369 357 372 colon carcinoma Modifier D003110 10021369 955 970 colon carcinoma Modifier D003110 10021369 1090 1096 cancer SpecificDisease D009369 100562|t|Familial deficiency of the seventh component of complement associated with recurrent bacteremic infections due to Neisseria. 100562|a|The serum of a 29-year old woman with a recent episode of disseminated gonococcal infection and a history of meningococcal meningitis and arthritis as a child was found to lack serum hemolytic complement activity. The seventh component of complement (C7) was not detected by functional or immunochemical assays, whereas other components were normal by hemolytic and immunochemical assessment. Her fresh serum lacked complement-mediated bactericidal activity against Neisseria gonorrhoeae, but the addition of fresh normal serum or purified C7 restored bactericidal activity as well as hemolytic activity. The absence of functional C7 activity could not be accounted for on the basis of an inhibitor. Opsonization and generation of chemotactic activity functioned normally. Complete absence of C7 was also found in one sibling who had the clinical syndrome of meningococcal meningitis and arthritis as a child and in this siblings clinically well eight-year-old son. HLA histocompatibility typing of the family members did not demonstrate evidence for genetic linkage of C7 deficiency with the major histocompatibility loci. This report represents the first cases of C7 deficiency associated with infectious complications and suggests that bactericidal activity may be important in host defense against bacteremic neisseria infections. 100562 0 58 Familial deficiency of the seventh component of complement SpecificDisease OMIM:610102 100562 85 123 bacteremic infections due to Neisseria DiseaseClass D016870 100562 183 216 disseminated gonococcal infection SpecificDisease D004673 100562 234 258 meningococcal meningitis SpecificDisease D008585 100562 263 272 arthritis DiseaseClass D001168 100562 734 758 absence of functional C7 Modifier OMIM:610102 100562 898 920 Complete absence of C7 SpecificDisease OMIM:610102 100562 984 1008 meningococcal meningitis SpecificDisease D008585 100562 1013 1022 arthritis DiseaseClass D001168 100562 1196 1209 C7 deficiency SpecificDisease OMIM:610102 100562 1292 1305 C7 deficiency SpecificDisease OMIM:610102 100562 1428 1459 bacteremic neisseria infections SpecificDisease D016870 10078749|t|GCH1 mutation in a patient with adult-onset oromandibular dystonia. 10078749|a|The authors report a mutation in exon 5 of GCH1 in a patient with adult-onset oromandibular dystonia and no obvious family history of dystonia. The patient responded positively to treatment with L-dopa. These findings demonstrate that GCH1 mutations must be considered even in patients with dystonic symptoms not typical of dopa-responsive dystonia. 10078749 44 66 oromandibular dystonia SpecificDisease D008538 10078749 146 168 oromandibular dystonia SpecificDisease D008538 10078749 202 210 dystonia DiseaseClass D004421 10078749 359 367 dystonic Modifier D004421 10078749 392 416 dopa-responsive dystonia SpecificDisease C538007 10085150|t|The hereditary hemochromatosis protein, HFE, specifically regulates transferrin-mediated iron uptake in HeLa cells. 10085150|a|HFE is the protein product of the gene mutated in the autosomal recessive disease hereditary hemochromatosis (Feder, J. N., Gnirke, A., Thomas, W., Tsuchihashi, Z., Ruddy, D. A., Basava, A., Dormishian, F., Domingo, R. J., Ellis, M. C., Fullan, A., Hinton, L. M., Jones, N. L., Kimmel, B. E., Kronmal, G. S., Lauer, P., Lee, V. K., Loeb, D. B., Mapa, F. A., McClelland, E., Meyer, N. C., Mintier, G. A., Moeller, N., Moore, T., Morikang, E., Prasss, C. E ., Quintana, L., Starnes, S. M ., Schatzman, R. C ., Brunke, K. J ., Drayna, D. T., Risch, N. J ., Bacon, B. R ., and Wolff, R. R . (1996) Nat. Genet. 13, 399-408). At the cell surface, HFE complexes with transferrin receptor (TfR), increasing the dissociation constant of transferrin (Tf) for its receptor 10-fold (Gross, C. N ., Irrinki, A., Feder, J. N ., and Enns, C. A . (1998) J. Biol. Chem. 273, 22068-22074; Feder, J. N., Penny, D. M., Irrinki, A., Lee, V. K., Lebron, J. A., Watson, N., Tsuchihashi, Z., Sigal, E., Bjorkman, P. J., and Schatzman, R. C. (1998) Proc. Natl . Acad. Sci. U S A 95, 1472-1477). HFE does not remain at the cell surface, but traffics with TfR to Tf-positive internal compartments (Gross et al., 1998). Using a HeLa cell line in which the expression of HFE is controlled by tetracycline, we show that the expression of HFE reduces 55Fe uptake from Tf by 33% but does not affect the endocytic or exocytic rates of TfR cycling. Therefore, HFE appears to reduce cellular acquisition of iron from Tf within endocytic compartments. HFE specifically reduces iron uptake from Tf, as non-Tf-mediated iron uptake from Fe-nitrilotriacetic acid is not altered. These results explain the decreased ferritin levels seen in our HeLa cell system and demonstrate the specific control of HFE over the Tf-mediated pathway of iron uptake. These results also have implications for the understanding of cellular iron homeostasis in organs such as the liver, pancreas, heart, and spleen that are iron loaded in hereditary hemochromatotic individuals lacking functional HFE. 10085150 4 30 hereditary hemochromatosis Modifier D006432 10085150 170 197 autosomal recessive disease DiseaseClass D030342 10085150 198 224 hereditary hemochromatosis SpecificDisease D006432 10085150 2094 2120 hereditary hemochromatotic Modifier D006432 10090880|t|Mutation and haplotype studies of familial Mediterranean fever reveal new ancestral relationships and evidence for a high carrier frequency with reduced penetrance in the Ashkenazi Jewish population. 10090880|a|Familial Mediterranean fever (FMF) is a recessive disorder characterized by episodes of fever with serositis or synovitis. The FMF gene (MEFV) was cloned recently, and four missense mutations were identified. Here we present data from non-Ashkenazi Jewish and Arab patients in whom we had not originally found mutations and from a new, more ethnically diverse panel. Among 90 symptomatic mutation-positive individuals, 11 mutations accounted for 79% of carrier chromosomes. Of the two mutations that are novel, one alters the same residue (680) as a previously known mutation, and the other (P369S) is located in exon 3. Consistent with another recent report, the E148Q mutation was observed in patients of several ethnicities and on multiple microsatellite haplotypes, but haplotype data indicate an ancestral relationships between non-Jewish Italian and Ashkenazi Jewish patients with FMF and other affected populations. Among approximately 200 anonymous Ashkenazi Jewish DNA samples, the MEFV carrier frequency was 21%, with E148Q the most common mutation. Several lines of evidence indicate reduced penetrance among Ashkenazi Jews, especially for E148Q, P369S, and K695R. Nevertheless, E148Q helps account for recessive inheritance in an Ashkenazi family previously reported as an unusual case of dominantly inherited FMF. The presence of three frequent MEFV mutations in multiple Mediterranean populations strongly suggests a heterozygote advantage in this geographic region. 10090880 34 62 familial Mediterranean fever SpecificDisease D010505 10090880 200 228 Familial Mediterranean fever SpecificDisease D010505 10090880 230 233 FMF SpecificDisease D010505 10090880 240 258 recessive disorder DiseaseClass D030342 10090880 299 308 serositis DiseaseClass D012700 10090880 312 321 synovitis DiseaseClass D013585 10090880 327 330 FMF Modifier D010505 10090880 1087 1090 FMF SpecificDisease D010505 10090880 1522 1525 FMF SpecificDisease D010505 10090885|t|Autoimmune lymphoproliferative syndrome with defective Fas: genotype influences penetrance. 10090885|a|Autoimmune lymphoproliferative syndrome (ALPS) is a disorder of lymphocyte homeostasis and immunological tolerance. Most patients have a heterozygous mutation in the APT1 gene, which encodes Fas (CD95, APO-1), mediator of an apoptotic pathway crucial to lymphocyte homeostasis. Of 17 unique APT1 mutations in unrelated ALPS probands, 12 (71%) occurred in exons 7-9, which encode the intracellular portion of Fas. In vitro, activated lymphocytes from all 17 patients showed apoptotic defects when exposed to an anti-Fas agonist monoclonal antibody. Similar defects were found in a Fas-negative cell line transfected with cDNAs bearing each of the mutations. In cotransfection experiments, Fas constructs with either intra- or extracellular mutations caused dominant inhibition of apoptosis mediated by wild-type Fas. Two missense Fas variants, not restricted to patients with ALPS, were identified. Variant A (-1) T at the Fas signal-sequence cleavage site, which mediates apoptosis less well than wild-type Fas and is partially inhibitory, was present in 13% of African American alleles. Among the ALPS-associated Fas mutants, dominant inhibition of apoptosis was much more pronounced in mutants affecting the intracellular, versus extracellular, portion of the Fas receptor. Mutations causing disruption of the intracellular Fas death domain also showed a higher penetrance of ALPS phenotype features in mutation-bearing relatives. Significant ALPS-related morbidity occurred in 44% of relatives with intracellular mutations, versus 0% of relatives with extracellular mutations. Thus, the location of mutations within APT1 strongly influences the development and the severity of ALPS. 10090885 0 39 Autoimmune lymphoproliferative syndrome SpecificDisease D056735 10090885 93 132 Autoimmune lymphoproliferative syndrome SpecificDisease D056735 10090885 134 138 ALPS SpecificDisease D056735 10090885 145 207 disorder of lymphocyte homeostasis and immunological tolerance CompositeMention D008232|D007154 10090885 412 416 ALPS Modifier D056735 10090885 968 972 ALPS SpecificDisease D056735 10090885 1191 1195 ALPS Modifier D056735 10090885 1471 1475 ALPS Modifier D056735 10090885 1538 1542 ALPS Modifier D056735 10090885 1773 1777 ALPS SpecificDisease D056735 10190819|t|Mutational analysis and genotype-phenotype correlation of 29 unrelated Japanese patients with X-linked adrenoleukodystrophy. 10190819|a|BACKGROUND X-linked adrenoleukodystrophy (ALD) is an inherited disease characterized by progressive neurologic dysfunction, occasionally associated with adrenal insufficiency. The classic form of ALD usually has onset in childhood (childhood cerebral ALD), with rapid neurologic deterioration leading to a vegetative state. Adult-onset cerebral ALD also presents with rapidly progressive neurologic dysfunction. Milder phenotypes such as adrenomyeloneuropathy and Addison disease only also have been recognized. Despite discovery of the causative gene, a molecular basis for the diverse clinical presentations remains to be elucidated. OBJECTIVES To conduct mutational analyses in 29 Japanese patients with ALD from 29 unrelated families, to obtain knowledge of the spectrum of mutations in this gene, and to study genotype-phenotype correlations in Japanese patients. METHODS The 29 patients comprised 13 patients with childhood cerebral ALD, 11 patients with adult-onset cerebral ALD, and 5 patients with adrenomyeloneuropathy. We conducted detailed mutational analyses of 29 unrelated Japanese patients with ALD by genomic Southern blot analysis and direct nucleotide sequence analysis of reverse transcriptase-polymerase chain reaction products derived from total RNA that was extracted from cultured skin fibroblasts, lymphoblastoid cells, or peripheral blood leukocytes. RESULTS Three patients with adult-onset cerebral ALD were identified as having large genomic rearrangements. The remaining 26 patients were identified as having 21 independent mutations, including 12 novel mutations resulting in small nucleotide alterations in the ALD gene. Eighteen (69%) of 26 mutations were missense mutations. Most missense mutations involved amino acids conserved in homologous gene products, including PMP70, mALDRP, and Pxa1p. The AG dinucleotide deletion at position 1081-1082, which has been reported previously to be the most common mutation in white patients (12% -17%), was also identified as the most common mutation in Japanese patients (12%). All phenotypes were associated with mutations resulting in protein truncation or subtle amino acid changes. There were no differences in phenotypic expressions between missense mutations involving conserved amino acids and those involving nonconserved amino acids. CONCLUSIONS There are no obvious correlations between the phenotypes of patients with ALD and their genotypes, suggesting that other genetic or environmental factors modify the phenotypic expressions of ALD.. 10190819 94 123 X-linked adrenoleukodystrophy SpecificDisease D000326 10190819 137 166 X-linked adrenoleukodystrophy SpecificDisease D000326 10190819 168 171 ALD SpecificDisease D000326 10190819 179 196 inherited disease DiseaseClass D030342 10190819 226 248 neurologic dysfunction SpecificDisease D009461 10190819 279 300 adrenal insufficiency DiseaseClass D000309 10190819 322 325 ALD SpecificDisease D000326 10190819 358 380 childhood cerebral ALD SpecificDisease D000326 10190819 394 418 neurologic deterioration DiseaseClass D009461 10190819 462 474 cerebral ALD SpecificDisease D000326 10190819 514 536 neurologic dysfunction DiseaseClass D009461 10190819 564 585 adrenomyeloneuropathy SpecificDisease D000326 10190819 590 605 Addison disease SpecificDisease D000224 10190819 834 837 ALD SpecificDisease D000326 10190819 1048 1070 childhood cerebral ALD SpecificDisease D000326 10190819 1101 1113 cerebral ALD SpecificDisease D000326 10190819 1135 1156 adrenomyeloneuropathy SpecificDisease D000326 10190819 1239 1242 ALD SpecificDisease D000326 10190819 1546 1558 cerebral ALD SpecificDisease D000326 10190819 1771 1774 ALD Modifier D000326 10190819 2533 2536 ALD SpecificDisease D000326 10190819 2650 2653 ALD SpecificDisease D000326 6859721|t|Absence of the seventh component of complement in a patient with chronic meningococcemia presenting as vasculitis. 6859721|a|A previously healthy 40-year-old man presenting with fever, arthritis, and cutaneous vasculitis was found to have chronic meningococcemia. Evaluation of his complement system showed an absence of functional and antigenic C7, compatible with a complete deficiency of the seventh component of complement. Study of the patients family spanning four generations showed heterozygous deficiency of C7 in five members. Chronic neisserial infection can be associated with C7 deficiency and must be distinguished from other causes of cutaneous vasculitis.. 6859721 0 46 Absence of the seventh component of complement SpecificDisease OMIM:610102 6859721 65 88 chronic meningococcemia SpecificDisease D008589 6859721 103 113 vasculitis SpecificDisease D014657 6859721 168 173 fever SpecificDisease D005334 6859721 175 184 arthritis SpecificDisease D001168 6859721 190 210 cutaneous vasculitis SpecificDisease D018366 6859721 229 252 chronic meningococcemia SpecificDisease D008589 6859721 367 416 deficiency of the seventh component of complement SpecificDisease OMIM:610102 6859721 493 509 deficiency of C7 SpecificDisease OMIM:610102 6859721 527 555 Chronic neisserial infection SpecificDisease D016870 6859721 579 592 C7 deficiency SpecificDisease OMIM:610102 6859721 640 660 cutaneous vasculitis SpecificDisease D018366 10071185|t|Genotype and phenotype in patients with dihydropyrimidine dehydrogenase deficiency. 10071185|a|Dihydropyrimidine dehydrogenase (DPD) deficiency is an autosomal recessive disease characterised by thymine-uraciluria in homozygous deficient patients and has been associated with a variable clinical phenotype. In order to understand the genetic and phenotypic basis for DPD deficiency, we have reviewed 17 families presenting 22 patients with complete deficiency of DPD. In this group of patients, 7 different mutations have been identified, including 2 deletions [295-298delTCAT, 1897delC], 1 splice-site mutation [IVS14 + 1G > A)] and 4 missense mutations (85T > C, 703C > T, 2658G > A, 2983G > T). Analysis of the prevalence of the various mutations among DPD patients has shown that the G-- > A point mutation in the invariant splice donor site is by far the most common (52%), whereas the other six mutations are less frequently observed. A large phenotypic variability has been observed, with convulsive disorders, motor retardation and mental retardation being the most abundant manifestations. A clear correlation between the genotype and phenotype has not been established. An altered beta-alanine, uracil and thymine homeostasis might underlie the various clinical abnormalities encountered in patients with DPD deficiency. 10071185 40 82 dihydropyrimidine dehydrogenase deficiency SpecificDisease D054067 10071185 84 132 Dihydropyrimidine dehydrogenase (DPD) deficiency SpecificDisease D054067 10071185 139 166 autosomal recessive disease DiseaseClass D030342 10071185 356 370 DPD deficiency SpecificDisease D054067 10071185 438 455 deficiency of DPD SpecificDisease D054067 10071185 745 748 DPD Modifier D054067 10071185 985 1005 convulsive disorders DiseaseClass D004829 10071185 1007 1024 motor retardation DiseaseClass D019957 10071185 1029 1047 mental retardation DiseaseClass D008607 10071185 1252 1274 clinical abnormalities DiseaseClass D013568 10071185 1304 1318 DPD deficiency SpecificDisease D054067 1323345|t|Molecular characterization of glucose-6-phosphate dehydrogenase (G6PD) deficiency by natural and amplification created restriction sites: five mutations account for most G6PD deficiency cases in Taiwan. 1323345|a|We have developed a rapid and simple method to diagnose the molecular defects of glucose-6-phosphate dehydrogenase (G6PD) deficiency in Chinese in Taiwan. This method involves the selective amplification of a DNA fragment from human G6PD gene with specific oligonucleotide primers followed by digestion with restriction enzymes that recognize artificially created or naturally occurring restriction sites. Ninety-four Chinese males with G6PD deficiency were studied. The results show that 50% (47 of 94) were G to T mutation at nucleotide (nt) 1376, 21. 3% (20 of 94) were G to A mutation at nt 1388, 7. 4% (7 of 94) were A to G mutation at nt 493, 7. 4% (7 of 94) were A to G mutation at nt 95, 4. 2% (4 of 94) were C to T mutation at nt 1024, 1. 1% (1 of 94) was G to T mutation at nt 392, and 1. 1% (1 of 94) was G to A mutation at nt 487. These results show that the former five mutations account for more than 90% of G6PD deficiency cases in Taiwan. Aside from showing that G to T change at nt 1376 is the most common mutation, our research indicates that nt 493 mutation is a frequent mutation among Chinese in Taiwan. We compared G6PD activity among different mutations, without discovering significant differences between them. 1323345 30 81 glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 1323345 170 185 G6PD deficiency SpecificDisease D005955 1323345 284 335 glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 1323345 640 655 G6PD deficiency SpecificDisease D005955 1323345 1125 1140 G6PD deficiency SpecificDisease D005955 2828430|t|Homozygous hypobetalipoproteinemia: a disease distinct from abetalipoproproteinemia at the molecular level. 2828430|a|apoB DNA, RNA, and protein from two patients with homozygous hypobetalipoproteinemia (HBL) were evaluated and compared with normal individuals. Southern blot analysis with 10 different cDNA probes revealed a normal gene without major insertions, deletions, or rearrangements. Northern and slot blot analyses of total liver mRNA from HBL patients documented a normal size apoB mRNA that was present in greatly reduced quantities. ApoB protein was detected within HBL hepatocytes utilizing immunohistochemical techniques; however, it was markedly reduced in quantity when compared with control samples. No apoB was detectable in the plasma of HBL individuals with an ELISA assay. These data are most consistent with a mutation in the coding portion of the apoB gene in HBL patients, leading to an abnormal apoB protein and apoB mRNA instability. These results are distinct from those previously noted in abetalipoproteinemia, which was characterized by an elevated level of hepatic apoB mRNA and accumulation of intracellular hepatic apoB protein.. 2828430 0 34 Homozygous hypobetalipoproteinemia SpecificDisease D006995 2828430 60 83 abetalipoproproteinemia SpecificDisease D000012 2828430 158 192 homozygous hypobetalipoproteinemia SpecificDisease D006995 2828430 194 197 HBL SpecificDisease D006995 2828430 441 444 HBL Modifier D006995 2828430 570 573 HBL Modifier D006995 2828430 749 752 HBL Modifier D006995 2828430 875 878 HBL Modifier D006995 2828430 1010 1030 abetalipoproteinemia SpecificDisease D000012 10471457|t|A population-based study of the clinical expression of the hemochromatosis gene. 10471457|a|BACKGROUND AND METHODS Hereditary hemochromatosis is associated with homozygosity for the C282Y mutation in the hemochromatosis (HFE) gene on chromosome 6, elevated serum transferrin saturation, and excess iron deposits throughout the body. To assess the prevalence and clinical expression of the HFE gene, we conducted a population-based study in Busselton, Australia. In 1994, we obtained blood samples for the determination of serum transferrin saturation and ferritin levels and the presence or absence of the C282Y mutation and the H63D mutation (which may contribute to increased hepatic iron levels) in 3011 unrelated white adults. We evaluated all subjects who had persistently elevated transferrin-saturation values (45 percent or higher) or were homozygous for the C282Y mutation. We recommended liver biopsy for subjects with serum ferritin levels of 300 ng per milliliter or higher. The subjects were followed for up to four years. RESULTS Sixteen of the subjects (0. 5 percent) were homozygous for the C282Y mutation, and 424 (14. 1 percent) were heterozygous. The serum transferrin saturation was 45 percent or higher in 15 of the 16 who were homozygous; in 1 subject it was 43 percent. Four of the homozygous subjects had previously been given a diagnosis of hemochromatosis, and 12 had not. Seven of these 12 patients had elevated serum ferritin levels in 1994; 6 of the 7 had further increases in 1998, and 1 had a decrease, although the value remained elevated. The serum ferritin levels in the four other homozygous patients remained in the normal range. Eleven of the 16 homozygous subjects underwent liver biopsy; 3 had hepatic fibrosis, and 1, who had a history of excessive alcohol consumption, had cirrhosis and mild microvesicular steatosis. Eight of the 16 homozygous subjects had clinical findings that were consistent with the presence of hereditary hemochromatosis, such as hepatomegaly, skin pigmentation, and arthritis. CONCLUSIONS In a population of white adults of northern European ancestry, 0. 5 percent were homozygous for the C282Y mutation in the HFE gene. However, only half of those who were homozygous had clinical features of hemochromatosis, and one quarter had serum ferritin levels that remained normal over a four-year period. 10471457 59 74 hemochromatosis Modifier D006432 10471457 105 131 Hereditary hemochromatosis SpecificDisease D006432 10471457 194 209 hemochromatosis Modifier D006432 10471457 281 301 excess iron deposits DiseaseClass D019190 10471457 1357 1372 hemochromatosis SpecificDisease D006432 10471457 1724 1740 hepatic fibrosis DiseaseClass D008103 10471457 1770 1799 excessive alcohol consumption SpecificDisease D000435 10471457 1805 1814 cirrhosis DiseaseClass D008103 10471457 1824 1848 microvesicular steatosis SpecificDisease D005234 10471457 1950 1976 hereditary hemochromatosis SpecificDisease D006432 10471457 1986 1998 hepatomegaly DiseaseClass D006529 10471457 2000 2017 skin pigmentation DiseaseClass D010859 10471457 2023 2032 arthritis DiseaseClass D001168 10471457 2252 2267 hemochromatosis SpecificDisease D006432 7574457|t|Overexpression of DM20 messenger RNA in two brothers with Pelizaeus-Merzbacher disease. 7574457|a|Pelizaeus-Merzbacher disease is a rare, sex-linked recessive, dysmyelinating disease of the central nervous system that has been associated with mutations in the myelin proteolipid protein (PLP) gene. Only 25% of patients studied with Pelizaeus-Merzbacher disease have exonic mutations in this gene, the underlying cause of the disease in the remaining patients is unknown. The PLP gene encodes two major alternatively spliced transcripts called PLP and DM20. PLP messenger RNA is specifically expressed in central nervous system tissue, whereas DM20 messenger RNA is found in central nervous system, cardiac, and other tissues. We studied cultured skin fibroblasts from 2 brothers with Pelizaeus-Merzbacher disease who exhibited no detectable exonic mutation of the PLP gene. Examination of RNA from these cells showed that the level of DM20 messenger RNA is elevated sixfold relative to male control skin fibroblasts. An unrelated female carrier, also with no detectable exonic mutation, showed a threefold increase in DM20 messenger RNA in cultured skin fibroblasts. Our findings suggest that in some patients, Pelizaeus-Merzbacher disease is caused by overexpression of PLP gene transcripts, and that in these families a 50% increase of DM20 messenger RNA in females, relative to the increase in affected males, can identify a female carrier.. 7574457 58 86 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 7574457 88 116 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 7574457 122 202 rare, sex-linked recessive, dysmyelinating disease of the central nervous system DiseaseClass D020279+D035583 7574457 323 351 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 7574457 775 803 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 7574457 1202 1230 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 10466420|t|Homozygosity for a novel DTDST mutation in a child with a 'broad bone-platyspondylic' variant of diastrophic dysplasia. 10466420|a|Atypical or variant forms of well-known chondrodysplasias may pose diagnostic problems. We report on a girl with clinical features suggesting diastrophic dysplasia but with unusual radiographic features including severe platyspondyly, wide metaphyses, and fibular overgrowth, which are partially reminiscent of metatropic dysplasia. The diagnosis was clarified by molecular analysis of the DTDST gene, which revealed homozygosity for a previously undescribed mutation leading to a Q454P substitution in the 10th transmembrane domain of the DTDST sulfate transporter. Molecular analysis may be of particular value in such atypical cases.. 10466420 97 118 diastrophic dysplasia SpecificDisease C536170 10466420 160 177 chondrodysplasias DiseaseClass D010009 10466420 262 283 diastrophic dysplasia SpecificDisease C536170 10466420 340 353 platyspondyly DiseaseClass D013122 10466420 431 451 metatropic dysplasia SpecificDisease C537356 1676565|t|Carrier detection and prenatal diagnosis of Pelizaeus-Merzbacher disease using a combination of anonymous DNA polymorphisms and the proteolipid protein (PLP) gene cDNA. 1676565|a|We report carrier identification and a prenatal diagnosis using DNA polymorphisms in 2 families with X-linked Pelizaeus-Merzbacher disease (PMD). In both families, the proteolipid protein (PLP) gene in the single affected male could be traced back to his unaffected maternal grandfather. Therefore, each family contains a new mutation. In the case of the prenatal diagnosis, the fetus was shown by cytogenetic analysis to be a female, who we predict will be a noncarrier of PMD based on her genotype with the PLP intragenic polymorphism.. 1676565 44 72 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 1676565 270 307 X-linked Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 1676565 309 312 PMD SpecificDisease OMIM:312080 1676565 643 646 PMD SpecificDisease OMIM:312080 1776638|t|Gardner syndrome in a boy with interstitial deletion of the long arm of chromosome 5. 1776638|a|We described a 15-year-old boy with Gardner syndrome (GS), mental retardation, and craniofacial abnormalities. High-resolution banding analysis showed an interstitial deletion of the long arm of chromosome 5 (q22. 1----q31 1----q31. 1). The breakpoints in the present case and in 3 previously reported 5q- patients with adenomatous polyposis coli suggest that the gene responsible for GS/or familial polyposis coli (FPC) is in the 5q22 region, a result consistent with the findings of linkage studies 1776638 0 16 Gardner syndrome SpecificDisease D005736 1776638 122 138 Gardner syndrome SpecificDisease D005736 1776638 140 142 GS SpecificDisease D005736 1776638 145 163 mental retardation DiseaseClass D008607 1776638 169 195 craniofacial abnormalities DiseaseClass D019465 1776638 406 432 adenomatous polyposis coli SpecificDisease D011125 1776638 471 473 GS SpecificDisease D005736 1776638 477 500 familial polyposis coli SpecificDisease D011125 1776638 502 505 FPC SpecificDisease D011125 1577763|t|Type I human complement C2 deficiency. A 28-base pair gene deletion causes skipping of exon 6 during RNA splicing. 1577763|a|Two variants of a genetic deficiency of complement protein C2 (C2D) have been previously identified. No C2 protein translation is detected in type I deficiency, while type II deficiency is characterized by a selective block in C2 secretion. Type I C2 deficiency was described in a family in which the C2 null allele (C2Q0) is associated with the major histocompatibility haplotype/complotype HLA-A25, B18, C2Q0, BfS, C4A4, C4B2, Drw2; this extended haplotype occurs in over 90% of C2-deficient individuals (common complotype/haplotype). To determine the molecular basis of type I C2 deficiency, the C2 gene and cDNA were characterized from a homozygous type I C2-deficient individual with the common associated haplotype/complotype. We found a 28-base pair deletion in the type I C2Q0 gene, beginning 9 base pairs upstream of the 3-end of exon 6, that generates a C2 transcript with a complete deletion of exon 6 (134 base pair) and a premature termination codon. In studies of eight kindred, the 28-base pair deletion was observed in all C2Q0 alleles associated with the common type I deficient complotype/haplotype; this deletion was not present in normal C2 nor in type II C2-deficient genes. These data demonstrate that 1) type I human complement C2 deficiency is caused by a 28-base pair genomic deletion that causes skipping of exon 6 during RNA splicing, resulting in generation of a premature termination codon, 2) the 28-base pair deletion in the type I C2Q0 gene is strongly associated with the HLA haplotype/complotype A25, B18, C2Q0, BfS, C4A4, C4B2, Drw2, suggesting that all C2-deficient individuals with this haplotype/complotype will harbor the 28-base pair C2 gene deletion, and 3) type II C2 deficiency is caused by a different, as yet uncharacterized, molecular genetic defect.. 1577763 0 37 Type I human complement C2 deficiency SpecificDisease OMIM:217000 1577763 141 176 deficiency of complement protein C2 SpecificDisease OMIM:217000 1577763 356 376 Type I C2 deficiency SpecificDisease OMIM:217000 1577763 596 608 C2-deficient Modifier OMIM:217000 1577763 688 708 type I C2 deficiency SpecificDisease OMIM:217000 1577763 768 787 type I C2-deficient Modifier OMIM:217000 1577763 1283 1303 type II C2-deficient Modifier OMIM:217000 1577763 1343 1380 type I human complement C2 deficiency SpecificDisease OMIM:217000 1577763 1705 1717 C2-deficient Modifier OMIM:217000 1577763 1815 1836 type II C2 deficiency SpecificDisease OMIM:217000 1577763 1897 1911 genetic defect DiseaseClass D030342 10767339|t|(Over)correction of FMR1 deficiency with YAC transgenics: behavioral and physical features. 10767339|a|Fragile X syndrome is a common cause of mental retardation involving loss of expression of the FMR1 gene. The role of FMR1 remains undetermined but the protein appears to be involved in RNA metabolism. Fmr1 knockout mice exhibit a phenotype with some similarities to humans, such as macroorchidism and behavioral abnormalities. As a step toward understanding the function of FMR1 and the determination of the potential for therapeutic approaches to fragile X syndrome, yeast artificial chromosome (YAC) transgenic mice were generated in order to determine whether the Fmr1 knockout mouse phenotype could be rescued. Several transgenic lines were generated that carried the entire FMR1 locus with extensive amounts of flanking sequence. We observed that the YAC transgene supported production of the human protein (FMRP) which was present at levels 10 to 15 times that of endogenous protein and was expressed in a cell- and tissue-specific manner. Macro-orchidism was absent in knockout mice carrying the YAC transgene indicating functional rescue by the human protein. Given the complex behavioral phenotype in fragile X patients and the mild phenotype previously reported for the Fmr1 knockout mouse, we performed a more thorough evaluation of the Fmr1 knockout phenotype using additional behavioral assays that had not previously been reported for this animal model. The mouse displayed reduced anxiety-related responses with increased exploratory behavior. FMR1 YAC transgenic mice overexpressing the human protein did produce opposing behavioral responses and additional abnormal behaviors were also observed. These findings have significant implications for gene therapy for fragile X syndrome since overexpression of the gene may harbor its own phenotype.. 10767339 20 35 FMR1 deficiency SpecificDisease OMIM:300624 10767339 92 110 Fragile X syndrome SpecificDisease D005600 10767339 132 150 mental retardation DiseaseClass D008607 10767339 375 389 macroorchidism SpecificDisease D005600 10767339 541 559 fragile X syndrome SpecificDisease D005600 10767339 1203 1212 fragile X Modifier D005600 10767339 1489 1496 anxiety Modifier D001008 10767339 1772 1790 fragile X syndrome SpecificDisease D005600 10208848|t|Fabry disease: identification of novel alpha-galactosidase A mutations and molecular carrier detection by use of fluorescent chemical cleavage of mismatches. 10208848|a|Fabry disease (FD) (angiokeratoma corporis diffusum) is an X-linked inborn error of glycosphingolipid metabolism caused by defects in the lysosomal alpha-galactosidase A gene (GLA). The enzymatic defect leads to the systemic accumulation of neutral glycosphingolipids with terminal alpha-galactosyl moieties. Clinically, affected hemizygous males have angiokeratoma, severe acroparesthesia, renal failure, and vasculopathy of the heart and brain. While demonstration of alpha-galactosidase deficiency in leukocytes is diagnostic in affected males, enzymatic detection of female carriers is often inconclusive, due to random X-chromosomal inactivation, underlining the need of molecular investigations for accurate genetic counseling. By use of chemical cleavage of mismatches adapted to fluorescence-based detection systems, we have characterized the mutations underlying alpha-Gal A deficiency in 16 individuals from six unrelated families with FD. The mutational spectrum included five missense mutations (C202W, C223G, N224D, R301Q, and Q327K) and one splice-site mutation [IVS3 G (-1) -- > C]. Studies at the mRNA level showed that the latter led to altered pre-mRNA splicing with consequent alteration of the mRNA translational reading frame and generation of a premature termination codon of translation. By use of this strategy, carrier status was accurately assessed in all seven at-risk females tested, whereas enzymatic dosages failed to diagnose or exclude heterozygosity.. 10208848 0 13 Fabry disease SpecificDisease D000795 10208848 158 171 Fabry disease SpecificDisease D000795 10208848 173 175 FD SpecificDisease D000795 10208848 178 209 angiokeratoma corporis diffusum SpecificDisease D000795 10208848 217 270 X-linked inborn error of glycosphingolipid metabolism DiseaseClass D008052 10208848 510 523 angiokeratoma SpecificDisease D000794 10208848 525 547 severe acroparesthesia SpecificDisease D010292 10208848 549 562 renal failure SpecificDisease D051437 10208848 568 603 vasculopathy of the heart and brain CompositeMention D014652 10208848 628 658 alpha-galactosidase deficiency SpecificDisease D000795 10208848 1030 1052 alpha-Gal A deficiency SpecificDisease D000795 10208848 1104 1106 FD SpecificDisease D000795 8533768|t|Evidence for linkage of bipolar disorder to chromosome 18 with a parent-of-origin effect. 8533768|a|A susceptibility gene on chromosome 18 and a parent-of-origin effect have been suggested for bipolar affective disorder (BPAD). We have studied 28 nuclear families selected for apparent unilineal transmission of the BPAD phenotype, by using 31 polymorphic markers spanning chromosome 18. Evidence for linkage was tested with affected-sib-pair and LOD score methods under two definitions of the affected phenotype. The affected-sibpair analyses indicated excess allele sharing for markers on 18p within the region reported previously. The greatest sharing was at D18S37 64% in bipolar and recurrent unipolar (RUP) sib pairs (P =. 0006). In addition, excess sharing of the paternally, but not maternally, transmitted alleles was observed at three markers on 18q at D18S41, 51 bipolar and RUP sib pairs were concordant for paternally transmitted alleles, and 21 pairs were discordant (P = 0004). The evidence for linkage to loci on both 18p and 18q was strongest in the 11 paternal pedigrees, i. e e., those in which the father or one of the fathers sibs is affected. In these pedigrees, the greatest allele sharing (81%; P =. 00002) and the highest LOD score (3. 51; phi = 0. 0) were observed at D18S41. Our results provide further support for linkage of BPAD to chromosome 18 and the first molecular evidence for a parent-of-origin effect operating in this disorder. The number of loci involved, and their precise location, require further study 8533768 24 40 bipolar disorder SpecificDisease D001714 8533768 183 209 bipolar affective disorder SpecificDisease D001714 8533768 211 215 BPAD SpecificDisease D001714 8533768 306 310 BPAD Modifier D001714 8533768 1345 1349 BPAD SpecificDisease D001714 8531967|t|BRCA1 mutations in a population-based sample of young women with breast cancer. 8531967|a|BACKGROUND. Inherited mutations in the BRCA1 gene are associated with a high risk of breast and ovarian cancer in some families. However, little is known about the contribution of BRCA1 mutations to breast cancer in the general population. We analyzed DNA samples from women enrolled in a population-based study of early-onset breast cancer to assess the spectrum and frequency of germ-line BRCA1 mutations in young women with breast cancer. METHODS. We studied 80 women in whom breast cancer was diagnosed before the age of 35, and who were not selected on the basis of family history. Genomic DNA was studied for BRCA1 mutations by analysis involving single-strand conformation polymorphisms and with allele-specific assays. Alterations were defined by DNA sequencing. RESULTS. Germ-line BRCA1 mutations were identified in 6 of the 80 women. Four additional rare sequence variants of unknown functional importance were also identified. Two of the mutations and three of the rare sequence variants were found among the 39 women who reported no family history of breast or ovarian cancer. None of the mutations and only one of the rare variants was identified in a reference population of 73 unrelated subjects. CONCLUSIONS. Alterations in BRCA1 were identified in approximately 10 percent of this cohort of young women with breast cancer. The risk of harboring a mutation was not limited to women with family histories of breast or ovarian cancer. These results represent a minimal estimate of the frequency of BRCA1 mutations in this population. Comprehensive methods of identifying BRCA1 mutations and understanding their importance will be needed before testing of women in the general population can be undertaken.. 8531967 65 78 breast cancer SpecificDisease D001943 8531967 165 190 breast and ovarian cancer CompositeMention D001943|D010051 8531967 279 292 breast cancer SpecificDisease D001943 8531967 407 420 breast cancer SpecificDisease D001943 8531967 507 520 breast cancer SpecificDisease D001943 8531967 559 572 breast cancer SpecificDisease D001943 8531967 1143 1167 breast or ovarian cancer CompositeMention D001943|D010051 8531967 1405 1418 breast cancer SpecificDisease D001943 8531967 1503 1527 breast or ovarian cancer CompositeMention D001943|D010051 10915776|t|Retinoschisin, the X-linked retinoschisis protein, is a secreted photoreceptor protein, and is expressed and released by Weri-Rb1 cells. 10915776|a|X-linked retinoschisis is characterized by microcystic-like changes of the macular region and schisis within the inner retinal layers, leading to visual deterioration in males. Many missense and protein-truncating mutations of the causative gene RS1 have now been identified and are thought to be inactivating. RS1 encodes a 224 amino acid protein, retinoschisin, which contains a discoidin domain but is of unknown function. We have generated a polyclonal antibody against a peptide from a unique region within retinoschisin, which detects a protein of approximately 28 kDa in retinal samples reduced with dithiothreitol, but multimers sized > 40 kDa under non-reducing conditions. A screen of human tissues with this antibody reveals retinoschisin to be retina specific and the antibody detects a protein of similar size in bovine and murine retinae. We investigated the expression pattern in the retina of both RS1 mRNA (using in situ hybridization with riboprobes) and retinoschisin (using immunohistochemistry). The antisense riboprobe detected RS1 mRNA only in the photoreceptor layer but the protein product of the gene was present both in the photoreceptors and within the inner portions of the retina. Furthermore, differentiated retinoblastoma cells (Weri-Rb1 cells) were found to express RS1 mRNA and to release retinoschisin. These results suggest that retinoschisin is released by photo-receptors and has functions within the inner retinal layers. Thus, X-linked retinoschisis is caused by abnormalities in a putative secreted photoreceptor protein and is the first example of a secreted photo-receptor protein associated with a retinal dystrophy.. 10915776 19 41 X-linked retinoschisis Modifier D041441 10915776 137 159 X-linked retinoschisis SpecificDisease D041441 10915776 283 303 visual deterioration DiseaseClass C531604 10915776 1376 1390 retinoblastoma Modifier D012175 10915776 1604 1626 X-linked retinoschisis SpecificDisease D041441 10915776 1779 1796 retinal dystrophy DiseaseClass D058499 1302003|t|Aberrant splicing of the CHM gene is a significant cause of choroideremia. 1302003|a|Choroideremia (CHM) is an X-linked progressive degeneration of the choroid and retina. 12% of unrelated male patients carry deletions of the partially cloned CHM gene. In Finland, there are more than 120 living CHM patients belonging to eight apparently unrelated pedigrees. Molecular deletions involving the CHM gene have been detected in three families. We have screened the remaining five families for point mutations. In one large family a single nucleotide (T) insertion into the donor splice site of exon C leads to two aberrantly spliced mRNAs both producing a premature stop codon. The mutation can be assayed easily by amplification and digestion with Msel. Our findings provide additional evidence for the pathogenetic role of CHM mutations and provide a diagnostic tool for one fifth of the worlds known CHM patients.. 1302003 25 28 CHM Modifier D015794 1302003 60 73 choroideremia SpecificDisease D015794 1302003 75 88 Choroideremia SpecificDisease D015794 1302003 90 93 CHM SpecificDisease D015794 1302003 101 160 X-linked progressive degeneration of the choroid and retina DiseaseClass D015785 1302003 233 236 CHM Modifier D015794 1302003 286 289 CHM Modifier D015794 1302003 384 387 CHM Modifier D015794 1302003 812 815 CHM Modifier D015794 1302003 890 893 CHM Modifier D015794 7811247|t|X-linked adrenoleukodystrophy (ALD): a novel mutation of the ALD gene in 6 members of a family presenting with 5 different phenotypes. 7811247|a|Fragments of the adrenoleukodystrophy (ALD) cDNA from a patient with adolescent ALD were amplified by polymerase chain reaction and subcloned. Bidirectional sequencing of the entire coding ALD gene disclosed a cytosine to guanine transversion at nucleotide 1451 in exon five, resulting in substitution of proline 484 by arginine. Five of nine siblings of the patient, comprising two cerebral ALD, one adrenomyeloneuropathy, one Addison only as well as the symptomatic mother (all accumulating very long chain fatty acids) carried this mutation, which was not found in the unaffected persons, in five unrelated ALD patients, and in twenty controls. We propose that this missense mutation generated the disease per se as well as the metabolic defect; the different phenotypes, however, must have originated by means of additional pathogenetic factors.. 7811247 0 29 X-linked adrenoleukodystrophy SpecificDisease D000326 7811247 31 34 ALD SpecificDisease D000326 7811247 61 64 ALD Modifier D000326 7811247 152 172 adrenoleukodystrophy Modifier D000326 7811247 174 177 ALD Modifier D000326 7811247 204 218 adolescent ALD SpecificDisease D000326 7811247 324 327 ALD Modifier D000326 7811247 518 530 cerebral ALD SpecificDisease D000326 7811247 536 557 adrenomyeloneuropathy SpecificDisease D000326 7811247 563 575 Addison only SpecificDisease D000224 7811247 745 748 ALD Modifier D000326 2352258|t|Statistical analysis of the two stage mutation model in von Hippel-Lindau disease, and in sporadic cerebellar haemangioblastoma and renal cell carcinoma. 2352258|a|Analysis of the age incidence curves for unilateral and bilateral retinoblastoma led Knudson to propose that hereditary tumours may arise by a single event and sporadic tumours by a two stage mutation process. It has been suggested recently that sporadic renal cell carcinoma may arise from a two stage mutation process. We analysed the age incidence curves for symptomatic renal cell carcinoma (n = 26) and cerebellar haemangioblastoma (n = 68) in 109 patients with von Hippel-Lindau (VHL) disease, and compared them to 104 patients with sporadic renal cell carcinoma and 43 patients with sporadic cerebellar haemangioblastoma. The age incidence curves for renal cell carcinoma and cerebellar haemangioblastoma in VHL disease were compatible with a single mutation model, whereas the age incidence curves for sporadic renal cell carcinoma and cerebellar haemangioblastoma suggested a two stage mutation process. These data are compatible with the VHL gene functioning as a recessive tumour suppressor gene. Sporadic cerebellar haemangioblastoma and some renal cell carcinoma may arise from somatic mutations inactivating both alleles at the VHL locus.. 2352258 56 81 von Hippel-Lindau disease SpecificDisease D006623 2352258 90 127 sporadic cerebellar haemangioblastoma SpecificDisease D018325 2352258 132 152 renal cell carcinoma SpecificDisease D002292 2352258 195 234 unilateral and bilateral retinoblastoma CompositeMention D012175 2352258 263 281 hereditary tumours DiseaseClass D009386 2352258 314 330 sporadic tumours DiseaseClass D009369 2352258 400 429 sporadic renal cell carcinoma SpecificDisease D002292 2352258 528 548 renal cell carcinoma SpecificDisease D002292 2352258 562 590 cerebellar haemangioblastoma SpecificDisease D018325 2352258 621 652 von Hippel-Lindau (VHL) disease SpecificDisease D006623 2352258 693 722 sporadic renal cell carcinoma SpecificDisease D002292 2352258 744 781 sporadic cerebellar haemangioblastoma SpecificDisease D018325 2352258 812 832 renal cell carcinoma SpecificDisease D002292 2352258 837 865 cerebellar haemangioblastoma SpecificDisease D018325 2352258 869 880 VHL disease SpecificDisease D006623 2352258 964 993 sporadic renal cell carcinoma SpecificDisease D002292 2352258 998 1026 cerebellar haemangioblastoma SpecificDisease D018325 2352258 1102 1105 VHL Modifier D006623 2352258 1138 1144 tumour Modifier D009369 2352258 1162 1199 Sporadic cerebellar haemangioblastoma SpecificDisease D018325 2352258 1209 1229 renal cell carcinoma SpecificDisease D002292 2352258 1296 1299 VHL Modifier D006623 10364518|t|Characterization of a germline mosaicism in families with Lowe syndrome, and identification of seven novel mutations in the OCRL1 gene. 10364518|a|The oculocerebrorenal syndrome of Lowe (OCRL) is an X-linked disorder characterized by major abnormalities of eyes, nervous system, and kidneys. Mutations in the OCRL1 gene have been associated with the disease. OCRL1 encodes a phosphatidylinositol 4, 5-biphosphate (PtdIns [4, 5] P2) 5-phosphatase. We have examined the OCRL1 gene in eight unrelated patients with OCRL and have found seven new mutations and one recurrent in-frame deletion. Among the new mutations, two nonsense mutations (R317X and E558X) and three other frameshift mutations caused premature termination of the protein. A missense mutation, R483G, was located in the highly conserved PtdIns (4, 5) P2 5-phosphatase domain. Finally, one frameshift mutation, 2799delC, modifies the C-terminal part of OCRL1, with an extension of six amino acids. Altogether, 70% of missense mutations are located in exon 15, and 52% of all mutations cluster in exons 11-15. We also identified two new microsatellite markers for the OCRL1 locus, and we detected a germline mosaicism in one family. This observation has direct implications for genetic counseling of Lowe syndrome families.. 10364518 58 71 Lowe syndrome SpecificDisease D009800 10364518 140 174 oculocerebrorenal syndrome of Lowe SpecificDisease D009800 10364518 176 180 OCRL SpecificDisease D009800 10364518 188 205 X-linked disorder DiseaseClass D040181 10364518 229 279 abnormalities of eyes, nervous system, and kidneys CompositeMention D000015 10364518 501 505 OCRL SpecificDisease D009800 10364518 1251 1264 Lowe syndrome Modifier D009800 10449429|t|Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice. 10449429|a|Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene, leading to the absence of the dystrophin protein in striated muscle. A significant number of these mutations are premature stop codons. On the basis of the observation that aminoglycoside treatment can suppress stop codons in cultured cells, we tested the effect of gentamicin on cultured muscle cells from the mdx mouse - an animal model for DMD that possesses a premature stop codon in the dystrophin gene. Exposure of mdx myotubes to gentamicin led to the expression and localization of dystrophin to the cell membrane. We then evaluated the effects of differing dosages of gentamicin on expression and functional protection of the muscles of mdx mice. We identified a treatment regimen that resulted in the presence of dystrophin in the cell membrane in all striated muscles examined and that provided functional protection against muscular injury. To our knowledge, our results are the first to demonstrate that aminoglycosides can suppress stop codons not only in vitro but also in vivo. Furthermore, these results raise the possibility of a novel treatment regimen for muscular dystrophy and other diseases caused by premature stop codon mutations. This treatment could prove effective in up to 15% of patients with DMD.. 10449429 88 115 Duchenne muscular dystrophy SpecificDisease D020388 10449429 117 120 DMD SpecificDisease D020388 10449429 512 515 DMD SpecificDisease D020388 10449429 1005 1020 muscular injury DiseaseClass D014947 10449429 1245 1263 muscular dystrophy SpecificDisease D009136 10449429 1392 1395 DMD SpecificDisease D020388 1939657|t|Molecular and metabolic basis for the metabolic disorder normotriglyceridemic abetalipoproteinemia. 1939657|a|We have previously described a disorder, normotriglyceridemic abetalipoproteinemia, that is characterized by the virtual absence of plasma low density lipoproteins and complete absence of apoB-100, but with apparently normal secretion of triglyceride-rich lipoproteins containing apoB-48. The patients plasma lipoproteins were shown on polyacrylamide gels and by antibody mapping to have a new truncated apoB variant, apoB-50, circulating along with her apoB-48. We have found this individual to be homozygous for a single C-to-T nucleotide substitution at apoB codon 2252, which produces a premature in-frame stop codon. Thus, this is a rare example of homozygous hypobetalipoproteinemia. Electron photomicrographs revealed that the diameters of particles in the d less than 1. 006 g/ml lipoprotein fraction, in both the postprandial and postabsorptive state, are bimodally distributed. The molar ratio of apoE to apoB in these particles is 3. 5 1, similar to normal VLDL. The plasma LDL interval contains both spherical and cuboidal particles. Autologous reinfusion of labeled d less than 1. 006 g/ml lipoproteins showed exponential disappearance from plasma, with an apparent half-removal time of 50 min, somewhat slower than for normal chylomicrons but within the normal range for VLDL. The calculated production rate for apoB was within the normal range in this subject. 1939657 38 56 metabolic disorder DiseaseClass D008659 1939657 57 98 normotriglyceridemic abetalipoproteinemia SpecificDisease D000012 1939657 141 182 normotriglyceridemic abetalipoproteinemia SpecificDisease D000012 1939657 754 788 homozygous hypobetalipoproteinemia SpecificDisease D006995 1409710|t|von Willebrand disease type B: a missense mutation selectively abolishes ristocetin-induced von Willebrand factor binding to platelet glycoprotein Ib. 1409710|a|von Willebrand factor (vWF) is a multimeric glycoprotein that mediates the adhesion of platelets to the subendothelium by binding to platelet glycoprotein Ib. For human vWF, this interaction can be induced in vitro by the antibiotic ristocetin or the snake venom protein botrocetin. A missense mutation, Gly-561-- > Ser, was identified within the proposed glycoprotein Ib binding domain of vWF in the proband with von Willebrand disease type B, a unique variant characterized by no ristocetin-induced, but normal botrocetin-induced, binding to glycoprotein Ib. The corresponding mutant recombinant protein, rvWF (G561S), formed normal multimers and exhibited the same functional defect as the patients plasma vWF, confirming that this mutation causes von Willebrand disease type B. These data show that botrocetin and ristocetin cofactor activities of vWF can be dissociated by a point mutation and confirm that these mediators promote vWF binding to platelets by different mechanisms. The normal botrocetin-induced binding and the defective ristocetin-induced binding of rvWF (G561S) suggest that the primary defect in von Willebrand disease type B may be a failure of normal allosteric regulation of the glycoprotein Ib binding function of vWF.. 1409710 0 29 von Willebrand disease type B SpecificDisease D014842 1409710 92 106 von Willebrand Modifier D014842 1409710 151 165 von Willebrand Modifier D014842 1409710 565 594 von Willebrand disease type B SpecificDisease D014842 1409710 902 931 von Willebrand disease type B SpecificDisease D014842 1409710 1271 1300 von Willebrand disease type B SpecificDisease D014842 8554067|t|A high incidence of BRCA1 mutations in 20 breast-ovarian cancer families. 8554067|a|We have analyzed 20 breast-ovarian cancer families, the majority of which show positive evidence of linkage to chromosome 17q12 for germ-line mutations in the BRCA1 gene. BRCA1 mutations cosegregating with breast and ovarian cancer susceptibility were identified in 16 families, including 1 family with a case of male breast cancer. Nine of these mutations have not been reported previously. The majority of mutations were found to generate a premature stop codon leading to the formation of a truncated BRCA1 protein of 2% -88% of the expected normal length. Two mutations altered the RING finger domain. Sequencing of genomic DNA led to the identification of a mutation in the coding region of BRCA1 in 12 families, and cDNA analysis revealed an abnormal or missing BRCA1 transcript in 4 of the 8 remaining families. A total of eight mutations were associated with a reduced quantity of BRCA1 transcript. We were unable to detect BRCA1 mutations in 4 of the 20 families, but only 1 of these was clearly linked to BRCA1. It is expected that the majority of clear examples of the breast-ovarian syndrome will be associated with germ-line mutations in the coding region of BRCA1.. 8554067 42 63 breast-ovarian cancer Modifier D061325 8554067 94 115 breast-ovarian cancer Modifier D061325 8554067 280 305 breast and ovarian cancer Modifier D061325 8554067 387 405 male breast cancer SpecificDisease D018567 8554067 1154 1177 breast-ovarian syndrome SpecificDisease D061325 8244393|t|Assignment of the human Na+/glucose cotransporter gene SGLT1 to chromosome 22q13.1. 8244393|a|The Na +/glucose cotransporter gene SGLT1 encodes the primary carrier protein responsible for the uptake of the dietary sugars glucose and galactose from the intestinal lumen. SGLT1 transport activity is currently exploited in oral rehydration therapy. The 75-kDa glycoprotein is localized in the brush border of the intestinal epithelium and is predicted to comprise 12 membrane spans. In two patients with the autosomal recessive disease glucose/galactose malabsorption, the underlying cause was found to be a missense mutation in SGLT1, and the Asp28-- > Asn change was demonstrated in vitro to eliminate SGLT1 transport activity. The SGLT1 gene was previously shown to reside on the distal q arm of chromosome 22 (11. 2-- > qter). We have used a cosmid probe for fluorescence in situ hybridization, which refines the localization to 22q13. 1, and provide an example of the utility of the SGLT1 probe as a diagnostic for genetic diseases associated with translocations of chromosome 22. 8244393 496 555 autosomal recessive disease glucose/galactose malabsorption SpecificDisease OMIM:606824 8244393 1008 1024 genetic diseases DiseaseClass D030342 1384323|t|A pseudodeficiency allele common in non-Jewish Tay-Sachs carriers: implications for carrier screening. 1384323|a|Deficiency of beta-hexosaminidase A (Hex A) activity typically results in Tay-Sachs disease. However, healthy subjects found to be deficient in Hex A activity (i. e., pseudodeficient) by means of in vitro biochemical tests have been described. We analyzed the HEXA gene of one pseudodeficient subject and identified both a C739-to-T substitution that changes Arg247----Trp on one allele and a previously identified Tay-Sachs disease mutation on the second allele. Six additional pseudodeficient subjects were found to have the C739-to-T mutation. This allele accounted for 32% (20/62) of non-Jewish enzyme-defined Tay-Sachs disease carriers but for none of 36 Jewish enzyme-defined carriers who did not have one of three known mutations common to this group. The C739-to-T allele, together with a " true " Tay-Sachs disease allele, causes Hex A pseudodeficiency. Given both the large proportion of non-Jewish carriers with this allele and that standard biochemical screening cannot differentiate between heterozygotes for the C739-to-T mutations and Tay-Sachs disease carriers, DNA testing for this mutation in at-risk couples is essential. 1384323 47 56 Tay-Sachs Modifier D013661 1384323 103 138 Deficiency of beta-hexosaminidase A SpecificDisease D013661 1384323 177 194 Tay-Sachs disease SpecificDisease D013661 1384323 234 252 deficient in Hex A SpecificDisease D013661 1384323 518 535 Tay-Sachs disease Modifier D013661 1384323 717 734 Tay-Sachs disease Modifier D013661 1384323 909 926 Tay-Sachs disease Modifier D013661 1384323 1153 1170 Tay-Sachs disease Modifier D013661 1307230|t|The presence of two different infantile Tay-Sachs disease mutations in a Cajun population. 1307230|a|A study was undertaken to characterize the mutation (s) responsible for Tay-Sachs disease (TSD) in a Cajun population in southwest Louisiana and to identify the origins of these mutations. Eleven of 12 infantile TSD alleles examined in six families had the beta-hexosaminidase A (Hex A) alpha-subunit exon 11 insertion mutation that is present in approximately 70% of Ashkenazi Jewish TSD heterozygotes. The mutation in the remaining allele was a single-base transition in the donor splice site of the alpha-subunit intron 9. To determine the origins of these two mutations in the Cajun population, the TSD carrier status was enzymatically determined for 90 members of four of the six families, and extensive pedigrees were constructed for all carriers. A single ancestral couple from France was found to be common to most of the carriers of the exon 11 insertion. Pedigree data suggest that this mutation has been in the Cajun population since its founding over 2 centuries ago and that it may be widely distributed within the population. In contrast, the intron 9 mutation apparently was introduced within the last century and probably is limited to a few Louisiana families.. 1307230 40 57 Tay-Sachs disease Modifier D013661 1307230 163 180 Tay-Sachs disease SpecificDisease D013661 1307230 182 185 TSD SpecificDisease D013661 1307230 303 306 TSD Modifier D013661 1307230 476 479 TSD Modifier D013661 1307230 694 697 TSD Modifier D013661 8088831|t|Canavan disease: genomic organization and localization of human ASPA to 17p13-ter and conservation of the ASPA gene during evolution. 8088831|a|Canavan disease, or spongy degeneration of the brain, is a severe leukodystrophy caused by the deficiency of aspartoacylase (ASPA). Recently, a missense mutation was identified in human ASPA coding sequence from patients with Canavan disease. The human ASPA gene has been cloned and found to span 29 kb of the genome. Human aspartoacylase is coded by six exons intervened by five introns. The exons vary from 94 (exon III) to 514 (exon VI) bases. The exon/intron splice junction sites follow the gt/ag consensus sequence rule. Southern blot analysis of genomic DNA from human/mouse somatic cell hybrid cell lines localized ASPA to human chromosome 17. The human ASPA locus was further mapped in the 17p13-ter region by fluorescence in situ hybridization. The bovine aspa gene has also been cloned, and its exon/intron organization is identical to that of the human gene. The 500-base sequence upstream of the initiator ATG codon in the human gene and that in the bovine gene are 77% identical. Human ASPA coding sequences cross-hybridize with genomic DNA from yeast, chicken, rabbit, cow, dog, mouse, rat, and monkey. The specificity of cross-species hybridization of coding sequences suggests that aspartoacylase has been conserved during evolution. It should now be possible to identify mutations in the noncoding genomic sequences that lead to Canavan disease and to study the regulation of ASPA.. 8088831 0 15 Canavan disease SpecificDisease D017825 8088831 134 149 Canavan disease SpecificDisease D017825 8088831 154 186 spongy degeneration of the brain SpecificDisease D017825 8088831 200 214 leukodystrophy SpecificDisease D007966 8088831 229 257 deficiency of aspartoacylase SpecificDisease D017825 8088831 360 375 Canavan disease SpecificDisease D017825 8088831 1481 1496 Canavan disease SpecificDisease D017825 6524872|t|Adrenoleukodystrophy: survey of 303 cases: biochemistry, diagnosis, and therapy. 6524872|a|Adrenoleukodystrophy (ALD) is a genetically determined disorder associated with progressive central demyelination and adrenal cortical insufficiency. All affected persons show increased levels of saturated unbranched very-long-chain fatty acids, particularly hexacosanoate (C26 0), because of impaired capacity to degrade these acids. This degradation normally takes place in a subcellular organelle called the peroxisome, and ALD, together with Zellwegers cerebrohepatorenal syndrome, is now considered to belong to the newly formed category of peroxisomal disorders. Biochemical assays permit prenatal diagnosis, as well as identification of most heterozygotes. We have identified 303 patients with ALD in 217 kindreds. These patients show a wide phenotypic variation. Sixty percent of patients had childhood ALD and 17% adrenomyeloneuropathy, both of which are X-linked, with the gene mapped to Xq28. Neonatal ALD, a distinct entity with autosomal recessive inheritance and points of resemblance to Zellwegers syndrome, accounted for 7% of the cases. Although excess C26 0 in the brain of patients with ALD is partially of dietary origin, dietary C26 0 restriction did not produce clear benefit. Bone marrow transplant lowered the plasma C26 0 level but failed to arrest neurological progression.. 6524872 0 20 Adrenoleukodystrophy SpecificDisease D000326 6524872 81 101 Adrenoleukodystrophy SpecificDisease D000326 6524872 103 106 ALD SpecificDisease D000326 6524872 161 194 progressive central demyelination DiseaseClass D003711 6524872 199 229 adrenal cortical insufficiency DiseaseClass D000309 6524872 509 512 ALD SpecificDisease D000326 6524872 528 566 Zellwegers cerebrohepatorenal syndrome SpecificDisease D015211 6524872 628 649 peroxisomal disorders DiseaseClass D018901 6524872 783 786 ALD SpecificDisease D000326 6524872 893 896 ALD SpecificDisease D000326 6524872 905 926 adrenomyeloneuropathy SpecificDisease D000326 6524872 986 998 Neonatal ALD SpecificDisease OMIM:202370 6524872 1084 1103 Zellwegers syndrome SpecificDisease D015211 6524872 1189 1192 ALD SpecificDisease D000326 10441343|t|French Machado-Joseph disease patients do not exhibit gametic segregation distortion: a sperm typing analysis. 10441343|a|Segregation distortion has been reported to occur in a number of the trinucleotide repeat disorders. On the basis of a sperm typing study performed in patients of Japanese descent with Machado-Joseph disease (MJD), it was reported that disease alleles are preferentially transmitted during meiosis. We performed a sperm typing study of five MJD patients of French descent and analysis of the pooled data shows a ratio of mutant to normal alleles of 379 436 (46. 5 53. 5%), which does not support meiotic segregation distortion. To confirm these results, sperm typing analysis was also performed using a polymorphic marker, D14S1050, closely linked to the MJD1 gene. Among 910 sperm analyzed, the allele linked to the disease chromosome was detected in 50. 3% of the samples and the allele linked to the normal chromosome was found in 49. 6% of the sperm. The difference in frequency of these two alleles is not significant (P = 0. 8423). Likelihood-based analysis of segregation distortion in the single sperm data using the SPERMSEG program also showed no support for segregation distortion at the gamete level in this patient population. The previous report on the Japanese patients also suggested that disease allele stability may be influenced by a trans effect of an intragenic polymorphism (987 G/C) in the wild-type allele. All of the French patients were heterozygous for this polymorphism. However, analysis of the variance in repeat number in sperm from the French MJD patients overlapped significantly with the variance in repeat number observed in the C/C homozygous Japanese patients. 10441343 7 29 Machado-Joseph disease Modifier D017827 10441343 180 210 trinucleotide repeat disorders DiseaseClass D030342 10441343 296 318 Machado-Joseph disease SpecificDisease D017827 10441343 320 323 MJD SpecificDisease D017827 10441343 452 455 MJD Modifier D017827 10441343 1588 1591 MJD Modifier D017827 3346018|t|Nebulin seen in DMD males including one patient with a large DNA deletion encompassing the DMD gene. 3346018|a|The presence of nebulin in a muscle specimen from a patient with Duchenne muscular dystrophy (DMD) due to a large deletion precludes the possibility that this protein is the DMD gene product.. 3346018 16 19 DMD Modifier D020388 3346018 91 94 DMD Modifier D020388 3346018 166 193 Duchenne muscular dystrophy SpecificDisease D020388 3346018 195 198 DMD SpecificDisease D020388 3346018 275 278 DMD Modifier D020388 8188241|t|The human gene for alkaptonuria (AKU) maps to chromosome 3q. 8188241|a|Alkaptonuria (AKU; McKusick no. 203500) is a rare autosomal recessive disorder caused by the lack of homogentisic acid oxidase activity. Patients excrete large amounts of homogentisic acid in their urine and a black ochronotic pigment is deposited in their cartilage and collagenous tissues. Ochronosis is the predominant clinical complication of the disease leading to ochronotic arthropathy, dark urine, pigment changes of the skin, and other clinical features. A mutation causing alkaptonuria in the mouse has mapped to chromosome 16. Considering conserved synteny, we were able to map the human gene to chromosome 3q in six alkaptonuria pedigrees of Slovak origin.. 8188241 19 31 alkaptonuria SpecificDisease D000474 8188241 33 36 AKU SpecificDisease D000474 8188241 61 73 Alkaptonuria SpecificDisease D000474 8188241 75 78 AKU SpecificDisease D000474 8188241 111 139 autosomal recessive disorder DiseaseClass D030342 8188241 353 363 Ochronosis SpecificDisease D009794 8188241 431 453 ochronotic arthropathy SpecificDisease D009794+D007592 8188241 544 556 alkaptonuria SpecificDisease D000474 8188241 689 701 alkaptonuria Modifier D000474 1317264|t|The APC gene, responsible for familial adenomatous polyposis, is mutated in human gastric cancer. 1317264|a|Although gastric cancer is the most common cancer in the world, genetic changes during its carcinogenesis are not well understood. Since some gastric cancers are considered to originate from the intestinal metaplasia, it is likely that the adenomatous polyposis coli (APC) gene, the mutation of which causes adenomatous polyps in the colon, is associated with carcinogenesis of gastric cancer. Based on this idea, DNAs isolated from gastric cancers were examined by means of a RNase protection analysis coupled with polymerase chain reaction followed by sequencing of the polymerase chain reaction products. By screening nearly one-half of the coding region of the APC gene in 44 tumors, somatic mutations were detected in three tumors a missense mutation, a nonsense mutation, and a 5-base pair deletion resulting in a frame shift which causes truncation of the gene product. These results suggest that the mutation of the APC gene also plays an important role during the carcinogenesis of at least some gastric cancers.. 1317264 4 7 APC Modifier D011125 1317264 30 60 familial adenomatous polyposis SpecificDisease D011125 1317264 82 96 gastric cancer SpecificDisease D013274 1317264 107 121 gastric cancer SpecificDisease D013274 1317264 141 147 cancer DiseaseClass D009369 1317264 240 255 gastric cancers SpecificDisease D013274 1317264 338 364 adenomatous polyposis coli Modifier D011125 1317264 366 369 APC Modifier D011125 1317264 406 437 adenomatous polyps in the colon SpecificDisease D018256 1317264 476 490 gastric cancer SpecificDisease D013274 1317264 531 546 gastric cancers SpecificDisease D013274 1317264 763 766 APC Modifier D011125 1317264 778 784 tumors DiseaseClass D009369 1317264 827 833 tumors DiseaseClass D009369 1317264 1023 1026 APC Modifier D011125 1317264 1104 1119 gastric cancers SpecificDisease D013274 10083734|t|Molecular epidemiology of C9 deficiency heterozygotes with an Arg95Stop mutation of the C9 gene in Japan. 10083734|a|Deficiency of the ninth component of human complement (C9) is the most common complement deficiency in Japan, with an incidence of approximately one homozygote in 1000, but is very rare in other countries. Genetic analyses of Japanese C9 deficiency have shown that a C-to-T transition leading to TGA stop codon for Arg95 in exon 4 of the C9 gene (Arg95Stop) is common in Japanese C9 deficiency. To determine the prevalence of heterozygous carriers of the Arg95Stop mutation in a Japanese population, we collected DNA samples from 300 individuals in two of the four main islands of Japan. Heterozygote detection was performed with an allele-specific polymerase chain reaction (PCR) system designed to detect exclusively only one of the normal and mutant alleles, followed by confirmation with PCR/single-strand conformation polymorphism (SSCP) analysis and direct sequencing. Twenty individuals were heterozygous for the Arg95Stop mutation. None was homozygous. The prevalence of carriers of the Arg95Stop mutation was 6. 7% (20/300). An estimated frequency (0. 12%) of complete C9 deficiency due to homozygous Arg95Stop mutation was consistent with frequencies determined by serological studies 10083734 26 39 C9 deficiency Modifier OMIM:613825 10083734 106 159 Deficiency of the ninth component of human complement SpecificDisease OMIM:613825 10083734 184 205 complement deficiency DiseaseClass D007153 10083734 341 354 C9 deficiency SpecificDisease OMIM:613825 10083734 486 499 C9 deficiency SpecificDisease OMIM:613825 10083734 1184 1197 C9 deficiency SpecificDisease OMIM:613825 2760209|t|Molecular analysis of a female Lesch-Nyhan patient. 2760209|a|We report the identification of a female patient with the X-linked recessive Lesch-Nyhan syndrome (hypoxanthine phosphoribosyltransferase [HPRT] deficiency). Cytogenetic and carrier studies revealed structurally normal chromosomes for this patient and her parents and demonstrated that this mutation arose through a de novo gametic event. Comparison of this patients DNA with the DNA of her parents revealed that a microdeletion, which occurred within a maternal gamete and involved the entire HPRT gene, was partially responsible for the disease in this patient. Somatic cell hybrids, generated to separate maternal and paternal X chromosomes, showed that expression of two additional X-linked enzymes, phosphoglycerate kinase and glucose-6-phosphate dehydrogenase, were expressed only in cells that contained the maternal X chromosome, suggesting the presence of a functionally inactive paternal X chromosome. Furthermore, comparison of methylation patterns within a region of the HPRT gene known to be important in gene regulation revealed differences between DNA from the father and the patient, in keeping with an active HPRT locus in the father and an inactive HPRT locus in the patient. Together these data indicate that nonrandom inactivation of the cytogenetically normal paternal X chromosome and a microdeletion of the HPRT gene on an active maternal X chromosome were responsible for the absence of HPRT in this patient.. 2760209 31 42 Lesch-Nyhan Modifier D007926 2760209 129 149 Lesch-Nyhan syndrome SpecificDisease D007926 2760209 151 207 hypoxanthine phosphoribosyltransferase [HPRT] deficiency SpecificDisease D007926 10323252|t|Changes at P183 of emerin weaken its protein-protein interactions resulting in X-linked Emery-Dreifuss muscular dystrophy. 10323252|a|Emery-Dreifuss muscular dystrophy (EDMD) is an X-linked recessive muscular dystrophy characterized by early contractures of the elbows, Achilles tendons and spine, slowly progressive muscle wasting and weakness, and cardiomyopathy associated with cardiac conduction defects. The emerin gene has been mapped to Xq28 and encodes a 34-kDa serine-rich protein, emerin, which has been localized to the nuclear envelope in a wide variety of tissues, including skeletal and cardiac muscle. Mutations spanning the emerin gene have been identified in patients with EDMD. We present here the effect, on emerin protein expression, of two missense mutations identified in unrelated EDMD patients. These alterations predict the replacement of a proline residue at position 183 with either a histidine or a threonine. Biochemical analysis has demonstrated that the mobility and expression levels of the mutant forms of emerin are indistinguishable from that of wild-type emerin, but that they have weakened interactions with nuclear lamina components. In comparison with the usual EDMD phenotype, patients with P183 missense mutations have a later age at onset of first symptoms, elbow contractures, ankle contractures, upper limb weakness and lower limb weakness, but there is no difference for the age at onset of cardiac involvement. This is the first report of protein studies on patients with missense mutations resulting in the clinical features of EDMD. These studies demonstrate the importance of proline 183 for the proper structure/function of emerin.. 10323252 79 121 X-linked Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10323252 123 156 Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10323252 158 162 EDMD SpecificDisease D020389 10323252 170 207 X-linked recessive muscular dystrophy DiseaseClass D040181+D009136 10323252 231 285 contractures of the elbows, Achilles tendons and spine CompositeMention D003286 10323252 306 320 muscle wasting DiseaseClass D009133 10323252 325 333 weakness DiseaseClass D018908 10323252 339 353 cardiomyopathy SpecificDisease D009202 10323252 370 396 cardiac conduction defects SpecificDisease OMIM:115080 10323252 679 683 EDMD SpecificDisease D020389 10323252 793 797 EDMD Modifier D020389 10323252 1190 1194 EDMD Modifier D020389 10323252 1289 1307 elbow contractures SpecificDisease D003286 10323252 1309 1327 ankle contractures SpecificDisease D003286 10323252 1329 1348 upper limb weakness SpecificDisease D018908 10323252 1353 1372 lower limb weakness SpecificDisease D018908 10323252 1425 1444 cardiac involvement DiseaseClass D006331 10323252 1564 1568 EDMD SpecificDisease D020389 7795653|t|Decreased expression of BRCA1 accelerates growth and is often present during sporadic breast cancer progression. 7795653|a|We have characterized expression of the familial breast and ovarian cancer gene, BRCA1, in cases of non-hereditary (sporadic) breast cancer and analyzed the effect of antisense inhibition of BRCA1 on the proliferative rate of mammary epithelial cells. BRCA1 mRNA levels are markedly decreased during the transition from carcinoma in situ to invasive cancer. Experimental inhibition of BRCA1 expression with antisense oligonucleotides produced accelerated growth of normal and malignant mammary cells, but had no effect on non-mammary epithelial cells. These studies suggest that BRCA1 may normally serve as a negative regulator of mammary epithelial cell growth whose function is compromised in breast cancer either by direct mutation or alterations in gene expression.. 7795653 77 99 sporadic breast cancer Modifier D001943 7795653 153 187 familial breast and ovarian cancer Modifier D061325 7795653 213 252 non-hereditary (sporadic) breast cancer SpecificDisease D001943 7795653 433 450 carcinoma in situ DiseaseClass D002278 7795653 454 469 invasive cancer DiseaseClass D009362 7795653 808 821 breast cancer SpecificDisease D001943 10827108|t|Additional copies of the proteolipid protein gene causing Pelizaeus-Merzbacher disease arise by separate integration into the X chromosome. 10827108|a|The proteolipid protein gene (PLP) is normally present at chromosome Xq22. Mutations and duplications of this gene are associated with Pelizaeus-Merzbacher disease (PMD). Here we describe two new families in which males affected with PMD were found to have a copy of PLP on the short arm of the X chromosome, in addition to a normal copy on Xq22. In the first family, the extra copy was first detected by the presence of heterozygosity of the AhaII dimorphism within the PLP gene. The results of FISH analysis showed an additional copy of PLP in Xp22. 1, although no chromosomal rearrangements could be detected by standard karyotype analysis. Another three affected males from the family had similar findings. In a second unrelated family with signs of PMD, cytogenetic analysis showed a pericentric inversion of the X chromosome. In the inv (X) carried by several affected family members, FISH showed PLP signals at Xp11. 4 and Xq22. A third family has previously been reported, in which affected members had an extra copy of the PLP gene detected at Xq26 in a chromosome with an otherwise normal banding pattern. The identification of three separate families in which PLP is duplicated at a noncontiguous site suggests that such duplications could be a relatively common but previously undetected cause of genetic disorders. 10827108 58 86 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 10827108 275 303 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 10827108 305 308 PMD SpecificDisease OMIM:312080 10827108 374 377 PMD SpecificDisease OMIM:312080 10827108 894 897 PMD SpecificDisease OMIM:312080 10827108 1449 1466 genetic disorders DiseaseClass D030342 1975560|t|Linkage relationships of the apolipoprotein C1 gene and a cytochrome P450 gene (CYP2A) to myotonic dystrophy. 1975560|a|We have studied the genetic linkage of two markers, the apolipoprotein C1 (APOC1) gene and a cytochrome P450 (CYP2A) gene, in relation to the gene for myotonic dystrophy (DM). A peak lod score of 9. 29 at 2 cM was observed for APOC1-DM, with a lod score of 8. 55 at 4 cM for CYP2A-DM. These two markers also show close linkage to each other (theta max = 0. 05, Zmax = 9. 09). From examination of the genotypes of the recombinant individuals, CYP2A appears to map proximal to DM because in one recombinant individual CYP2A, APOC2 and CKMM had all recombined with DM. Evidence from another CYP2A-DM recombinant individual places CYP2A proximal to APOC2 and CKMM. Localisation of CYP2A on a panel of somatic cell hybrids also suggests that it is proximal to DM and APOC2/C1/E gene cluster. 1975560 90 108 myotonic dystrophy SpecificDisease D009223 1975560 261 279 myotonic dystrophy SpecificDisease D009223 1975560 281 283 DM SpecificDisease D009223 10737981|t|beta-galactosidase gene mutations affecting the lysosomal enzyme and the elastin-binding protein in GM1-gangliosidosis patients with cardiac involvement. 10737981|a|GM1-gangliosidosis is a lysosomal storage disorder caused by deficiency of acid beta-galactosidase (GLB1). We report five new beta-galactosidase gene mutations in nine Italian patients and one fetus, segregating in seven unrelated families. Six of the eight patients with the infantile, severe form of the disease presented cardiac involvement, a feature rarely associated with GM1-gangliosidosis. Molecular analysis of the patients RNA and DNA identified two new RNA splicing defects, three new and three previously described amino acid substitutions. Interestingly, all patients with cardiac involvement were homozygous for one of these mutations R59H, Y591C, Y591N, or IVS14-2A > G. In contrast, all other patients were compound heterozygous for one of the following mutations R201H, R482H, G579D, IVS8 + 2T > C. Although we could not directly correlate the presence of cardiac abnormalities with specific genetic lesions, the mutations identified in patients with cardiomyopathy fell in the GLB1 cDNA region common to the lysosomal enzyme and the Hbeta-Gal-related protein, also known as the elastin binding protein (EBP). Consequently, both molecules are affected by the mutations, and they may contribute differently to the occurrence of specific clinical manifestations.. 10737981 100 118 GM1-gangliosidosis Modifier D016537 10737981 133 152 cardiac involvement SpecificDisease D006331 10737981 154 172 GM1-gangliosidosis SpecificDisease D016537 10737981 178 204 lysosomal storage disorder DiseaseClass D016464 10737981 215 252 deficiency of acid beta-galactosidase SpecificDisease D016537 10737981 478 497 cardiac involvement DiseaseClass D006331 10737981 532 550 GM1-gangliosidosis SpecificDisease D016537 10737981 740 759 cardiac involvement DiseaseClass D006331 10737981 1029 1050 cardiac abnormalities DiseaseClass D018376 10737981 1065 1080 genetic lesions DiseaseClass D020022 10737981 1124 1138 cardiomyopathy DiseaseClass D009202 1679035|t|Exclusion of linkage between familial Mediterranean fever and the human serum amyloid A (SAA) gene cluster. 1679035|a|We studied the relationship between the autosomal recessive trait familial Mediterranean fever (FMF) and the serum amyloid A (SAA) genes by comparing alleles of a highly polymorphic dinucleotide repeat and a conventional restriction fragment length polymorphism (RFLP) in the SAA gene cluster in Israeli FMF kindreds. By haplotype analysis, our data indicate a minimum crossover frequency of 22% between the SAA gene marker and FMF. By conventional linkage analysis this eliminates a minimum of 10. 4 cM including and surrounding the SAA gene cluster as the site of the FMF mutation although SAA proteins are prominent physiologic markers of the acute attacks. 1679035 29 57 familial Mediterranean fever SpecificDisease D010505 1679035 174 202 familial Mediterranean fever SpecificDisease D010505 1679035 204 207 FMF SpecificDisease D010505 1679035 412 415 FMF Modifier D010505 1679035 536 539 FMF SpecificDisease D010505 1679035 678 681 FMF Modifier D010505 10802667|t|Mice deficient in Six5 develop cataracts: implications for myotonic dystrophy. 10802667|a|Expansion of a CTG trinucleotide repeat in the 3 UTR of the gene DMPK at the DM1 locus on chromosome 19 causes myotonic dystrophy, a dominantly inherited disease characterized by skeletal muscle dystrophy and myotonia, cataracts and cardiac conduction defects. Targeted deletion of Dm15, the mouse orthologue of human DMPK, produced mice with a mild myopathy and cardiac conduction abnormalities, but without other features of myotonic dystrophy, such as myotonia and cataracts. We, and others, have demonstrated that repeat expansion decreases expression of the adjacent gene SIX5 (refs 7, 8), which encodes a homeodomain transcription factor. To determine whether SIX5 deficiency contributes to the myotonic dystrophy phenotype, we disrupted mouse Six5 by replacing the first exon with a beta-galactosidase reporter. Six5-mutant mice showed reporter expression in multiple tissues, including the developing lens. Homozygous mutant mice had no apparent abnormalities of skeletal muscle function, but developed lenticular opacities at a higher rate than controls. Our results suggest that SIX5 deficiency contributes to the cataract phenotype in myotonic dystrophy, and that myotonic dystrophy represents a multigenic disorder.. 10802667 5 22 deficient in Six5 SpecificDisease D030342 10802667 31 40 cataracts SpecificDisease D002386 10802667 59 77 myotonic dystrophy SpecificDisease D009223 10802667 190 208 myotonic dystrophy SpecificDisease D009223 10802667 212 240 dominantly inherited disease DiseaseClass D030342 10802667 267 283 muscle dystrophy SpecificDisease D009136 10802667 288 296 myotonia DiseaseClass D009222 10802667 298 307 cataracts SpecificDisease D002386 10802667 312 338 cardiac conduction defects DiseaseClass OMIM:115080 10802667 429 437 myopathy DiseaseClass D009135 10802667 442 474 cardiac conduction abnormalities DiseaseClass D006327 10802667 506 524 myotonic dystrophy SpecificDisease D009223 10802667 534 542 myotonia DiseaseClass D009222 10802667 547 556 cataracts SpecificDisease D002386 10802667 745 760 SIX5 deficiency SpecificDisease D030342 10802667 780 798 myotonic dystrophy Modifier D009223 10802667 1033 1074 abnormalities of skeletal muscle function DiseaseClass D009139 10802667 1090 1110 lenticular opacities SpecificDisease D002386 10802667 1168 1183 SIX5 deficiency SpecificDisease D030342 10802667 1203 1211 cataract Modifier D002386 10802667 1225 1243 myotonic dystrophy SpecificDisease D009223 10802667 1254 1272 myotonic dystrophy SpecificDisease D009223 10802667 1286 1305 multigenic disorder DiseaseClass D030342 1338764|t|Screening for germ-line mutations in familial adenomatous polyposis patients: 61 new patients and a summary of 150 unrelated patients. 1338764|a|We report here the result of a screening for germ-line mutations in the adenomatous polyposis coli (APC) gene in 61 new familial adenomatous polyposis (FAP) patients as well as a summary of the results of 150 patients. Examination of the entire coding region of the APC gene, based on a ribonuclease protection assay coupled with the polymerase chain reaction (PCR), disclosed mutations that were considered to cause significant defects in the APC product in 97 of 150 unrelated FAP patients. Our findings revealed the following characteristics of the germ-line mutations of APC 1) the great majority of the mutations were found to truncate the APC product; 2) almost all of the mutations were located within the first half of the coding region; 3) no correlation was observed between the locations of germ-line mutations and extracolonic manifestations in FAP patients; 4) more than 80% of base substitutions in the APC gene were from cytosine to other nucleotides, nearly one-third of which occurred at the GpG site. Our results provide information helpful to an understanding of the APC gene and will also contribute to presymptomatic diagnosis of members in FAP families.. 1338764 37 67 familial adenomatous polyposis Modifier D011125 1338764 207 233 adenomatous polyposis coli Modifier D011125 1338764 235 238 APC Modifier D011125 1338764 255 285 familial adenomatous polyposis Modifier D011125 1338764 287 290 FAP Modifier D011125 1338764 401 404 APC Modifier D011125 1338764 579 582 APC Modifier D011125 1338764 614 617 FAP Modifier D011125 1338764 781 784 APC Modifier D011125 1338764 993 996 FAP Modifier D011125 1338764 1053 1056 APC Modifier D011125 1338764 1222 1225 APC Modifier D011125 1338764 1298 1301 FAP Modifier D011125 1999552|t|Hereditary deficiency of C5 in association with discoid lupus erythematosus. 1999552|a|A 29-year-old woman with discoid lupus erythematosus had undetectable classic pathway complement activity. Hypocomplementemia was due to selective deficiency of C5. One of her children was also deficient. To our knowledge this is the first documented case of an association between discoid lupus erythematosus and C5 deficiency.. 1999552 11 27 deficiency of C5 SpecificDisease OMIM:609536 1999552 48 75 discoid lupus erythematosus SpecificDisease D008179 1999552 102 129 discoid lupus erythematosus SpecificDisease D008179 1999552 184 202 Hypocomplementemia DiseaseClass D007153 1999552 224 240 deficiency of C5 SpecificDisease OMIM:609536 1999552 359 386 discoid lupus erythematosus SpecificDisease D008179 1999552 391 404 C5 deficiency SpecificDisease OMIM:609536 10818206|t|Isolation, genomic organization, and expression analysis of the mouse and rat homologs of MEFV, the gene for familial mediterranean fever. 10818206|a|Familial Mediterranean fever (FMF) is a recessive disorder characterized by episodes of fever with serositis or synovitis. Recently the FMF gene (MEFV) was cloned; the protein product, pyrin/marenostrin, is thought to regulate inflammation in myeloid cells. In this manuscript we report the mouse and rat homologs of MEFV. The murine gene contains ten exons with a coding sequence of 2304 bp, while the rat homolog has nine exons with a coding sequence of 2253 bp. A considerable amino acid sequence homology was observed between the mouse and human (47. 6% identity and 65. 5% similarity) and between the mouse and rat genes (73. 5% identity and 82. 1% similarity). The predicted rodent proteins have several important domains and signals found in human pyrin, including a B-box zinc finger domain, Robbins-Dingwall nuclear localization signal, and coiled-coil domain. However, perhaps because of an ancient frame-shift mutation, neither the mouse nor the rat protein has an intact C-terminal B30. 2 domain, in which most FMF-associated mutations have been found in human MEFV. Nevertheless, like the human gene, mouse Mefv is expressed in peripheral blood granulocytes but not lymphocytes. Consistent with its expression in granulocytes, Mefv was detected at high levels in the primary follicles and marginal zones of the splenic white pulp. Mefv is localized on mouse Chromosome (Chr) 16, region A3-B1, extending a region of synteny with human Chr 16p13. 3. Development of knockout and knockin mouse models may provide further insights into the functional evolution of this gene. 10818206 109 137 familial mediterranean fever SpecificDisease D010505 10818206 139 167 Familial Mediterranean fever SpecificDisease D010505 10818206 169 172 FMF SpecificDisease D010505 10818206 179 197 recessive disorder DiseaseClass D030342 10818206 227 232 fever DiseaseClass D005334 10818206 238 247 serositis SpecificDisease D012700 10818206 251 260 synovitis SpecificDisease D013585 10818206 275 278 FMF Modifier D010505 10818206 1162 1165 FMF Modifier D010505 1684569|t|Analysis of X-chromosome inactivation and presumptive expression of the Wiskott-Aldrich syndrome (WAS) gene in hematopoietic cell lineages of a thrombocytopenic carrier female of WAS. 1684569|a|We report on a thrombocytopenic female belonging to a pedigree with the Wiskott-Aldrich syndrome (WAS). Restriction fragment length polymorphism (RFLP) analysis with probe M27 beta, closely linked to the WAS gene, demonstrated that she is a carrier of WAS. Both small-sized and normal-sized platelets were present, suggesting that, unlike the vast majority of WAS carriers, she does not manifest nonrandom X-chromosome inactivation in the thrombopoietic cell lineage. Study of X-chromosome inactivation by means of RFLP and methylation analysis demonstrated that the pattern of X-chromosome inactivation was nonrandom in T lymphocytes, but random in granulocytes. While this is the first complete report on the occurrence of thrombocytopenia in a carrier female of WAS as the result of atypical lyonization, it also suggests that expression of the WAS gene occurs at (or extends up to) a later stage than the multipotent stem cell along the hematopoietic differentiation pathway.. 1684569 72 96 Wiskott-Aldrich syndrome Modifier D014923 1684569 98 101 WAS Modifier D014923 1684569 179 182 WAS SpecificDisease D014923 1684569 199 215 thrombocytopenic Modifier D013921 1684569 256 280 Wiskott-Aldrich syndrome SpecificDisease D014923 1684569 282 285 WAS SpecificDisease D014923 1684569 388 391 WAS Modifier D014923 1684569 436 439 WAS SpecificDisease D014923 1684569 544 547 WAS Modifier D014923 1684569 909 925 thrombocytopenia SpecificDisease D013921 1684569 949 952 WAS SpecificDisease D014923 1684569 1032 1035 WAS Modifier D014923 10724175|t|hCds1-mediated phosphorylation of BRCA1 regulates the DNA damage response. 10724175|a|Mutations in the BRCA1 (ref. 1) tumour suppressor gene are found in almost all of the families with inherited breast and ovarian cancers and about half of the families with only breast cancer. Although the biochemical function of BRCA1 is not well understood, it is important for DNA damage repair and cell-cycle checkpoint. BRCA1 exists in nuclear foci but is hyperphosphorylated and disperses after DNA damage. It is not known whether BRCA1 phosphorylation and dispersion and its function in DNA damage response are related. In yeast the DNA damage response and the replication-block checkpoint are mediated partly through the Cds1 kinase family. Here we report that the human Cds1 kinase (hCds1/Chk2) regulates BRCA1 function after DNA damage by phosphorylating serine 988 of BRCA1. We show that hCds1 and BRCA1 interact and co-localize within discrete nuclear foci but separate after gamma irradiation. Phosphorylation of BRCA1 at serine 988 is required for the release of BRCA1 from hCds1. This phosphorylation is also important for the ability of BRCA1 to restore survival after DNA damage in the BRCA1-mutated cell line HCC1937.. 10724175 107 113 tumour Modifier D009369 10724175 175 211 inherited breast and ovarian cancers CompositeMention D061325 10724175 253 266 breast cancer SpecificDisease D001943 10556285|t|Ataxin-3 with an altered conformation that exposes the polyglutamine domain is associated with the nuclear matrix. 10556285|a|Spinocerebellar ataxia type-3 or Machado-Joseph disease (SCA3/MJD) is a member of the CAG/polyglutamine repeat disease family. In this family of disorders, a normally polymorphic CAG repeat becomes expanded, resulting in expression of an expanded polyglutamine domain in the disease gene product. Experimental models of polyglutamine disease implicate the nucleus in pathogenesis; however, the link between intranuclear expression of expanded polyglutamine and neuronal dysfunction remains unclear. Here we demonstrate that ataxin-3, the disease protein in SCA3/MJD, adopts a unique conformation when expressed within the nucleus of transfected cells. The monoclonal antibody 1C2 is known preferentially to bind expanded polyglutamine, but we find that it also binds a fragment of ataxin-3 containing a normal glutamine repeat. In addition, expression of ataxin-3 within the nucleus exposes the glutamine domain of the full-length non-pathological protein, allowing it to bind the monoclonal antibody 1C2. Fractionation and immunochemical experiments indicate that this novel conformation of intranuclear ataxin-3 is not due to proteolysis, suggesting instead that association with nuclear protein (s) alters the structure of full-length ataxin-3 which exposes the polyglutamine domain. This conformationally altered ataxin-3 is bound to the nuclear matrix. The pathological form of ataxin-3 with an expanded polyglutamine domain also associates with the nuclear matrix. These data suggest that an early event in the pathogenesis of SCA3/MJD may be an altered conformation of ataxin-3 within the nucleus that exposes the polyglutamine domain.. 10556285 115 144 Spinocerebellar ataxia type-3 SpecificDisease D017827 10556285 148 170 Machado-Joseph disease SpecificDisease D017827 10556285 172 176 SCA3 SpecificDisease D017827 10556285 177 180 MJD SpecificDisease D017827 10556285 201 233 CAG/polyglutamine repeat disease DiseaseClass D030342 10556285 435 456 polyglutamine disease DiseaseClass D030342 10556285 576 596 neuronal dysfunction DiseaseClass D009461 10556285 672 676 SCA3 SpecificDisease D017827 10556285 677 680 MJD SpecificDisease D017827 10556285 1648 1652 SCA3 SpecificDisease D017827 10556285 1653 1656 MJD SpecificDisease D017827 10077614|t|A zinc finger truncation of murine WT1 results in the characteristic urogenital abnormalities of Denys-Drash syndrome . 10077614|a|The Wilms tumor -suppressor gene, WT1, plays a key role in urogenital development, and WT1 dysfunction is implicated in both neoplastic (Wilms tumor, mesothelioma, leukemias, and breast cancer) and nonneoplastic (glomerulosclerosis) disease. The analysis of diseases linked specifically with WT1 mutations, such as Denys-Drash syndrome (DDS), can provide valuable insight concerning the role of WT1 in development and disease. DDS is a rare childhood disease characterized by a nephropathy involving mesangial sclerosis, XY pseudohermaphroditism, and/or Wilms tumor (WT). DDS patients are constitutionally heterozygous for exonic point mutations in WT1, which include mutations predicted to truncate the protein within the C-terminal zinc finger (ZF) region. We report that heterozygosity for a targeted murine Wt1 allele, Wt1 (tmT396), which truncates ZF3 at codon 396, induces mesangial sclerosis characteristic of DDS in adult heterozygous and chimeric mice. Male genital defects also were evident and there was a single case of Wilms tumor in which the transcript of the nontargeted allele showed an exon 9 skipping event, implying a causal link between Wt1 dysfunction and Wilms tumorigenesis in mice. However, the mutant WT1 (tmT396) protein accounted for only 5% of WT1 in both heterozygous embryonic stem cells and the WT. This has implications regarding the mechanism by which the mutant allele exerts its effect. 10077614 69 93 urogenital abnormalities DiseaseClass D014564 10077614 97 117 Denys-Drash syndrome SpecificDisease D030321 10077614 124 135 Wilms tumor Modifier D009396 10077614 207 222 WT1 dysfunction SpecificDisease D030342 10077614 245 255 neoplastic Modifier D009369 10077614 257 268 Wilms tumor SpecificDisease D009396 10077614 270 282 mesothelioma SpecificDisease D008654 10077614 284 293 leukemias SpecificDisease D007938 10077614 299 312 breast cancer SpecificDisease D001943 10077614 318 331 nonneoplastic Modifier D004194 10077614 333 351 glomerulosclerosis SpecificDisease D005921 10077614 435 455 Denys-Drash syndrome SpecificDisease D030321 10077614 457 460 DDS SpecificDisease D030321 10077614 547 550 DDS SpecificDisease D030321 10077614 598 609 nephropathy SpecificDisease D007674 10077614 620 639 mesangial sclerosis SpecificDisease C537346 10077614 644 665 pseudohermaphroditism SpecificDisease D012734 10077614 674 685 Wilms tumor SpecificDisease D009396 10077614 687 689 WT SpecificDisease D009396 10077614 692 695 DDS Modifier D030321 10077614 999 1018 mesangial sclerosis SpecificDisease C537346 10077614 1037 1040 DDS SpecificDisease D030321 10077614 1082 1102 Male genital defects DiseaseClass D005832 10077614 1152 1163 Wilms tumor SpecificDisease D009396 10077614 1278 1293 Wt1 dysfunction SpecificDisease D030342 10077614 1298 1317 Wilms tumorigenesis SpecificDisease D009396 10077614 1447 1449 WT SpecificDisease D009396 7106752|t|Heterogeneity of "Mediterranean type" glucose-6-phosphate dehydrogenase (G6PD) deficiency in Spain and description of two new variants associated with favism. 7106752|a|Glucose-6-phosphate dehydrogenase (G6PD); EC 1. 1. 1. 49 from thirty-six unrelated Spanish males was partially purified from blood, and the variants were characterized biochemically and electrophoretically according to the methods recommended by the world Health Organization. Subjects were from multiple geographic regions within Spain, and all suffered from hemolytic anemia, either acute (34 cases) or chronic nonspherocytic (2 cases). Almost all the variants studied presented residual erythrocyte G6PD activity ranging from 0 to 10% of normal, and five different mutants were responsible for the deficient phenotype. Three variants were similar to others previously described G6PD Mediterranean (11 cases), G6PD Athens-like (3 cases), and G6PD Union (2 cases). The remaining variants were different from the numerous variants already reported and have been considered as new mutants. Provisionally they are called G6PD Betica (19 cases) and G6PD Menorca (1 case). The present study constitutes the first attempt to characterize the deficient G6PD variants found in Spain and supplies new data on the relationship between molecular characteristics of deficient variants and their clinical manifestations. The most important findings can be summarized as follows (1) The Spanish population is characterized by an important heterogeneity in G6PD deficiency. (2) Although G6PD Mediterranean is very frequent, it presents a relatively high degree of polymorphism. (3) Favism has been observed associated with all kinds of variants described here. (4) G6PD Betica, which is the most frequent variant found in subjects of Southern Spanish origin, has been observed associated with favism in all cases except one. 7106752 38 89 glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 7106752 151 157 favism SpecificDisease D005236 7106752 519 535 hemolytic anemia SpecificDisease D000743 7106752 1197 1211 deficient G6PD Modifier D005955 7106752 1504 1519 G6PD deficiency SpecificDisease D005955 7106752 1629 1635 Favism SpecificDisease D005236 7106752 1840 1846 favism SpecificDisease D005236 10480214|t|Early onset of X-linked Emery-Dreifuss muscular dystrophy in a boy with emerin gene deletion. 10480214|a|A boy developed contractures of the Achilles tendons at 3 years and of the postcervical muscles at 7 years, although neither contractures of the elbows nor cardiac abnormality were recognized by the age of 9 years. Muscle computed tomography scanning revealed changes characteristic of muscle involvement. Emerin was not detected in the biopsied muscle, and RT-PCR and PCR-based genomic DNA analyses of the emerin gene demonstrated no amplification product in the patient. These results confirmed the diagnosis of X-linked Emery-Dreifuss muscular dystrophy (EDMD), and reinforce the necessity of molecular genetic diagnosis of the membrane protein emerin in younger patients with possible EDMD before appearance of the typical symptoms, to avoid sudden cardiac death.. 10480214 15 57 X-linked Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10480214 110 146 contractures of the Achilles tendons DiseaseClass D003286 10480214 219 245 contractures of the elbows DiseaseClass D003286 10480214 250 269 cardiac abnormality DiseaseClass D018376 10480214 608 650 X-linked Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10480214 652 656 EDMD SpecificDisease D020389 10480214 783 787 EDMD SpecificDisease D020389 10480214 840 860 sudden cardiac death SpecificDisease D016757 1682919|t|PRAD1, a candidate BCL1 oncogene: mapping and expression in centrocytic lymphoma. 1682919|a|Rearrangement of the BCL1 (B-cell lymphoma 1) region on chromosome 11q13 appears to be highly characteristic of centrocytic lymphoma and also is found infrequently in other B-cell neoplasms. Rearrangement is thought to deregulate a nearby protooncogene, but transcribed sequences in the immediate vicinity of BCL1 breakpoints had not been identified. PRAD1, previously designated D11S287E, was identified on 11q13 as a chromosomal breakpoint region rearranged with the parathyroid hormone gene in a subset of parathyroid adenomas; this highly conserved putative oncogene, which encodes a novel cyclin, has been linked to BCL1 and implicated also in subsets of breast and squamous cell neoplasms with 11q13 amplification. We report pulsed-field gel electrophoresis data showing BCL1 and PRAD1 to be no more than 130 kilobases apart. PRAD1 mRNA is abundantly expressed in seven of seven centrocytic lymphomas (Kiel classification), in contrast to 13 closely related but noncentrocytic lymphomas. Three of the seven centrocytic lymphomas had detectable BCL1 DNA rearrangement. Also, two unusual cases of CLL with BCL1 rearrangement overexpressed PRAD1, in contrast to five CLL controls. Thus, PRAD1 is an excellent candidate " BCL1 oncogene. " Its overexpression may be a key consequence of rearrangement of the BCL1 vicinity in B-cell neoplasms and a unifying pathogenetic feature in centrocytic lymphoma. 1682919 60 80 centrocytic lymphoma DiseaseClass D008223 1682919 194 214 centrocytic lymphoma DiseaseClass D008223 1682919 255 271 B-cell neoplasms SpecificDisease D016393 1682919 591 611 parathyroid adenomas DiseaseClass D010282 1682919 742 776 breast and squamous cell neoplasms CompositeMention D001943|D018307 1682919 967 988 centrocytic lymphomas DiseaseClass D008223 1682919 1050 1074 noncentrocytic lymphomas DiseaseClass D008223 1682919 1095 1116 centrocytic lymphomas DiseaseClass D008223 1682919 1408 1424 B-cell neoplasms SpecificDisease D016393 1682919 1464 1484 centrocytic lymphoma DiseaseClass D008223 7202134|t|Adrenoleukodystrophy: increased plasma content of saturated very long chain fatty acids. 7202134|a|With a new method we measured the saturated very long chain fatty acids in the plasma of adrenoleukodystrophy (ALD) hemizygotes, ALD heterozygotes, and controls. ALD hemizygotes showed increased levels of hexacosanoate (C26 fatty acid) which represented 0. 081 +/- 0. 0066% (SEM) of total fatty acids, compared to 0. 015 +/- 0. 0032% in the controls. C25, C24, and C23 fatty acids were also increased, but the C22 and C20 fatty acids were normal. C26 levels were also increased in most ALD heterozygotes, with a mean level 0. 057 +/- 0. 0063% of total fatty acids. The technique can be used for diagnosis and carrier identification, and in the evaluation of therapy. 7202134 0 20 Adrenoleukodystrophy SpecificDisease D000326 7202134 178 198 adrenoleukodystrophy Modifier D000326 7202134 200 203 ALD Modifier D000326 7202134 218 221 ALD Modifier D000326 7202134 251 254 ALD Modifier D000326 7202134 575 578 ALD Modifier D000326 6337374|t|Glucose-6-phosphate dehydrogenase deficiency inhibits in vitro growth of Plasmodium falciparum. 6337374|a|Glucose-6-phosphate dehydrogenase (G6PD; EC 1. 1. 1. 49) -deficient red blood cells from male hemizygotes and female heterozygotes from the island of Sardinia were studied for their ability to support growth in vitro of the malaria-causing organism Plasmodium falciparum. Parasite growth was approximately one-third of normal in both hemi- and heterozygotes for G6PD deficiency. In Sardinians with the beta 0-thalassemia trait, parasite growth was normal except when G6PD deficiency occurred together with the thalassemia trait. The data support the hypothesis that G6PD deficiency may confer a selective advantage in a malarious area; the female heterozygote may be at a particular advantage because resistance to malaria equals that of male hemizygotes, but the risk of fatal hemolysis may be less. However, more female heterozygotes must be studied to confirm this hypothesis. No protective effect of beta 0-thalassemia trait could be demonstrated in vitro. 6337374 0 44 Glucose-6-phosphate dehydrogenase deficiency SpecificDisease D005955 6337374 96 163 Glucose-6-phosphate dehydrogenase (G6PD; EC 1. 1. 1. 49) -deficient Modifier D005955 6337374 320 327 malaria Modifier D008288 6337374 458 473 G6PD deficiency SpecificDisease D005955 6337374 563 578 G6PD deficiency SpecificDisease D005955 6337374 606 617 thalassemia Modifier D013789 6337374 662 677 G6PD deficiency SpecificDisease D005955 6337374 716 725 malarious Modifier D008288 6337374 811 818 malaria SpecificDisease D008288 6337374 868 883 fatal hemolysis SpecificDisease D006461 3718019|t|Heterozygous C2 deficiency associated with angioedema, myasthenia gravis, and systemic lupus erythematosus. 3718019|a|We describe a patient with myasthenia gravis, systemic lupus erythematosus, and angioedema associated with heterozygous complement factor 2 (C2) deficiency. The significance of this association is controversial, though the association of C2 deficiency with certain histocompatibility antigens suggests possible linkage to immune response genes. To our knowledge this is the first report of heterozygous C2 deficiency in association with this combination of autoimmune disorders, and we discuss the aetiological implications.. 3718019 0 26 Heterozygous C2 deficiency SpecificDisease OMIM:217000 3718019 43 53 angioedema SpecificDisease D000799 3718019 55 72 myasthenia gravis SpecificDisease D009157 3718019 78 106 systemic lupus erythematosus SpecificDisease D008180 3718019 135 152 myasthenia gravis SpecificDisease D009157 3718019 154 182 systemic lupus erythematosus SpecificDisease D008180 3718019 188 198 angioedema SpecificDisease D000799 3718019 228 263 complement factor 2 (C2) deficiency SpecificDisease OMIM:217000 3718019 346 359 C2 deficiency SpecificDisease OMIM:217000 3718019 511 524 C2 deficiency SpecificDisease OMIM:217000 3718019 565 585 autoimmune disorders DiseaseClass D001327 2963536|t|Inherited C3 deficiency with recurrent infections and glomerulonephritis. 2963536|a|A 10-year-old Laotian boy had homozygous deficiency of the third component of complement and recurrent bacterial infections beginning at age 5 months. Cellular and humoral immunity were normal, as were polymorphonuclear leukocyte chemotaxis and bactericidal activities. Serum complement-mediated hemolytic, chemotactic, and opsonic activities were deficient. In vitro addition of purified C3 to patient serum restored hemolytic complement to normal levels, and plasma infusion during each of four episodes of pneumonia significantly enhanced serum opsonic activity for as long as 36 hours. A renal biopsy specimen revealed mesangiopathic glomerulonephritis, although significant levels of circulating IgG immune complexes were not detected. These findings further support the association of C3 deficiency with immune-complex disease and suggest that plasma infusion may be an adjunct to antibiotic therapy in the management of severe pyogenic infections in patients with C3 deficiency.. 2963536 10 23 C3 deficiency SpecificDisease OMIM:613779 2963536 54 72 glomerulonephritis SpecificDisease D005921 2963536 115 162 deficiency of the third component of complement SpecificDisease OMIM:613779 2963536 177 197 bacterial infections DiseaseClass D001424 2963536 583 592 pneumonia SpecificDisease D011014 2963536 697 730 mesangiopathic glomerulonephritis SpecificDisease D005921 2963536 865 878 C3 deficiency SpecificDisease OMIM:613779 2963536 884 906 immune-complex disease DiseaseClass D007105 2963536 1008 1027 pyogenic infections DiseaseClass D007239 2963536 1045 1058 C3 deficiency SpecificDisease OMIM:613779 7586656|t|Southern analysis reveals a large deletion at the hypoxanthine phosphoribosyltransferase locus in a patient with Lesch-Nyhan syndrome. 7586656|a|Whole genomic hprt clones were used in Southern analysis to screen the integrity of the hprt gene in a family that includes a patient with HPRT enzyme deficiency causal to Lesch-Nyhan syndrome. A 5 kb DNA sequence deletion was found to have its endpoints in the first and third introns. The probes identified the carrier status of female family members, aided by an RFLP carried by the mothers normal X-chromosome.. 7586656 113 133 Lesch-Nyhan syndrome SpecificDisease D007926 7586656 274 296 HPRT enzyme deficiency SpecificDisease D007926 7586656 307 327 Lesch-Nyhan syndrome SpecificDisease D007926 10426999|t|Association of BRCA1 with the hRad50-hMre11-p95 complex and the DNA damage response. 10426999|a|BRCA1 encodes a tumor suppressor that is mutated in familial breast and ovarian cancers. Here, it is shown that BRCA1 interacts in vitro and in vivo with hRad50, which forms a complex with hMre11 and p95/nibrin. Upon irradiation, BRCA1 was detected in discrete foci in the nucleus, which colocalize with hRad50. Formation of irradiation-induced foci positive for BRCA1, hRad50, hMre11, or p95 was dramatically reduced in HCC/1937 breast cancer cells carrying a homozygous mutation in BRCA1 but was restored by transfection of wild-type BRCA1. Ectopic expression of wild-type, but not mutated, BRCA1 in these cells rendered them less sensitive to the DNA damage agent, methyl methanesulfonate. These data suggest that BRCA1 is important for the cellular responses to DNA damage that are mediated by the hRad50-hMre11-p95 complex.. 10426999 101 106 tumor Modifier D009369 10426999 137 172 familial breast and ovarian cancers CompositeMention D061325 10426999 515 528 breast cancer Modifier D001943 3678494|t|Nebulin and titin expression in Duchenne muscular dystrophy appears normal. 3678494|a|Monoclonal antibodies which recognize different epitopes on either titin or nebulin show normal staining patterns on frozen sections of three muscle biopsies of Duchenne muscular dystrophy (DMD). Gel electrophoresis and immunoblotting performed on two of these muscle biopsies show the normal pattern of titin and nebulin polypeptides. Since the donor of one of these biopsies has a large deletion of the 5-region of the DMD gene, our results argue against the recent proposal that nebulin is the gene mutated in DMD.. 3678494 32 59 Duchenne muscular dystrophy SpecificDisease D020388 3678494 237 264 Duchenne muscular dystrophy SpecificDisease D020388 3678494 266 269 DMD SpecificDisease D020388 3678494 497 500 DMD Modifier D020388 3678494 589 592 DMD SpecificDisease D020388 8281152|t|Myotonic dystrophy kinase is a component of neuromuscular junctions. 8281152|a|The clinical manifestation of myotonic dystrophy (DM) is correlated to the extent of expansion of an unstable [CTG] n DNA motif. Recent studies have demonstrated that this trinucleotide motif forms part of the last, 3 untranslated exon of a gene which potentially encodes multiple protein isoforms of a serine/threonine protein kinase (myotonic dystrophy protein kinase, DM-PK). We report here on the development of antisera against synthetic DM-PK peptide antigens and their use in biochemical and histochemical studies. Immunoreactive DM-kinase protein of 53 kD is present at low levels in skeletal and cardiac muscle extracts of DM patients and normal controls. Immunohistochemical staining revealed that DM-PK is localised prominently at sites of neuromuscular and myotendinous junctions (NMJs and MTJs) of human and rodent skeletal muscles. Furthermore, very low levels of immunoreactive DM-PK protein are present in the sarcoplasm of predominantly type I fibres in various muscles. Strikingly, presence of the protein can also be demonstrated for NMJs of muscular tissues of adult and congenital cases of DM, with no gross changes in structural organisation. Our findings provide a basis for further characterisation of the role of the kinase in protein assembly processes or signal mediation at synaptic sites and ultimately for the understanding of the complex pathophysiology of DM.. 8281152 0 18 Myotonic dystrophy Modifier D009223 8281152 99 117 myotonic dystrophy SpecificDisease D009223 8281152 119 121 DM SpecificDisease D009223 8281152 405 423 myotonic dystrophy Modifier D009223 8281152 701 703 DM Modifier D009223 8281152 1180 1182 DM SpecificDisease D009223 8281152 1457 1459 DM SpecificDisease D009223 7316485|t|New genetic variants of glucose 6-phosphate dehydrogenase (G6PD) in Italy. 7316485|a|Six new variants of human erythrocyte G6PD have been characterized. All of them were found in Italian males and all were associated with enzyme deficiency, but only two with signs of haemolysis. These and other variants reported in the literature, which must thus far be regarded as sporadic, are found to map in parts of Italy where common types of G6PD deficiency are also prevalent.. 7316485 212 229 enzyme deficiency DiseaseClass D008661 7316485 258 268 haemolysis SpecificDisease D006461 7316485 425 440 G6PD deficiency SpecificDisease D005955 10677309|t|ATM-heterozygous germline mutations contribute to breast cancer-susceptibility. 10677309|a|Approximately 0. 5% -1% of the general population has been estimated to be heterozygous for a germline mutation in the ATM gene. Mutations in the ATM gene are responsible for the autosomal recessive disorder ataxia-telangiectasia (A-T) (MIM 208900). The finding that ATM-heterozygotes have an increased relative risk for breast cancer was supported by some studies but not confirmed by others. In view of this discrepancy, we examined the frequency of ATM germline mutations in a selected group of Dutch patients with breast cancer. We have analyzed ATM germline mutations in normal blood lymphocytes, using the protein-truncation test followed by genomic-sequence analysis. A high percentage of ATM germline mutations was demonstrated among patients with sporadic breast cancer. The 82 patients included in this study had developed breast cancer at age < 45 and had survived >/= 5 years (mean 15 years), and in 33 (40%) of the patients a contralateral breast tumor had been diagnosed. Among these patients we identified seven (8. 5%) ATM germline mutations, of which five are distinct. One splice-site mutation (IVS10-6T-- > G) was detected three times in our series. Four heterozygous carriers were patients with bilateral breast cancer. Our results indicate that the mutations identified in this study are " A-T disease-causing " mutations that might be associated with an increased risk of breast cancer in heterozygotes. We conclude that ATM heterozygotes have an approximately ninefold-increased risk of developing a type of breast cancer characterized by frequent bilateral occurrence, early age at onset, and long-term survival. The specific characteristics of our population of patients may explain why such a high frequency was not found in other series. 10677309 50 63 breast cancer Modifier D001943 10677309 259 287 autosomal recessive disorder DiseaseClass D030342 10677309 288 309 ataxia-telangiectasia SpecificDisease D001260 10677309 311 314 A-T SpecificDisease D001260 10677309 401 414 breast cancer SpecificDisease D001943 10677309 598 611 breast cancer SpecificDisease D001943 10677309 836 858 sporadic breast cancer SpecificDisease D001943 10677309 913 926 breast cancer SpecificDisease D001943 10709732|t|Autoimmune lymphoproliferative syndrome (ALPS) in a child from consanguineous parents: a dominant or recessive disease? 10709732|a|Autoimmune lymphoproliferative syndrome (ALPS) is characterized by autoimmune features and lymphoproliferations and is generally caused by defective Fas-mediated apoptosis. This report describes a child with clinical features of ALPS without detectable Fas expression on freshly isolated blood leukocytes. Detection of FAS transcripts via real-time quantitative PCR made a severe transcriptional defect unlikely. Sequencing of the FAS gene revealed a 20-nucleotide duplication in the last exon affecting the cytoplasmic signaling domain. The patient was homozygous for this mutation, whereas the consanguineous parents and the siblings were heterozygous. The patient reported here is a human homologue of the Fas-null mouse, inasmuch as she carries an autosomal homozygous mutation in the FAS gene and she shows the severe and accelerated ALPS phenotype. The heterozygous family members did not have the ALPS phenotype, indicating that the disease-causing FAS mutation in this family is autosomal recessive.. 10709732 0 39 Autoimmune lymphoproliferative syndrome SpecificDisease D056735 10709732 41 45 ALPS SpecificDisease D056735 10709732 120 159 Autoimmune lymphoproliferative syndrome SpecificDisease D056735 10709732 161 165 ALPS SpecificDisease D056735 10709732 349 353 ALPS SpecificDisease D056735 10709732 959 963 ALPS Modifier D056735 10709732 1024 1028 ALPS Modifier D056735 8551426|t|A prevalent mutation for galactosemia among black Americans. 8551426|a|OBJECTIVE To define the mutation causing galactosemia in patients of black American origin who have no galactose-1-phosphate uridyltransferase (GALT) activity in erythrocytes but good clinical outcome. METHODS We discovered a mutation caused by a C-- > T transition at base-pair 1158 of the GALT gene that results in a serine-to-leucine substitution at codon 135 (S135L). We developed a method with which to screen populations for its prevalence. We compared galactose-1-phosphate uridyltransferase among erythrocytes, leukocytes, and transformed lymphoblasts, as well as total body oxidation of D- (13C) -galactose to 13CO2 among three genotypes for GALT (S135L/S135L, Q188R/Q188R, and Normal/Normal). RESULTS We found a 48% prevalence of the S135L mutation among 17 black American patients with classic galactosemia and a 1% prevalence in a population of 50 black Americans without galactosemia. The S135L mutation was not found in 84 white patients with G/G galactosemia nor in 87 white control subjects without galactosemia. We found normal whole body oxidation of D- (13C) -galactose by the patient homozygous for S135L and various degrees of enzyme impairment among different tissues. CONCLUSIONS The S135L mutation in the GALT gene is a prevalent cause of galactosemia among black patients. Because GALT activity varies in different tissues of patients homozygous for S135L, they may have a better clinical outcome than patients who are homozygous for Q188R when both are treated from infancy.. 8551426 25 37 galactosemia SpecificDisease D005693 8551426 103 115 galactosemia SpecificDisease D005693 8551426 861 881 classic galactosemia SpecificDisease D005693 8551426 948 960 galactosemia SpecificDisease D005693 8551426 1025 1037 galactosemia SpecificDisease D005693 8551426 1079 1091 galactosemia SpecificDisease D005693 8551426 1328 1340 galactosemia SpecificDisease D005693 10699184|t|Constitutive and regulated modes of splicing produce six major myotonic dystrophy protein kinase (DMPK) isoforms with distinct properties. 10699184|a|Myotonic dystrophy (DM) is the most prevalent inherited neuromuscular disease in adults. The genetic defect is a CTG triplet repeat expansion in the 3-untranslated region of the myotonic dystrophy protein kinase (DMPK) gene, consisting of 15 exons. Using a transgenic DMPK-overexpressor mouse model, we demonstrate here that the endogenous mouse DMPK gene and the human DMPK transgene produce six major alternatively spliced mRNAs which have almost identical cell type-dependent distribution frequencies and expression patterns. Use of a cryptic 5 splice site in exon 8, which results in absence or presence of 15 nucleotides specifying a VSGGG peptide motif, and/or use of a cryptic 3 splice site in exon 14, which leads to a frameshift in the mRNA reading frame, occur as independent stochastic events in all tissues examined. In contrast, the excision of exons 13/14 that causes a frameshift and creates a C-terminally truncated protein is clearly cell type dependent and occurs predominantly in smooth muscle. We generated all six full-length mouse cDNAs that result from combinations of these three major splicing events and show that their transfection into cells in culture leads to production of four different approximately 74 kDa full-length (heart-, skeletal muscle- or brain-specific) and two C-terminally truncated approximately 68 kDa (smooth muscle-specific) isoforms. Information on DMPK mRNA and protein isoform expression patterns will be useful for recognizing differential effects of (CTG) (n) expansion in DM manifestation.. 10699184 63 81 myotonic dystrophy Modifier D009223 10699184 139 157 Myotonic dystrophy SpecificDisease D009223 10699184 159 161 DM SpecificDisease D009223 10699184 185 216 inherited neuromuscular disease DiseaseClass D009468+D030342 10699184 232 246 genetic defect DiseaseClass D030342 10699184 317 335 myotonic dystrophy Modifier D009223 10699184 1666 1668 DM Modifier D009223 10417286|t|Linkage analysis in a large Brazilian family with van der Woude syndrome suggests the existence of a susceptibility locus for cleft palate at 17p11.2-11.1. 10417286|a|van der Woude syndrome (VWS), which has been mapped to 1q32-41, is characterized by pits and/or sinuses of the lower lip, cleft lip/palate (CL/P), cleft palate (CP), bifid uvula, and hypodontia (H). The expression of VWS, which has incomplete penetrance, is highly variable. Both the occurrence of CL/P and CP within the same genealogy and a recurrence risk < 40% for CP among descendants with VWS have suggested that the development of clefts in this syndrome is influenced by modifying genes at other loci. To test this hypothesis, we have conducted linkage analysis in a large Brazilian kindred with VWS, considering as affected the individuals with CP, regardless of whether it is associated with other clinical signs of VWS. Our results suggest that a gene at 17p11. 2-11 2-11. 1, together with the VWS gene at 1p32-41, enhances the probability of CP in an individual carrying the two at-risk genes. If this hypothesis is confirmed in other VWS pedigrees, it will represent one of the first examples of a gene, mapped through linkage analysis, which modifies the expression of a major gene. 10417286 50 72 van der Woude syndrome SpecificDisease C536528 10417286 126 138 cleft palate SpecificDisease D002972 10417286 156 178 van der Woude syndrome SpecificDisease C536528 10417286 180 183 VWS SpecificDisease C536528 10417286 240 276 pits and/or sinuses of the lower lip CompositeMention D008047 10417286 278 294 cleft lip/palate SpecificDisease D002971|D002972 10417286 296 300 CL/P SpecificDisease D002971|D002972 10417286 303 315 cleft palate SpecificDisease D002972 10417286 317 319 CP SpecificDisease D002972 10417286 322 333 bifid uvula SpecificDisease C531732 10417286 339 349 hypodontia SpecificDisease D000848 10417286 351 352 H SpecificDisease D000848 10417286 373 376 VWS SpecificDisease C536528 10417286 454 458 CL/P SpecificDisease D002971|D002972 10417286 463 465 CP SpecificDisease D002972 10417286 524 526 CP SpecificDisease D002972 10417286 550 553 VWS SpecificDisease C536528 10417286 593 599 clefts DiseaseClass D002971|D002972 10417286 759 762 VWS SpecificDisease C536528 10417286 809 811 CP SpecificDisease D002972 10417286 881 884 VWS SpecificDisease C536528 10417286 960 963 VWS Modifier C536528 10417286 1009 1011 CP SpecificDisease D002972 10417286 1102 1105 VWS Modifier C536528 3393536|t|Diverse point mutations in the human glucose-6-phosphate dehydrogenase gene cause enzyme deficiency and mild or severe hemolytic anemia. 3393536|a|Glucose-6-phosphate dehydrogenase (G6PD; EC 1. 1. 1. 49) deficiency is a common genetic abnormality affecting an estimated 400 million people worldwide. Clinical and biochemical analyses have identified many variants exhibiting a range of phenotypes, which have been well characterized from the hematological point of view. However, until now, their precise molecular basis has remained unknown. We have cloned and sequenced seven mutant G6PD alleles. In the nondeficient polymorphic African variant G6PD A we have found a single point mutation. The other six mutants investigated were all associated with enzyme deficiency. In one of the commonest, G6PD Mediterranean, which is associated with favism among other clinical manifestations, a single amino acid replacement was found (serine----phenylalanine) it must be responsible for the decreased stability and the reduced catalytic efficiency of this enzyme. Single point mutations were also found in G6PD Metaponto (Southern Italy) and in G6PD Ilesha (Nigeria), which are asymptomatic, and in G6PD Chatham, which was observed in an Indian boy with neonatal jaundice. In G6PD " Matera, " which is now known to be the same as G6PD A-, two separate point mutations were found, one of which is the same as in G6PD A. In G6PD Santiago, a de novo mutation (glycine----arginine) is associated with severe chronic hemolytic anemia. The mutations observed show a striking predominance of C----T transitions, with CG doublets involved in four of seven cases. Thus, diverse point mutations may account largely for the phenotypic heterogeneity of G6PD deficiency. 3393536 82 99 enzyme deficiency DiseaseClass D008661 3393536 119 135 hemolytic anemia SpecificDisease D000743 3393536 137 204 Glucose-6-phosphate dehydrogenase (G6PD; EC 1. 1. 1. 49) deficiency SpecificDisease D005955 3393536 217 236 genetic abnormality DiseaseClass D030342 3393536 743 760 enzyme deficiency DiseaseClass D008661 3393536 832 838 favism DiseaseClass D005236 3393536 1239 1256 neonatal jaundice DiseaseClass D007567 3393536 1497 1513 hemolytic anemia SpecificDisease D000743 3393536 1726 1741 G6PD deficiency SpecificDisease D005955 2215607|t|Increased high-density lipoprotein levels caused by a common cholesteryl-ester transfer protein gene mutation. 2215607|a|BACKGROUND AND METHODS. The plasma cholesteryl-ester transfer protein (CETP) catalyzes the transfer of cholesteryl esters from high-density lipoprotein (HDL) to other lipoproteins. We recently described a Japanese family with increased HDL levels and CETP deficiency due to a splicing defect of the CETP gene. To assess the frequency and phenotype of this condition, we screened 11 additional families with high HDL levels by means of a radioimmunoassay for CETP and DNA analysis. RESULTS. We found the same CETP gene mutation in four families from three different regions of Japan. Analysis of restriction-fragment-length polymorphisms of the mutant CETP allele showed that all probands were homozygous for the identical haplotype. Family members homozygous for CETP deficiency (n = 10) had moderate hypercholesterolemia (mean total cholesterol level [+/- SD], 7. 01 +/- 0. 83 mmol per liter), markedly increased levels of HDL cholesterol (4. 24 +/- 1. 01 mmol per liter) and apolipoprotein A-I, and decreased levels of low-density lipoprotein cholesterol (1. 99 +/- 0. 80 mmol per liter) and apolipoprotein B. Members heterozygous for the deficiency (n = 20), whose CETP levels were in the lower part of the normal range, had moderately increased levels of HDL cholesterol and apolipoprotein A-I and an increased ratio of HDL subclass 2 to HDL subclass 3, as compared with unaffected family members (1. 5 +/- 0. 8 vs. 0. 7 +/- 0. 4). CETP deficiency was not found in six unrelated subjects with elevated HDL cholesterol levels who were from different parts of the United States. CONCLUSIONS. CETP deficiency appears to be a frequent cause of increased HDL levels in the population of Japan, possibly because of a founder effect. The results that we observed in heterozygotes suggest that CETP normally plays a part in the regulation of levels of HDL subclass 2. There was no evidence of premature atherosclerosis in the families with CETP deficiency. In fact, the lipoprotein profile of persons with CETP deficiency is potentially antiatherogenic and may be associated with an increased life span. 2215607 362 377 CETP deficiency SpecificDisease OMIM:143470 2215607 874 889 CETP deficiency SpecificDisease OMIM:143470 2215607 912 932 hypercholesterolemia SpecificDisease D006937 2215607 1547 1562 CETP deficiency SpecificDisease OMIM:143470 2215607 1705 1720 CETP deficiency SpecificDisease OMIM:143470 2215607 2000 2025 premature atherosclerosis SpecificDisease D050197 2215607 2047 2062 CETP deficiency SpecificDisease OMIM:143470 2215607 2113 2128 CETP deficiency SpecificDisease OMIM:143470 7523157|t|Hereditary deficiency of the seventh component of complement and recurrent meningococcal infection: investigations of an Irish family using a novel haemolytic screening assay for complement activity and C7 M/N allotyping. 7523157|a|Terminal complement component deficiency predisposes to meningococcal infection and is inherited in an autosomal co-dominant manner. An Irish family is described, in which 2 of 3 brothers had recurrent meningococcal infection. A novel screening assay was used to investigate for terminal complement deficiency and the 2 affected brothers were found to be completely deficient in the seventh component of complement (C7). Enzyme-linked immunosorbent assay for C7 revealed lower than normal levels in the remaining brother and parents. C7 M/N protein polymorphism allotyping, used to investigate the segregation of the C7 deficiency genes, showed that the apparently complement sufficient brother was heterozygous C7 deficient and a carrier of one of the deficiency genes. Complement screening should be carried out in any individual suffering recurrent meningococcal infection or infection with an uncommon meningococcal serogroup. Identification of complement deficient patients allows the implementation of strategies to prevent recurrent infection.. 7523157 11 60 deficiency of the seventh component of complement SpecificDisease OMIM:610102 7523157 75 98 meningococcal infection SpecificDisease D008589 7523157 222 262 Terminal complement component deficiency SpecificDisease D007153 7523157 278 301 meningococcal infection SpecificDisease D008589 7523157 424 447 meningococcal infection SpecificDisease D008589 7523157 501 531 terminal complement deficiency SpecificDisease D007153 7523157 577 636 completely deficient in the seventh component of complement SpecificDisease OMIM:610102 7523157 839 852 C7 deficiency Modifier OMIM:610102 7523157 934 946 C7 deficient SpecificDisease OMIM:610102 7523157 1074 1097 meningococcal infection SpecificDisease D008589 7523157 1128 1141 meningococcal Modifier D008589 7523157 1171 1191 complement deficient Modifier D007153 3162536|t|Mild and severe muscular dystrophy associated with deletions in Xp21 of the human X chromosome. 3162536|a|We have analysed over 300 patients suffering from Duchenne or Becker muscular dystrophy (DMD or BMD). Deletions have been characterised which encompass either the pERT87 (DXS164) locus only, the XJ1. 1 (DXS206) and HIP25 loci only, or all three loci. These loci have been shown to lie within the DMD region covering several hundred kilobases (kb) of DNA. One mildly affected BMD patient possesses a deletion of at least 110 kb including exons of the DMD gene. Other patients with similar exon deletions, or smaller deletions, show the more severe phenotype typical of DMD. We conclude from these studies that the severity of the clinical phenotype cannot be explained on the basis of the size of the deletion. We discuss this in the context of candidate gene sequences. 3162536 16 34 muscular dystrophy DiseaseClass D009136 3162536 146 183 Duchenne or Becker muscular dystrophy CompositeMention D020388|C537666 3162536 185 188 DMD SpecificDisease D020388 3162536 192 195 BMD SpecificDisease C537666 3162536 392 395 DMD Modifier D020388 3162536 471 474 BMD Modifier C537666 3162536 546 549 DMD Modifier D020388 3162536 664 667 DMD SpecificDisease D020388 10631148|t|Meiotic segregation analysis of RB1 alleles in retinoblastoma pedigrees by use of single-sperm typing. 10631148|a|In hereditary retinoblastoma, different epidemiological studies have indicated a preferential paternal transmission of mutant retinoblastoma alleles to offspring, suggesting the occurrence of a meiotic drive. To investigate this mechanism, we analyzed sperm samples from six individuals from five unrelated families affected with hereditary retinoblastoma. Single-sperm typing techniques were performed for each sample by study of two informative short tandem repeats located either in or close to the retinoblastoma gene (RB1). The segregation probability of mutant RB1 alleles in sperm samples was assessed by use of the SPERMSEG program, which includes experimental parameters, recombination fractions between the markers, and segregation parameters. A total of 2, 952 single sperm from the six donors were analyzed. We detected a significant segregation distortion in the data as a whole (P =. 0099) and a significant heterogeneity in the segregation rate across donors (. 0092). Further analysis shows that this result can be explained by segregation distortion in favor of the normal allele in one donor only and that it does not provide evidence of a significant segregation distortion in the other donors. The segregation distortion favoring the mutant RB1 allele does not seem to occur during spermatogenesis, and, thus, meiotic drive may result either from various mechanisms, including a fertilization advantage or a better mobility in sperm bearing a mutant RB1 gene, or from the existence of a defectively imprinted gene located on the human X chromosome. 10631148 47 61 retinoblastoma Modifier D012175 10631148 106 131 hereditary retinoblastoma SpecificDisease D012175 10631148 229 243 retinoblastoma Modifier D012175 10631148 433 458 hereditary retinoblastoma SpecificDisease D012175 10631148 605 619 retinoblastoma Modifier D012175 10861298|t|Functional differences of the PDS gene product are associated with phenotypic variation in patients with Pendred syndrome and non-syndromic hearing loss (DFNB4). 10861298|a|The PDS gene encodes a transmembrane protein, known as pendrin, which functions as a transporter of iodide and chloride. Mutations in this gene are responsible for Pendred syndrome and autosomal recessive non-syndromic hearing loss at the DFNB4 locus on chromosome 7q31. A screen of 20 individuals from the midwestern USA with non-syndromic hearing loss and dilated vestibular aqueducts identified three people (15%) with PDS mutations. To determine whether PDS mutations in individuals with Pendred syndrome differ functionally from PDS mutations in individuals with non-syndromic hearing loss, we compared three common Pendred syndrome allele variants (L236P, T416P and E384G), with three PDS mutations reported only in individuals with non-syndromic hearing loss (V480D, V653A and I490L/G497S). The mutations associated with Pendred syndrome have complete loss of pendrin-induced chloride and iodide transport, while alleles unique to people with DFNB4 are able to transport both iodide and chloride, albeit at a much lower level than wild-type pendrin. We hypothesize that this residual level of anion transport is sufficient to eliminate or postpone the onset of goiter in individuals with DFNB4. We propose a model for pendrin function in the thyroid in which pendrin transports iodide across the apical membrane of the thyrocyte into the colloid space.. 10861298 30 33 PDS Modifier C536648 10861298 105 121 Pendred syndrome SpecificDisease C536648 10861298 126 152 non-syndromic hearing loss SpecificDisease C537845 10861298 154 159 DFNB4 SpecificDisease OMIM:600791 10861298 166 169 PDS Modifier C536648 10861298 326 342 Pendred syndrome SpecificDisease C536648 10861298 347 393 autosomal recessive non-syndromic hearing loss SpecificDisease OMIM:600791 10861298 401 406 DFNB4 Modifier OMIM:600791 10861298 489 515 non-syndromic hearing loss SpecificDisease C537845 10861298 584 587 PDS Modifier C536648 10861298 620 623 PDS Modifier C536648 10861298 654 670 Pendred syndrome SpecificDisease C536648 10861298 696 699 PDS Modifier C536648 10861298 730 756 non-syndromic hearing loss SpecificDisease C537845 10861298 783 799 Pendred syndrome Modifier C536648 10861298 853 856 PDS Modifier C536648 10861298 901 927 non-syndromic hearing loss SpecificDisease C537845 10861298 990 1006 Pendred syndrome SpecificDisease C536648 10861298 1112 1117 DFNB4 SpecificDisease OMIM:600791 10861298 1330 1336 goiter DiseaseClass D006042 10861298 1357 1362 DFNB4 SpecificDisease OMIM:600791 1353340|t|Late-onset metachromatic leukodystrophy: molecular pathology in two siblings. 1353340|a|We report on a new allele at the arylsulfatase A (ARSA) locus causing late-onset metachromatic leukodystrophy (MLD). In that allele arginine84, a residue that is highly conserved in the arylsulfatase gene family, is replaced by glutamine. In contrast to alleles that cause early-onset MLD, the arginine84 to glutamine substitution is associated with some residual ARSA activity. A comparison of genotypes, ARSA activities, and clinical data on 4 individuals carrying the allele of 81 patients with MLD examined, further validates the concept that different degrees of residual ARSA activity are the basis of phenotypical variation in MLD.. 1353340 11 39 metachromatic leukodystrophy SpecificDisease D007966 1353340 159 187 metachromatic leukodystrophy SpecificDisease D007966 1353340 189 192 MLD SpecificDisease D007966 1353340 363 366 MLD SpecificDisease D007966 1353340 576 579 MLD Modifier D007966 1353340 712 715 MLD SpecificDisease D007966 10200300|t|Defective CD95/APO-1/Fas signal complex formation in the human autoimmune lymphoproliferative syndrome, type Ia. 10200300|a|Heterozygous mutations in the CD95 (APO-1/Fas) receptor occur in most individuals with autoimmune lymphoproliferative syndrome (ALPS) and dominantly interfere with apoptosis by an unknown mechanism. We show that local or global alterations in the structure of the cytoplasmic death domain from nine independent ALPS CD95 death-domain mutations result in a failure to bind the FADD/MORT1 signaling protein. Despite heterozygosity for the abnormal allele, lymphocytes from ALPS patients showed markedly decreased FADD association and a loss of caspase recruitment and activation after CD95 crosslinking. These data suggest that intracytoplasmic CD95 mutations in ALPS impair apoptosis chiefly by disrupting death-domain interactions with the signaling protein FADD/MORT1.. 10200300 63 111 autoimmune lymphoproliferative syndrome, type Ia SpecificDisease D056735 10200300 200 239 autoimmune lymphoproliferative syndrome SpecificDisease D056735 10200300 241 245 ALPS SpecificDisease D056735 10200300 424 428 ALPS Modifier D056735 10200300 584 588 ALPS Modifier D056735 10200300 774 778 ALPS SpecificDisease D056735 10408771|t|Classical galactosemia and mutations at the galactose-1-phosphate uridyl transferase (GALT) gene. 10408771|a|Classical galactosemia is caused by a deficiency in activity of the enzyme galactose-1-phosphate uridyl transferase (GALT), which, in turn, is caused by mutations at the GALT gene. The disorder exhibits considerable allelic heterogeneity and, at the end of 1998, more than 150 different base changes were recorded in 24 different populations and ethnic groups in 15 countries worldwide. The mutations most frequently cited are Q188R, K285N, S135L, and N314D. Q188R is the most common mutation in European populations or in those predominantly of European descent. Overall, it accounts for 60-70% of mutant chromosomes, but there are significant differences in its relative frequency in individual populations. Individuals homoallelic for Q188R tend to have a severe phenotype and this is in keeping with the virtually complete loss of enzyme activity observed in in vitro expression systems. Globally, K285N is rarer, but in many European populations it can be found on 25-40% of mutant chromosomes. It is invariably associated with a severe phenotype. S135L is found almost exclusively in African Americans. In vitro expression results are discrepant, but some individuals carrying S135L appear to exhibit GALT activity in some tissues. Duarte 1 (or Los Angeles) and Duarte 2 (or Duarte) variants carry the same amino acid substitution, N314D, even though D1 is associated with increased erythrocyte GALT activity and D2 with reduced activity. N314D is in linkage disequilibrium with other base changes that differ on the D1 and D2 alleles. N314D does not impair GALT activity in in vitro expression systems. However, there are differences in the abundance of GALT protein in lymphoblastoid cells lines from D2 and D1 individuals. It is unclear whether the specific molecular changes that distinguish the D1 and D2 alleles account for the different activities. The considerable genetic heterogeneity documented to date undoubtedly contributes to the phenotypic heterogeneity that is observed in galactosemia. The additional effects of nonallelic variation and other constitutional factors on phenotypic variability remain to be elucidated.. 10408771 0 22 Classical galactosemia SpecificDisease D005693 10408771 98 120 Classical galactosemia SpecificDisease D005693 10408771 2094 2106 galactosemia SpecificDisease D005693 10636421|t|X-linked retinoschisis with point mutations in the XLRS1 gene. 10636421|a|BACKGROUND X-linked retinoschisis (XLRS) is a relatively rare vitreoretinal dystrophy that causes visual loss in young men. Recently, a gene responsible for this disease, designated XLRS1, was identified, and several deleterious gene mutations were reported. OBJECTIVE To analyze Japanese patients clinically diagnosed as having XLRS formutational changes in the XLRS1 gene. METHODS Ten patients with XLRS underwent full ophthalmologic examination, including slitlamp biomicroscopy and dilated funduscopy. Genomic DNA was isolated from leukocytes, and all exons of the XLRS1 gene were amplified by polymerase chain reaction and analyzed using a direct sequencing method. RESULTS Point mutations in the XLRS1 gene were identified in all 10 patients. The mutations were identical in each of 2 pairs of brothers. Six of the point mutations represented missense mutations, 1 was a nonsense mutation, and 1 was a frameshift mutation. Five of the mutations are newly reported herein. CONCLUSIONS The discovery of new point mutations in this study increases the available information regarding the spectrum of genetic abnormalities and clinical manifestations of XLRS. However, the limited data failed to reveal a correlation between mutation and disease phenotype. CLINICAL RELEVANCE Identification of mutations in the XLRS1 gene and expanded information on clinical manifestations will facilitate early diagnosis, appropriate early therapy, and genetic counseling regarding the prognosis of XLRS.. 10636421 0 22 X-linked retinoschisis SpecificDisease D041441 10636421 75 97 X-linked retinoschisis SpecificDisease D041441 10636421 99 103 XLRS SpecificDisease D041441 10636421 126 149 vitreoretinal dystrophy DiseaseClass D058499 10636421 162 173 visual loss SpecificDisease C531604 10636421 394 398 XLRS Modifier D041441 10636421 467 471 XLRS SpecificDisease D041441 10636421 1171 1192 genetic abnormalities DiseaseClass D030342 10636421 1224 1228 XLRS SpecificDisease D041441 10636421 1555 1559 XLRS SpecificDisease D041441 10429004|t|Relationship among genotype, biochemical phenotype, and cognitive performance in females with phenylalanine hydroxylase deficiency: report from the Maternal Phenylketonuria Collaborative Study. 10429004|a|OBJECTIVE To examine the relationship of phenylalanine hydroxylase (PAH) genotypes to biochemical phenotype and cognitive development in maternal phenylketonuria (PKU). METHODOLOGY PAH gene mutations were examined in 222 hyperphenylalaninemic females enrolled in the Maternal PKU Collaborative Study (MPKUCS). A total of 84 different mutations were detected, and complete genotype was obtained in 199 individuals. Based on previous knowledge about mutation-phenotype associations, 78 of the mutations could be assigned to one of four classes of severity (severe PKU, moderate PKU, mild PKU, and mild hyperphenylalaninemia [MHP]). Then, 189 MPKUCS subjects were grouped according to the various combinations of mutation classifications. The sample sizes were large enough for statistical testing in four groups with at least one mutation that completely abolishes enzyme activity. These patients are considered functionally hemizygous. RESULTS The biochemical phenotype predicted from the genotype in functionally hemizygous patients was related significantly to the assigned phenylalanine level. Cognitive performance (IQ) was also significantly related to genotype. The IQ of PAH-deficient mothers with a severe PKU mutation in combination with a MHP mutation or a mild PKU mutation was 99 and 96, respectively, whereas the IQ of PKU mothers with two severe PKU mutations or with one severe and one moderate PKU mutation was 83 and 84, respectively. Of the patients with PKU, 92% had been treated during childhood. Those who were untreated or treated late had lower than average IQ scores for their group of mutation combinations. Females with moderate or mild PKU who were treated early and treated for > 6 years showed IQ scores 10 points above average for their group. CONCLUSIONS The reproductive outcome in maternal phenylketonuria is dependent on prenatal metabolic control and postnatal environmental circumstances. Both factors depend on the intellectual resources of the mother with PKU. The significant relationship among genotype, biochemical phenotype, and cognitive performance observed in the present study is of importance for the development of an optimal strategy for future treatment of females with PKU who plan pregnancy.. 10429004 94 130 phenylalanine hydroxylase deficiency SpecificDisease OMIM:261600 10429004 148 172 Maternal Phenylketonuria Modifier D017042 10429004 332 356 maternal phenylketonuria SpecificDisease D017042 10429004 358 361 PKU SpecificDisease D010661 10429004 417 438 hyperphenylalaninemic Modifier D010661 10429004 463 475 Maternal PKU Modifier D017042 10429004 758 761 PKU SpecificDisease D010661 10429004 772 775 PKU SpecificDisease D010661 10429004 782 785 PKU SpecificDisease D010661 10429004 791 817 mild hyperphenylalaninemia SpecificDisease D010661 10429004 819 822 MHP SpecificDisease D010661 10429004 1374 1387 PAH-deficient Modifier OMIM:261600 10429004 1410 1413 PKU Modifier D010661 10429004 1445 1448 MHP Modifier D010661 10429004 1468 1471 PKU Modifier D010661 10429004 1528 1531 PKU Modifier D010661 10429004 1556 1559 PKU Modifier D010661 10429004 1606 1609 PKU Modifier D010661 10429004 1669 1672 PKU SpecificDisease D010661 10429004 1859 1862 PKU SpecificDisease D010661 10429004 2011 2035 maternal phenylketonuria SpecificDisease D017042 10429004 2191 2194 PKU SpecificDisease D010661 10429004 2417 2420 PKU SpecificDisease D010661 1380672|t|Uncoupling of hypomyelination and glial cell death by a mutation in the proteolipid protein gene. 1380672|a|Proteolipid protein (PLP; M (r) 30, 000) is a highly conserved major polytopic membrane protein in myelin but its cellular function remains obscure. Neurological mutant mice can often provide model systems for human genetic disorders . Mutations of the X-chromosome-linked PLP gene are lethal, identified first in the jimpy mouse and subsequently in patients with Pelizaeus-Merzbacher disease. The unexplained phenotype of these mutations includes degeneration and premature cell death of oligodendrocytes with associated hypomyelination. Here we show that a new mouse mutant rumpshaker is defined by the amino-acid substitution Ile-to-Thr at residue 186 in a membrane-embedded domain of PLP. Surprisingly, rumpshaker mice, although myelin-deficient, have normal longevity and a full complement of morphologically normal oligodendrocytes. Hypomyelination can thus be genetically separated from the PLP-dependent oligodendrocyte degeneration. We suggest that PLP has a vital function in glial cell development, distinct from its later role in myelin assembly, and that this dichotomy of action may explain the clinical spectrum of Pelizaeus-Merzbacher disease.. 1380672 14 29 hypomyelination SpecificDisease D003711 1380672 34 50 glial cell death SpecificDisease D004194 1380672 314 331 genetic disorders DiseaseClass D030342 1380672 462 490 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 1380672 546 603 degeneration and premature cell death of oligodendrocytes CompositeMention D056784 1380672 620 635 hypomyelination SpecificDisease D003711 1380672 831 847 myelin-deficient SpecificDisease D003711 1380672 937 952 Hypomyelination SpecificDisease D003711 1380672 1010 1038 oligodendrocyte degeneration SpecificDisease D056784 1380672 1228 1256 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 10594001|t|Mutational analysis of the HGO gene in Finnish alkaptonuria patients. 10594001|a|Alkaptonuria (AKU), the prototypic inborn error of metabolism, has recently been shown to be caused by loss of function mutations in the homogentisate-1, 2-dioxygenase gene (HGO). So far 17 mutations have been characterised in AKU patients of different ethnic origin. We describe three novel mutations (R58fs, R330S, and H371R) and one common AKU mutation (M368V), detected by mutational and polymorphism analysis of the HGO gene in five Finnish AKU pedigrees. The three novel AKU mutations are most likely specific for the Finnish population and have originated recently.. 10594001 47 59 alkaptonuria Modifier D000474 10594001 70 82 Alkaptonuria SpecificDisease D000474 10594001 84 87 AKU SpecificDisease D000474 10594001 105 131 inborn error of metabolism DiseaseClass D008661 10594001 297 300 AKU Modifier D000474 10594001 413 416 AKU Modifier D000474 10594001 516 519 AKU Modifier D000474 10594001 547 550 AKU Modifier D000474 8441467|t|Putative X-linked adrenoleukodystrophy gene shares unexpected homology with ABC transporters. 8441467|a|Adrenoleukodystrophy (ALD) is an X-linked disease affecting 1/20, 000 males either as cerebral ALD in childhood or as adrenomyeloneuropathy (AMN) in adults. Childhood ALD is the more severe form, with onset of neurological symptoms between 5-12 years of age. Central nervous system demyelination progresses rapidly and death occurs within a few years. AMN is a milder form of the disease with onset at 15-30 years of age and a more progressive course. Adrenal insufficiency (Addisons disease) may remain the only clinical manifestation of ALD. The principal biochemical abnormality of ALD is the accumulation of very-long-chain fatty acids (VLCFA) because of impaired beta-oxidation in peroxisomes. The normal oxidation of VLCFA-CoA in patients fibroblasts suggested that the gene coding for the VLCFA-CoA synthetase could be a candidate gene for ALD. Here we use positional cloning to identify a gene partially deleted in 6 of 85 independent patients with ALD. In familial cases, the deletions segregated with the disease. An identical deletion was detected in two brothers presenting with different clinical ALD phenotypes. Candidate exons were identified by computer analysis of genomic sequences and used to isolate complementary DNAs by exon connection and screening of cDNA libraries. The deduced protein sequence shows significant sequence identity to a peroxisomal membrane protein of M (r) 70K that is involved in peroxisome biogenesis and belongs to the ATP-binding cassette superfamily of transporters.. 8441467 9 38 X-linked adrenoleukodystrophy Modifier D000326 8441467 94 114 Adrenoleukodystrophy SpecificDisease D000326 8441467 116 119 ALD SpecificDisease D000326 8441467 127 143 X-linked disease DiseaseClass D040181 8441467 180 192 cerebral ALD SpecificDisease D000326 8441467 212 233 adrenomyeloneuropathy SpecificDisease D000326 8441467 235 238 AMN SpecificDisease D000326 8441467 261 264 ALD SpecificDisease D000326 8441467 353 389 Central nervous system demyelination DiseaseClass D003711 8441467 446 449 AMN SpecificDisease D000326 8441467 546 567 Adrenal insufficiency DiseaseClass D000309 8441467 569 585 Addisons disease SpecificDisease D000224 8441467 633 636 ALD SpecificDisease D000326 8441467 679 682 ALD SpecificDisease D000326 8441467 941 944 ALD SpecificDisease D000326 8441467 1051 1054 ALD SpecificDisease D000326 8441467 1204 1207 ALD Modifier D000326 10618304|t|Cardiac Na(+) channel dysfunction in Brugada syndrome is aggravated by beta(1)-subunit. 10618304|a|BACKGROUND Mutations in the gene encoding the human cardiac Na (+) channel alpha-subunit (hH1) are responsible for chromosome 3-linked congenital long-QT syndrome (LQT3) and idiopathic ventricular fibrillation (IVF). An auxiliary beta (1) -subunit, widely expressed in excitable tissues, shifts the voltage dependence of steady-state inactivation toward more negative potentials and restores normal gating kinetics of brain and skeletal muscle Na (+) channels expressed in Xenopus oocytes but has little if any functional effect on the cardiac isoform. Here, we characterize the altered effects of a human beta (1) -subunit (hbeta (1)) on the heterologously expressed hH1 mutation (T1620M) previously associated with IVF. METHODS AND RESULTS When expressed alone in Xenopus oocytes, T1620M exhibited no persistent currents, in contrast to the LQT3 mutant channels, but the midpoint of steady-state inactivation (V (1/2)) was significantly shifted toward more positive potentials than for wild-type hH1. Coexpression of hbeta (1) did not significantly alter current decay or recovery from inactivation of wild-type hH1; however, it further shifted the V (1/2) and accelerated the recovery from inactivation of T1620M. Oocyte macropatch analysis revealed that the activation kinetics of T1620M were normal. CONCLUSIONS It is suggested that coexpression of hbeta (1) exposes a more severe functional defect that results in a greater overlap in the relationship between channel inactivation and activation (window current) in T1620M, which is proposed to be a potential pathophysiological mechanism of IVF in vivo. One possible explanation for our finding is an altered alpha-/beta (1) -subunit association in the mutant.. 10618304 37 53 Brugada syndrome SpecificDisease D053840 10618304 224 251 congenital long-QT syndrome SpecificDisease D008133 10618304 253 257 LQT3 SpecificDisease C537034 10618304 263 298 idiopathic ventricular fibrillation SpecificDisease C537182 10618304 300 303 IVF SpecificDisease C537182 10618304 806 809 IVF SpecificDisease C537182 10618304 933 937 LQT3 Modifier C537034 10618304 1689 1692 IVF SpecificDisease C537182 10369876|t|Autosomal recessive familial neurohypophyseal diabetes insipidus with continued secretion of mutant weakly active vasopressin. 10369876|a|Familial neurohypophyseal diabetes insipidus is an autosomal dominant disorder characterized by post-natal development of arginine vasopressin (AVP) deficiency due to mutations in the AVP gene. All published mutations affect the signal peptide or the neurophysin-II carrier protein and are presumed to interfere with processing of the preprohormone, leading to neuronal damage. We studied an unusual Palestinian family consisting of asymptomatic first cousin parents and three children affected with neurohypophyseal diabetes insipidus, suggesting autosomal recessive inheritance. All three affected children were homozygous and the parents heterozygous for a single novel mutation (C301- > T) in exon 1, replacing Pro7 of mature AVP with Leu (Leu-AVP). Leu-AVP was a weak agonist with approximately 30-fold reduced binding to the human V2 receptor. Measured by radioimmunoassay with a synthetic Leu-AVP standard, serum Leu-AVP levels were elevated in all three children and further increased during water deprivation to as high as 30 times normal. The youngest child (2 years old) was only mildly affected but had Leu-AVP levels similar to her severely affected 8-year-old brother, suggesting that unknown mechanisms may partially compensate for a deficiency of active AVP in very young children.. 10369876 0 64 Autosomal recessive familial neurohypophyseal diabetes insipidus SpecificDisease OMIM:125700 10369876 127 171 Familial neurohypophyseal diabetes insipidus SpecificDisease OMIM:125700 10369876 178 205 autosomal dominant disorder DiseaseClass D030342 10369876 249 286 arginine vasopressin (AVP) deficiency SpecificDisease OMIM:125700 10369876 488 503 neuronal damage DiseaseClass D009410 10369876 627 662 neurohypophyseal diabetes insipidus SpecificDisease D020790 10369876 1376 1400 deficiency of active AVP DiseaseClass OMIM:125700 2575483|t|Autosomal dominant aniridia linked to the chromosome 11p13 markers catalase and D11S151 in a large Dutch family. 2575483|a|In a large pedigree with autosomal dominant aniridia, we found close linkage between the aniridia locus AN2 and the markers catalase (CAT) (zeta = 7. 27 at theta = 0. 00) and D11S151 (zeta = 3. 86 at theta = 0. 10) flanking the AN2 locus on 11p13. Positive lod scores were also obtained for the 11p13----11p14 markers D11S16 and FSHB with the linkage group CAT/AN2/D11S151. We conclude that the autosomal dominant aniridia in this family is due to a mutation at the AN2 locus on 11p13. We have excluded linkage (zeta less than -2 at theta less than 0. 18) between the aniridia and the chromosome 2p25 marker D2S1 (linked to ACP1). 2575483 19 27 aniridia SpecificDisease D015783 2575483 157 165 aniridia SpecificDisease D015783 2575483 202 210 aniridia Modifier D015783 2575483 527 535 aniridia SpecificDisease D015783 2575483 681 689 aniridia SpecificDisease D015783 10607954|t|Knobloch syndrome involving midline scalp defect of the frontal region. 10607954|a|We report on a 4-year-old boy with Knobloch syndrome. He has vitreoretinal degeneration, high myopia, cataract, telecanthus, hypertelorism, and a high-arched palate. He also has a defect of the anterior midline scalp with involvement of the frontal bone as documented by a computed tomography (CT) scan. The brain was normal on CT scan and magnetic resonance imaging. We present a review of the 23 published cases with this syndrome. Our patient illustrates the importance of investigating for underlying ocular and central nervous system pathology whenever midline scalp defects are present.. 10607954 0 17 Knobloch syndrome SpecificDisease C537209 10607954 28 70 midline scalp defect of the frontal region DiseaseClass C538225 10607954 107 124 Knobloch syndrome SpecificDisease C537209 10607954 133 159 vitreoretinal degeneration SpecificDisease D012162 10607954 161 172 high myopia SpecificDisease D009216 10607954 174 182 cataract SpecificDisease D002386 10607954 184 195 telecanthus SpecificDisease D030342 10607954 197 210 hypertelorism SpecificDisease D006972 10607954 218 236 high-arched palate SpecificDisease D007569 10607954 252 288 defect of the anterior midline scalp DiseaseClass C538225 10607954 630 651 midline scalp defects DiseaseClass C538225 10602116|t|Pendred syndrome: phenotypic variability in two families carrying the same PDS missense mutation. 10602116|a|Pendred syndrome comprises congenital sensorineural hearing loss, thyroid goiter, and positive perchlorate discharge test. Recently, this autosomal recessive disorder was shown to be caused by mutations in the PDS gene, which encodes an anion transporter called pendrin. Molecular analysis of the PDS gene was performed in two consanguineous large families from Southern Tunisia comprising a total of 23 individuals affected with profound congenital deafness; the same missense mutation, L445W, was identified in all affected individuals. A widened vestibular aqueduct was found in all patients who underwent computed tomography (CT) scan exploration of the inner ear. In contrast, goiter was present in only 11 affected individuals, who interestingly had a normal result of the perchlorate discharge test whenever performed. The present results question the sensitivity of the perchlorate test for the diagnosis of Pendred syndrome and support the use of a molecular analysis of the PDS gene in the assessment of individuals with severe to profound congenital hearing loss associated with inner ear morphological anomaly even in the absence of a thyroid goiter.. 10602116 0 16 Pendred syndrome SpecificDisease C536648 10602116 75 78 PDS Modifier C536648 10602116 98 114 Pendred syndrome SpecificDisease C536648 10602116 125 162 congenital sensorineural hearing loss SpecificDisease D006319 10602116 164 178 thyroid goiter SpecificDisease D006042 10602116 236 264 autosomal recessive disorder DiseaseClass D030342 10602116 308 311 PDS Modifier C536648 10602116 395 398 PDS Modifier C536648 10602116 537 556 congenital deafness SpecificDisease D003638 10602116 639 666 widened vestibular aqueduct SpecificDisease OMIM:600791 10602116 780 786 goiter SpecificDisease D006042 10602116 1014 1030 Pendred syndrome SpecificDisease C536648 10602116 1082 1085 PDS Modifier C536648 10602116 1148 1171 congenital hearing loss SpecificDisease D003638 10602116 1188 1219 inner ear morphological anomaly SpecificDisease D007759 10602116 1245 1259 thyroid goiter SpecificDisease D006042 10470088|t|The type of somatic mutation at APC in familial adenomatous polyposis is determined by the site of the germline mutation: a new facet to Knudson's 'two-hit' hypothesis. 10470088|a|APC is often cited as a prime example of a tumor suppressor gene. Truncating germline and somatic mutations (or, infrequently, allelic loss) occur in tumors in FAP (familial adenomatous polyposis). Most sporadic colorectal cancers also have two APC mutations. Clues from attenuated polyposis, missense germline variants with mild disease and the somatic mutation cluster region (codons 1, 250-1, 450) indicate, however, that APC mutations might not result in simple loss of protein function. We have found that FAP patients with germline APC mutations within a small region (codons 1, 194-1, 392 at most) mainly show allelic loss in their colorectal adenomas, in contrast to other FAP patients, whose second hits tend to occur by truncating mutations in the mutation cluster region. Our results indicate that different APC mutations provide cells with different selective advantages, with mutations close to codon 1, 300 providing the greatest advantage. Allelic loss is selected strongly in cells with one mutation near codon 1, 300. A different germline-somatic APC mutation association exists in FAP desmoids. APC is not, therefore, a classical tumor suppressor. Our findings also indicate a new mechanism for disease severity if a broader spectrum of mutations is selected in tumors, the somatic mutation rate is effectively higher and more tumors grow.. 10470088 39 69 familial adenomatous polyposis SpecificDisease D011125 10470088 212 217 tumor Modifier D009369 10470088 319 325 tumors DiseaseClass D009369 10470088 329 332 FAP SpecificDisease D011125 10470088 334 364 familial adenomatous polyposis SpecificDisease D011125 10470088 372 399 sporadic colorectal cancers SpecificDisease D015179 10470088 414 417 APC Modifier D011125 10470088 440 460 attenuated polyposis Modifier C538265 10470088 594 597 APC Modifier D011125 10470088 680 683 FAP Modifier D011125 10470088 707 710 APC Modifier D011125 10470088 808 827 colorectal adenomas SpecificDisease D000236 10470088 850 853 FAP Modifier D011125 10470088 988 991 APC Modifier D011125 10470088 1233 1236 APC Modifier D011125 10470088 1268 1280 FAP desmoids SpecificDisease D018222 10470088 1317 1322 tumor Modifier D009369 10470088 1450 1456 tumors DiseaseClass D009369 10470088 1515 1521 tumors DiseaseClass D009369 7769092|t|Purification of human very-long-chain acyl-coenzyme A dehydrogenase and characterization of its deficiency in seven patients. 7769092|a|Mitochondrial very-long-chain acyl-coenzyme A dehydrogenase (VLCAD) was purified from human liver. The molecular masses of the native enzyme and the subunit were estimated to be 154 and 70 kD, respectively. The enzyme was found to catalyze the major part of mitochondrial palmitoylcoenzyme A dehydrogenation in liver, heart, skeletal muscle, and skin fibroblasts (89-97, 86-99, 96-99, and 78-87%, respectively). Skin fibroblasts from 26 patients suspected of having a disorder of mitochondrial beta-oxidation were analyzed for VLCAD protein using immunoblotting, and 7 of them contained undetectable or trace levels of the enzyme. The seven deficient fibroblast lines were characterized by measuring acyl-coenzyme A dehydrogenation activities, overall palmitic acid oxidation, and VLCAD protein synthesis using pulse-chase, further confirming the diagnosis of VLCAD deficiency. These results suggested the heterogenous nature of the mutations causing the deficiency in the seven patients. Clinically, all patients with VLCAD deficiency exhibited cardiac disease. At least four of them presented with hypertrophic cardiomyopathy. This frequency (> 57%) was much higher than that observed in patients with other disorders of mitochondrial long-chain fatty acid oxidation that may be accompanied by cardiac disease in infants.. 7769092 986 1002 VLCAD deficiency SpecificDisease C536353 7769092 1145 1161 VLCAD deficiency SpecificDisease C536353 7769092 1172 1187 cardiac disease DiseaseClass D006331 7769092 1226 1253 hypertrophic cardiomyopathy SpecificDisease D002312 7769092 1422 1437 cardiac disease DiseaseClass D006331 1282899|t|A germ line mutation within the coding sequence for the putative 5-phosphoribosyl-1-pyrophosphate binding site of hypoxanthine-guanine phosphoribosyltransferase (HPRT) in a Lesch-Nyhan patient: missense mutations within a functionally important region probably cause disease. 1282899|a|Lesch-Nyhan syndrome caused by a complete deficiency of hypoxanthine guanine phosphoribosyltransferase (HPRT) is the result of a heterogeneous group of germ line mutations. Identification of each mutant gene provides valuable information as to the type of mutation that occurs spontaneously. We report here a newly identified HPRT mutation in a Japanese patient with Lesch-Nyhan syndrome. This gene, designated HPRT Tokyo, had a single nucleotide change from G to A, as identified by sequencing cDNA amplified by the polymerase chain reaction. Allele specific oligonucleotide hybridization analysis using amplified genomic DNA showed that the mutant gene was transmitted from the maternal germ line. This mutation would lead to an amino acid substitution of Asp for Gly at the amino acid position 140 located within the putative 5-phosphoribosyl-1-pyrophosphate (PRPP) binding region. Missense mutations in human HPRT deficient patients thus far reported tend to accumulate in this functionally active region. However, a comparison of the data suggested that both missense and synonymous mutations can occur at any coding sequence of the human germ line HPRT gene, but that a limited percentage of all the missense mutations cause disease. The probability that a mutation will cause disease tends to be higher when the missense mutation is within a functionally important sequence.. 1282899 173 184 Lesch-Nyhan Modifier D007926 1282899 276 296 Lesch-Nyhan syndrome SpecificDisease D007926 1282899 309 378 complete deficiency of hypoxanthine guanine phosphoribosyltransferase SpecificDisease D007926 1282899 643 663 Lesch-Nyhan syndrome SpecificDisease D007926 1282899 1189 1203 HPRT deficient Modifier D007926 1301201|t|In vitro and in vivo correlations for I65T and M1V mutations at the phenylalanine hydroxylase locus. 1301201|a|Mutations at the phenylalanine hydroxylase (PAH) locus are the major cause of hyperphenylalaninemia. We have previously described four mutations (M1V, IVS12nt1, R408W, and S349P) at the PAH locus in French Canadians with ancestry in eastern Quebec. Here we report (1) identification of another mutation, on a haplotype 9 chromosome, which converts codon 65 from isoleucine (ATT) to threonine (ACT), (2) expression analysis of the I65T mutation in COS cells demonstrating 75% loss of both immunoreactive protein and enzyme activity, and (3) expression analysis of the most prevalent PKU allele (M1V) in eastern Quebec, showing nondetectable levels of PAH protein and activity, a finding compatible with a mutation in the translation initiation codon. Homozygosity for M1V and codominant inheritance of I65T/R408W were both associated with classical phenylketonuria.. 1301201 179 200 hyperphenylalaninemia DiseaseClass D010661 1301201 683 686 PKU Modifier D010661 1301201 939 964 classical phenylketonuria SpecificDisease D010661 10557317|t|Experimental hemochromatosis due to MHC class I HFE deficiency: immune status and iron metabolism. 10557317|a|The puzzling linkage between genetic hemochromatosis and histocompatibility loci became even more so when the gene involved, HFE, was identified. Indeed, within the well defined, mainly peptide-binding, MHC class I family of molecules, HFE seems to perform an unusual yet essential function. As yet, our understanding of HFE function in iron homeostasis is only partial; an even more open question is its possible role in the immune system. To advance on both of these avenues, we report the deletion of HFE alpha1 and alpha2 putative ligand binding domains in vivo. HFE-deficient animals were analyzed for a comprehensive set of metabolic and immune parameters. Faithfully mimicking human hemochromatosis, mice homozygous for this deletion develop iron overload, characterized by a higher plasma iron content and a raised transferrin saturation as well as an elevated hepatic iron load. The primary defect could, indeed, be traced to an augmented duodenal iron absorption. In parallel, measurement of the gut mucosal iron content as well as iron regulatory proteins allows a more informed evaluation of various hypotheses regarding the precise role of HFE in iron homeostasis. Finally, an extensive phenotyping of primary and secondary lymphoid organs including the gut provides no compelling evidence for an obvious immune-linked function for HFE.. 10557317 13 28 hemochromatosis SpecificDisease D006432 10557317 48 62 HFE deficiency SpecificDisease D006432 10557317 136 151 hemochromatosis SpecificDisease D006432 10557317 666 679 HFE-deficient Modifier D006432 10557317 789 804 hemochromatosis SpecificDisease D006432 144081|t|[Genetic heterogeneity of G6PD deficiency: mutant alleles of G6PD in the Shekii district of Azerbaijan] 144081|a|Examination on G6PD deficiency in 349 patients of Shekii district hospital (Azerbaijan) revealed 16 hemi-, 4 homo- and 9 heterozygotic carriers of the defect. Gd- frequency, calculated from the data obtained (7. 7%), may be compared to neighbouring regions frequencies (6-30%). Carriers of G6PD deficiency are residents of 11 villages located in Alasani-Aphtalan valley, highly endemic with malaria in the past; nearly all marriages are endogamic. Physico-chemical and kinetic study of 10 mutant forms of G6PD, according to WHO program, led to identification of 5 variants of the II class (Shekii, Bideiz, Shirin-Bulakh, Okhut I and Zakataly) and 2 variants of the III class (Okhut II and Martinique-like). Resemblance of the majority of variants in electrophoretic mobility and the level of erythrocyte enzyme activity permit to suggest the existence of a common parental mutant G6PD allele distributed in this area. 144081 26 41 G6PD deficiency SpecificDisease D005955 144081 119 134 G6PD deficiency SpecificDisease D005955 144081 394 409 G6PD deficiency SpecificDisease D005955 144081 495 502 malaria SpecificDisease D008288 6453040|t|[Gd- allele distribution patterns in Azerbaijan. III. The identification of mutant forms of glucose-6-phosphate dehydrogenase] 6453040|a|In 28 families with G6PD deficiency living in 3 settlements of Shekii district of Azerbaijan 11 G6PD variants of II and III classes differing by kinetic properties were identified according WHO program. 9 of them are characterized with the same electrophoretic mobility. Comparison of G6PD spectra in two subpopulations and in a mixed group permits to make a conclusion about existence of common and rare G6PD alleles in examined population. They distribute by gene drift supported by natural selection. Among 7 samples of G6PD with normal and increased activity two new variants of IV class -- Nukha and Bash-Kungut -- were found.. 6453040 147 162 G6PD deficiency SpecificDisease D005955 10878391|t|Determination of carrier status for the Wiskott-Aldrich syndrome by flow cytometric analysis of Wiskott-Aldrich syndrome protein expression in peripheral blood mononuclear cells. 10878391|a|The Wiskott-Aldrich syndrome (WAS) is caused by defects in the WAS protein (WASP) gene on the X chromosome. Previous study disclosed that flow cytometric analysis of intracellular WASP expression (FCM-WASP analysis) in lymphocytes was useful for the diagnosis of WAS patients. Lymphocytes from all WAS patients showed WASPdim instead of WASPbright. Here we report that FCM-WASP analysis in monocytes could be a useful tool for the WAS carrier diagnosis. Monocytes from all nine WAS carriers showed varied population of WASPdim together with WASPbright. None of control individuals possessed the WASPdim population. In contrast, lymphocytes from all the carriers except two lacked the WASPdim population. The difference of the WASPdim population in monocytes and lymphocytes observed in WAS carriers suggests that WASP plays a more critical role in the development of lymphocytes than in that of monocytes. The present studies suggest that a skewed X-chromosomal inactivation pattern observed in WAS carrier peripheral blood cells is not fixed at the hemopoietic stem cell level but progresses after the lineage commitment.. 10878391 40 64 Wiskott-Aldrich syndrome SpecificDisease D014923 10878391 96 120 Wiskott-Aldrich syndrome Modifier D014923 10878391 183 207 Wiskott-Aldrich syndrome SpecificDisease D014923 10878391 209 212 WAS SpecificDisease D014923 10878391 242 245 WAS Modifier D014923 10878391 442 445 WAS Modifier D014923 10878391 477 480 WAS Modifier D014923 10878391 610 613 WAS Modifier D014923 10878391 657 660 WAS Modifier D014923 10878391 965 968 WAS Modifier D014923 10878391 1174 1177 WAS Modifier D014923 2161209|t|Linkage of DNA markers at Xq28 to adrenoleukodystrophy and adrenomyeloneuropathy present within the same family. 2161209|a|We present a large kindred that contained patients with either adrenoleukodystrophy (ALD) or adrenomyeloneuropathy (AMN). The pedigree clearly supported the X-linked mode of inheritance of the nonneonatal form of ALD/AMN. Analysis with DNA markers at Xq28 suggested segregation of both ALD and AMN with an identical haplotype. This indicated that nonneonatal ALD and AMN are caused by a mutation in the same gene at Xq28. It showed, furthermore, that phenotypic differences between ALD and AMN are not necessarily the consequence of allelic heterogeneity due to different mutations within the same gene. The maximal lod score for linkage of the ALD/AMN gene and the multiallelic anonymous DNA marker at DXS52 was 3. 0 at a recombination fraction of 0. 00. This made a prenatal or presymptomatic diagnosis and heterozygote detection by DNA analysis with this marker reliable. 2161209 34 54 adrenoleukodystrophy SpecificDisease D000326 2161209 59 80 adrenomyeloneuropathy SpecificDisease D000326 2161209 176 196 adrenoleukodystrophy SpecificDisease D000326 2161209 198 201 ALD SpecificDisease D000326 2161209 206 227 adrenomyeloneuropathy SpecificDisease D000326 2161209 229 232 AMN SpecificDisease D000326 2161209 326 329 ALD SpecificDisease D000326 2161209 330 333 AMN SpecificDisease D000326 2161209 399 402 ALD SpecificDisease D000326 2161209 407 410 AMN SpecificDisease D000326 2161209 472 475 ALD SpecificDisease D000326 2161209 480 483 AMN SpecificDisease D000326 2161209 595 598 ALD SpecificDisease D000326 2161209 603 606 AMN SpecificDisease D000326 2161209 758 761 ALD Modifier D000326 2161209 762 765 AMN Modifier D000326 8326491|t|Further investigation of the HEXA gene intron 9 donor splice site mutation frequently found in non-Jewish Tay-Sachs disease patients from the British Isles. 8326491|a|In a previous study we found that a Tay-Sachs disease (TSD) causing mutation in the intron 9 donor splice site of the HEXA gene occurs at high frequency in non-Jewish patients and carriers from the British Isles. It was found more frequently in subjects of Irish, Scottish, and Welsh origin compared with English origin (63% and 31% respectively). We have now tested, in a blind study, 26 American TSD carriers and 28 non-carriers who have British ancestry for the intron 9 splice site mutation. Six of the carriers and none of the controls were positive for the mutation. All six had Irish ancestry, compared with nine of the 20 other (intron 9 mutation negative) TSD carriers (p < 0. 05). These results confirm the previously found high frequency of the intron 9 mutation in non-Jewish TSD families of British Isles, particularly Irish, origin, and reinforce the need to screen such families for this mutation. 8326491 106 123 Tay-Sachs disease Modifier D013661 8326491 193 210 Tay-Sachs disease SpecificDisease D013661 8326491 212 215 TSD SpecificDisease D013661 8326491 555 558 TSD Modifier D013661 8326491 822 825 TSD Modifier D013661 8326491 945 948 TSD Modifier D013661 7959767|t|The murine homologues of the Huntington disease gene (Hdh) and the alpha-adducin gene (Add1) map to mouse chromosome 5 within a region of conserved synteny with human chromosome 4p16.3. 7959767|a|Huntington disease (HD) is a severe autosomal dominant neurodegenerative disorder associated with a novel gene (IT15). Recently, we reported the cloning of Hdh, the murine homologue of IT15. Here, using an interspecific backcross, we have mapped both Hdh and the mouse homologue of human alpha-adducin (Add1), a membrane-associated cytoskeletal protein gene. Both of these genes map in the same position on mouse chromosome 5 in a region associated with ancestral chromosomal rearrangements and show no recombination with D5H4S43, D5H4S115, and D5H4S62, the murine homologues of D4S43, D4S115, and D4S62, respectively. Further mapping studies of humans, mice, and other mammalian species should reveal the nature of the rearrangements affecting this chromosomal segment during mammalian evolution.. 7959767 29 47 Huntington disease Modifier D006816 7959767 186 204 Huntington disease SpecificDisease D006816 7959767 206 208 HD SpecificDisease D006816 7959767 222 267 autosomal dominant neurodegenerative disorder DiseaseClass D019636 2562820|t|Linkage analysis of the apolipoprotein C2 gene and myotonic dystrophy on human chromosome 19 reveals linkage disequilibrium in a French-Canadian population. 2562820|a|The gene for human apolipoprotein C2 (APOC2), situated on the proximal long arm of chromosome 19, is closely linked to the gene for the most common form of adult muscular dystrophy, myotonic dystrophy (DM). Six APOC2 RFLPs (TaqI, BglI, BanI, BamHI, NcoI, and AvaII) have been identified to date. We have conducted a comprehensive DM linkage study utilizing all six RFLPs and involving 50 families and 372 individuals. The most informative RFLPs are, in descending order, NcoI (lod = 6. 64, theta = 0. 05), BglI (lod = 6. 12, theta = 0. 05), AvaII (lod = 6. 02, theta = 0. 03), BanI (lod = 5. 76, theta = 0. 04), TaqI (lod = 4. 29, theta = 0. 06), and BamHI (lod = 1. 75, theta = 0. 01). A substantial increase in the lod scores over those seen with the individual RFLPs was obtained when the linkage of the entire APOC2 haplotype (composed of the six RFLPs) was studied (lod = 17. 87, theta = 0. 04). We have observed significant inter-APOC2 RFLP linkage disequilibrium. Consequently, the three most informative RFLPs have been found to be BanI, TaqI, and either BglI, AvaII, or NcoI polymorphisms. We also demonstrate linkage disequilibrium between DM and APOC2 in our French-Canadian population (standardized disequilibrium constant phi =. 22, chi 2 = 5. 12, df = 1, P less than 0. 04). This represents the first evidence of linkage disequilibrium between APOC2 and the DM locus. 2562820 51 69 myotonic dystrophy SpecificDisease D009223 2562820 319 337 muscular dystrophy DiseaseClass D009136 2562820 339 357 myotonic dystrophy SpecificDisease D009223 2562820 359 361 DM SpecificDisease D009223 2562820 487 489 DM Modifier D009223 2562820 1529 1531 DM Modifier D009223 10094552|t|A retrospective anonymous pilot study in screening newborns for HFE mutations in Scandinavian populations. 10094552|a|We have retrospectively analyzed 837 random anonymized dried blood spot (DBS) samples from neonatal screening programs in Scandinavia for mutations in HFE, the candidate gene for hemochromatosis. We have found C282Y allele frequencies of 2. 3% (+ 2. 0%) (-1. 3%) in Greenland, 4. 5% +/-1. 9% in Iceland, 5. 1% +/-2. 3% in the Faeroe Islands, and 8. 2% +/-2. 7% in Denmark. The high prevalence of HFE mutations in Denmark suggests that population screening for the C282Y mutation could be highly advantageous in terms of preventive health care. Long-term follow-up evaluation of C282Y homozygotes and H63D/C282Y compound heterozygotes will give an indication of the penetrance of the mutations. 10094552 286 301 hemochromatosis SpecificDisease D006432 10484981|t|Hereditary TP53 codon 292 and somatic P16INK4A codon 94 mutations in a Li-Fraumeni syndrome family. 10484981|a|Li-Fraumeni syndrome is an autosomal dominant disorder that is characterized by various types of cancer in childhood and adult cases. Although hereditary TP53 mutation is very rare in different human cancers, it has been frequently reported in Li-Fraumeni syndrome. On the other hand, hereditary mutations of TP57KIP2, P15INK4B, and P16INK4A, which affect the cell cycle similar to TP53, were observed in some types of cancer. In a Turkish family with the diagnosis of Li-Fraumeni syndrome, we analyzed the mutation pattern of TP53, P57KIP2, P15INK4B, and P16INK4A in the peripheral blood, and loss of heterozygosity (homo/hemizygous deletion) pattern of TP53 and P15INK4B/P16INK4A in two tumor tissues. The propositus had a seminoma, his daughter a medulloblastoma, and one of his healthy cousins, a TP53 codon 292 missense point mutation (AAA-- > ATA; Lys-- > Ile) in the peripheral blood cells. Tumor tissue obtained from the propositus with the seminoma revealed loss of heterozygosity in the TP53 gene. In the analyses of tumor tissues from the propositus and his daughter, a P16INK4A codon 94 missense point mutation (GCG-- > GAG; Ala-- > Glu) was observed with the hereditary TP53 mutation. P16INK4A codon 94 mutation observed in our family is a novel mutation in Li-Fraumeni syndrome. No other gene alteration in TP53, P57KIP2, P15INK4B, and P16INK4A was observed. Existence of the P16INK4A mutation and the hereditary TP53 mutation with or without loss of heterozygosity in the TP53 gene (seminoma/medulloblastoma) may be evidence for a common mechanism involved in tumorogenesis. The gene alterations in TP53 and P16INK4A genes may be used as tumor markers in our family.. 10484981 71 91 Li-Fraumeni syndrome Modifier D016864 10484981 100 120 Li-Fraumeni syndrome SpecificDisease D016864 10484981 127 154 autosomal dominant disorder DiseaseClass D030342 10484981 197 203 cancer DiseaseClass D009369 10484981 300 307 cancers DiseaseClass D009369 10484981 344 364 Li-Fraumeni syndrome SpecificDisease D016864 10484981 519 525 cancer DiseaseClass D009369 10484981 569 589 Li-Fraumeni syndrome SpecificDisease D016864 10484981 789 794 tumor Modifier D009369 10484981 825 833 seminoma SpecificDisease D018239 10484981 850 865 medulloblastoma SpecificDisease D008527 10484981 1049 1057 seminoma SpecificDisease D018239 10484981 1127 1132 tumor Modifier D009369 10484981 1371 1391 Li-Fraumeni syndrome SpecificDisease D016864 10484981 1598 1606 seminoma SpecificDisease D018239 10484981 1607 1622 medulloblastoma SpecificDisease D008527 10484981 1675 1688 tumorogenesis DiseaseClass D002471 10484981 1753 1758 tumor Modifier D009369 2884570|t|An amino-acid substitution involved in phenylketonuria is in linkage disequilibrium with DNA haplotype 2. 2884570|a|Phenylketonuria (PKU) is an autosomal recessive human genetic disorder caused by a deficiency of hepatic phenylalanine hydroxylase (PAH, phenylalanine 4-monooxygenase, EC 1. 14. 16. 1). PKU is a common inborn error of amino-acid metabolism in caucasian populations and approximately 1 in 50 individuals are carriers of a PKU allele. To define the molecular basis of PKU, we characterized twelve restriction fragment-length polymorphism (RFLP) haplotypes of the PAH locus in the northern European population and observed that 90% of the PKU alleles in this population are confined to four common RFLP haplotypes. We have recently reported a splicing mutation in the PAH gene that is associated with RFLP haplotype 3 which is present at about 40% of mutant alleles. We now report the molecular lesion associated with the RFLP haplotype 2 mutant allele. This defect is caused by a C-to-T transition in exon 12 resulting in an amino-acid substitution (Arg to Trp) at residue 408 of PAH. Direct hybridization analysis of the point mutation using a specific oligonucleotide probe demonstrated that this mutation is also in linkage disequilibrium with RFLP haplotype 2 alleles that make up about 20% of mutant PAH genes 2884570 39 54 phenylketonuria SpecificDisease D010661 2884570 106 121 Phenylketonuria SpecificDisease D010661 2884570 123 126 PKU SpecificDisease D010661 2884570 134 176 autosomal recessive human genetic disorder DiseaseClass D030342 2884570 189 236 deficiency of hepatic phenylalanine hydroxylase SpecificDisease OMIM:261600 2884570 292 295 PKU SpecificDisease D010661 2884570 308 345 inborn error of amino-acid metabolism DiseaseClass D000592 2884570 427 430 PKU Modifier D010661 2884570 472 475 PKU SpecificDisease D010661 2884570 642 645 PKU Modifier D010661 2884570 888 904 molecular lesion DiseaseClass D030342 8258524|t|The effects of dystrophin gene mutations on the ERG in mice and humans. 8258524|a|PURPOSE. The authors earlier findings of a negative electroretinogram (ERG) in a boy with Duchenne muscular dystrophy (DMD) led them to investigate dystrophin gene deletions and ERGs in five boys with DMD. The authors wanted to determined whether there were similar ERG findings in an animal model for DMD, the mdx mouse. METHODS. Ganzfeld ERGs were recorded in five boys with DMD after a complete ophthalmic examination. The dystrophin gene was analyzed by Southern blot hybridization. ERGs were recorded in anesthetized mdx and control mice with a modified Grass photostimulator (Grass Instrument Company, Quincy, MA). RESULTS. Ophthalmic examinations in all five boys had normal findings, yet an abnormal negative ERG was recorded for each subject. The subjects gene deletions were variable, ranging from large deletions to no detectable deletions. The ERGs of the mdx mice were normal and did not differ significantly from those of the control mice. CONCLUSIONS. The authors believe the unique ERG recorded for the human subjects is a manifestation of DMD associated with defects at the dystrophin gene locus and represents a new clinical entity. The ERG of the mdx mouse may be spared for several reasons, including milder effects of the mouse gene defect, differences in muscle and retinal gene product, or species differences in the biochemical role of dystrophin. The ERG shows promise of becoming a noninvasive diagnostic tool for DMD and its milder allelic forms.. 8258524 162 189 Duchenne muscular dystrophy SpecificDisease D020388 8258524 191 194 DMD SpecificDisease D020388 8258524 273 276 DMD SpecificDisease D020388 8258524 374 377 DMD SpecificDisease D020388 8258524 449 452 DMD SpecificDisease D020388 8258524 1128 1131 DMD SpecificDisease D020388 8258524 1512 1515 DMD SpecificDisease D020388 1324223|t|Eight novel inactivating germ line mutations at the APC gene identified by denaturing gradient gel electrophoresis. 1324223|a|Familial adenomatous polyposis (FAP) is a dominantly inherited condition predisposing to colorectal cancer. The recent isolation of the responsible gene (adenomatous polyposis coli or APC) has facilitated the search for germ line mutations in affected individuals. Previous authors have used the RNase protection assay and the single-strand conformation polymorphisms procedure to screen for mutations. In this study we used denaturing gradient gel electrophoresis (DGGE). DGGE analysis of 10 APC exons (4, 5, 7, 8, 9, 10, 12, 13, 14, and part of 15) in 33 unrelated Dutch FAP patients has led to the identification of eight novel germ line mutations resulting in stop codons or frameshifts. The results reported here indicate that (1) familial adenomatous polyposis is caused by an extremely heterogeneous spectrum of point mutations; (2) all the mutations found in this study are chain terminating; and (3) DGGE represents a rapid and sensitive technique for the detection of mutations in the unusually large APC gene. An extension of the DGGE analysis to the entire coding region in a sufficient number of clinically well-characterized, unrelated patients will facilitate the establishment of genotype-phenotype correlations. On the other hand, the occurrence of an extremely heterogeneous spectrum of mutations spread throughout the entire length of the large APC gene among the FAP patients indicates that this approach may not be useful as a rapid presymptomatic diagnostic procedure in a routine laboratory. Nevertheless, the above DGGE approach has incidentally led to the identification of a common polymorphism in exon 13. Such intragenic polymorphisms offer a practical approach to a more rapid procedure for presymptomatic diagnosis of FAP by linkage analysis in informative families.. 1324223 52 55 APC Modifier D011125 1324223 116 146 Familial adenomatous polyposis SpecificDisease D011125 1324223 148 151 FAP SpecificDisease D011125 1324223 205 222 colorectal cancer SpecificDisease D015179 1324223 270 296 adenomatous polyposis coli SpecificDisease D011125 1324223 300 303 APC SpecificDisease D011125 1324223 609 612 APC Modifier D011125 1324223 689 692 FAP Modifier D011125 1324223 852 882 familial adenomatous polyposis SpecificDisease D011125 1324223 1127 1130 APC Modifier D011125 1324223 1480 1483 APC Modifier D011125 1324223 1499 1502 FAP Modifier D011125 1324223 1864 1867 FAP SpecificDisease D011125 10533068|t|Novel mutations in XLRS1 causing retinoschisis, including first evidence of putative leader sequence change. 10533068|a|Juvenile retinoschisis is an X-linked recessive disease caused by mutations in the XLRS1 gene. We screened 31 new unrelated patients and families for XLRS1 mutations in addition to previously reported mutations for 60 of our families (Retinoschisis Consortium, Hum Mol Genet 1998; 7 1185-1192). Twenty-three different mutations including 12 novel ones were identified in 28 patients. Mutations identified in this study include 19 missense mutations, two nonsense mutations, one intragenic deletion, four microdeletions, one insertion, and one intronic sequence substitution that is likely to result in a splice site defect. Two novel mutations, c. 38T-- > C (L13P) and c. 667T-- > C (C223R), respectively, present the first genetic evidence for the functional significance of the putative leader peptide sequence and for the functional significance at the carboxyl terminal of the XLRS1 protein beyond the discoidin domain. Mutations in 25 of the families were localized to exons 4-6, emphasizing the critical functional significance of the discoidin domain of the XLRS1 protein 10533068 33 46 retinoschisis SpecificDisease D041441 10533068 109 131 Juvenile retinoschisis SpecificDisease D041441 10533068 138 164 X-linked recessive disease DiseaseClass D040181 10533068 344 357 Retinoschisis Modifier D041441 6585184|t|Clinical use of DNA markers linked to the gene for Duchenne muscular dystrophy. 6585184|a|Seventy families with Duchenne muscular dystrophy (DMD) known to the Institute of Child Health fall into three categories with respect to potential linkage analysis with the X chromosome DNA markers RC8 and L1. 28 that bridge the DMD gene. Families in which there is at least one obligatory female heterozygote (n = 13). Here prediction and exclusion of DMD gene transmission may be possible, the accuracy being dependent on the closeness of the linkage of the DNA marker (s) to the DMD gene; an illustrative case is reported. Families in which there is a single affected boy, who also has one or more healthy brothers (n = 26). Given an informative restriction fragment length polymorphism (RFLP), the probability that the boy represents a new mutation can be reassessed; it is also possible to exclude the DMD gene in a sister. Families with a single affected boy with no brother (n = 30). Here exclusion of the DMD gene in a sister may be possible. Only in one family was there no possibility of useful linkage analysis. The linkage analysis required is described, and the need to check DMD families for informative RFLPs is stressed. 6585184 51 78 Duchenne muscular dystrophy SpecificDisease D020388 6585184 102 129 Duchenne muscular dystrophy SpecificDisease D020388 6585184 131 134 DMD SpecificDisease D020388 6585184 310 313 DMD Modifier D020388 6585184 434 437 DMD Modifier D020388 6585184 563 566 DMD Modifier D020388 6585184 888 891 DMD Modifier D020388 6585184 994 997 DMD Modifier D020388 6585184 1170 1173 DMD Modifier D020388 8471773|t|Molecular characterization of glucose-6-phosphate dehydrogenase (G6PD) deficiency in patients of Chinese descent and identification of new base substitutions in the human G6PD gene. 8471773|a|The underlying DNA changes associated with glucose-6-phosphate dehydrogenase (G6PD) -deficient Asians have not been extensively investigated. To fill this gap, we sequenced the G6PD gene of 43 G6PD-deficient Chinese whose G6PD was well characterized biochemically. DNA samples were obtained from peripheral blood of these individuals for sequencing using a direct polymerase chain reaction (PCR) sequencing procedure. From these 43 samples, we have identified five different types of nucleotide substitutions in the G6PD gene at cDNA 1388 from G to A (Arg to His); at cDNA 1376 from G to T (Arg to Leu); at cDNA 1024 from C to T (Leu to Phe); at cDNA 392 from G to T (Gly to Val); at cDNA 95 from A to G (His to Arg). These five nucleotide substitutions account for over 83% of our 43 G6PD-deficient samples and these substitutions have not been reported in non-Asians. The substitutions found at cDNA 392 and cDNA 1024 are new findings. The substitutions at cDNA 1376 and 1388 account for over 50% of the 43 samples examined indicating a high prevalence of these two alleles among G6PD-deficient Chinese. Our findings add support to the notion that diverse point mutations may account largely for much of the phenotypic heterogeneity of G6PD deficiency.. 8471773 30 81 glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 8471773 225 276 glucose-6-phosphate dehydrogenase (G6PD) -deficient Modifier D005955 8471773 375 389 G6PD-deficient Modifier D005955 8471773 968 982 G6PD-deficient Modifier D005955 8471773 1265 1279 G6PD-deficient Modifier D005955 8471773 1421 1436 G6PD deficiency SpecificDisease D005955 8314592|t|Norrie disease gene: characterization of deletions and possible function. 8314592|a|Positional cloning experiments have resulted recently in the isolation of a candidate gene for Norrie disease (pseudoglioma; NDP), a severe X-linked neurodevelopmental disorder. Here we report the isolation and analysis of human genomic DNA clones encompassing the NDP gene. The gene spans 28 kb and consists of 3 exons, the first of which is entirely contained within the 5 untranslated region. Detailed analysis of genomic deletions in Norrie patients shows that they are heterogeneous, both in size and in position. By PCR analysis, we found that expression of the NDP gene was not confined to the eye or to the brain. An extensive DNA and protein sequence comparison between the human NDP gene and related genes from the database revealed homology with cysteine-rich protein-binding domains of immediate--early genes implicated in the regulation of cell proliferation. We propose that NDP is a molecule related in function to these genes and may be involved in a pathway that regulates neural cell differentiation and proliferation.. 8314592 0 14 Norrie disease Modifier C537849 8314592 169 183 Norrie disease SpecificDisease C537849 8314592 185 197 pseudoglioma SpecificDisease C537849 8314592 199 202 NDP SpecificDisease C537849 8314592 214 250 X-linked neurodevelopmental disorder DiseaseClass D038901 8314592 339 342 NDP Modifier C537849 8314592 512 518 Norrie Modifier C537849 8314592 642 645 NDP Modifier C537849 8314592 763 766 NDP Modifier C537849 122435|t|Prenatal diagnosis of Wolman disease. 122435|a|Two pregnancies at risk for Wolman disease were monitored by assay and electrophoresis of acid lipase in cultured amniotic-fluid cells. Cells from patient 1 had 5% of control levels of acid lipase, using 14C-triolein as substrate; however, when artificial substrates (esters of 4-methylumbelliferone and p-nitrophenol) were used to measure acid lipase, these cells had 30% of control levels. Electrophoresis of cell extracts revealed the absence of the A form of acid lipase, consistent with the diagnosis of Wolman disease. Analysis of fetal tissues following prostaglandin termination of this pregnancy confirmed the diagnosis. Assay of fetal-skin fibroblasts with 14C-triolein, as well as with artificial substrates, showed marked deficiency of acid lipase activity. Electrophoresis of fetal-tissue extracts also demonstrated the absence of the A form of acid lipase. Amniotic-fluid cells from patient 2 showed normal levels of acid lipase with all substrates tested; the electrophoretic pattern of acid lipase was normal. The results suggest that the prenatal diagnosis of Wolman disease be made using the radioassay of acid lipase and/or electrophoresis.. 122435 22 36 Wolman disease SpecificDisease D015223 122435 66 80 Wolman disease SpecificDisease D015223 122435 547 561 Wolman disease SpecificDisease D015223 122435 1115 1129 Wolman disease SpecificDisease D015223 7795652|t|Somatic mutations in the BRCA1 gene in sporadic ovarian tumours. 7795652|a|The BRCA1 gene on chromosome 17q21 is responsible for an autosomal dominant syndrome of increased susceptibility to breast and ovarian cancer but no somatic mutations in tumours have yet been described. To study the potential role of BRCA1 in sporadic carcinogenesis, we analysed the genomic DNA of tumour and normal fractions of 47 ovarian cancers for mutations in BRCA1 using the single-strand conformation polymorphism technique. We now describe somatic mutations in the DNA of four tumours which also had loss of heterozygosity (LOH) at a BRCA1 intragenic marker. Our data support a tumour suppressor mechanism for BRCA1; somatic mutations and LOH may result in inactivation of BRCA1 in at least a small number of ovarian cancers.. 7795652 39 63 sporadic ovarian tumours SpecificDisease D010051 7795652 122 149 autosomal dominant syndrome DiseaseClass D030342 7795652 181 206 breast and ovarian cancer CompositeMention D061325 7795652 235 242 tumours DiseaseClass D009369 7795652 364 370 tumour DiseaseClass D009369 7795652 398 413 ovarian cancers SpecificDisease D010051 7795652 551 558 tumours DiseaseClass D009369 7795652 652 658 tumour Modifier D009369 7795652 783 798 ovarian cancers SpecificDisease D010051 2180286|t|Skewed X inactivation in a female MZ twin results in Duchenne muscular dystrophy. 2180286|a|One of female MZ twins presented with muscular dystrophy. Physical examination, creatine phosphokinase levels, and muscle biopsy were consistent with Duchenne muscular dystrophy (DMD). However, because of her sex she was diagnosed as having limb-girdle muscular dystrophy. With cDNA probes to the DMD gene, a gene deletion was detected in the twins and their mother. The de novo mutation which arose in the mother was shown by novel junction fragments generated by HindIII, PstI, or TaqI when probed with cDNA8. Additional evidence of a large gene deletion was given by novel SfiI junction fragments detected by probes p20, J-Bir, and J-66 on pulsed-field gel electrophoresis (PFGE). Immunoblot analysis of muscle from the affected twin showed dystrophin of normal size but of reduced amount. Immunofluorescent visualization of dystrophin revealed foci of dystrophin-positive fibers adjacent to foci of dystrophin-negative fibers. These data indicate that the affected twin is a manifesting carrier of an abnormal DMD gene, her myopathy being a direct result of underexpression of dystrophin. Cytogenetic analysis revealed normal karyotypes, eliminating the possibility of a translocation affecting DMD gene function. Both linkage analysis and DNA fingerprint analysis revealed that each twin has two different X chromosomes, eliminating the possibility of uniparental disomy as a mechanism for DMD expression. On the basis of methylation differences of the paternal and maternal X chromosomes in these MZ twins, we propose uneven lyonization (X chromosome inactivation) as the underlying mechanism for disease expression in the affected female.. 2180286 53 80 Duchenne muscular dystrophy SpecificDisease D020388 2180286 120 138 muscular dystrophy DiseaseClass D009136 2180286 232 259 Duchenne muscular dystrophy SpecificDisease D020388 2180286 261 264 DMD SpecificDisease D020388 2180286 323 353 limb-girdle muscular dystrophy SpecificDisease D049288 2180286 379 382 DMD Modifier D020388 2180286 1096 1099 DMD Modifier D020388 2180286 1110 1118 myopathy DiseaseClass D009135 2180286 1281 1284 DMD Modifier D020388 2180286 1439 1457 uniparental disomy SpecificDisease D024182 2180286 1477 1480 DMD Modifier D020388 1316718|t|Coincident Kaposi sarcoma and T-cell lymphoma in a patient with the Wiskott-Aldrich syndrome. 1316718|a|A 24 year old male with a history of eczema, recurrent mild infections, and thrombocytopenia consistent with the Wiskott-Aldrich syndrome (WAS) presented with a mediastinal mass, generalized lymphadenopathy, splenomegaly, and severe thrombocytopenia. Studies of immune function including immunoglobulin levels and T-cell subsets were normal. Furthermore, his T lymphocytes proliferated normally in response to phytohemagglutinin, concanavalin A, and the combination of neuraminidase/galactose oxidase. However, their proliferative responses to anti-CD43 antibody and periodate were diminished, consistent with the clinical diagnosis of WAS. An initial inguinal lymph node biopsy surprisingly revealed Kaposi sarcoma. However, following splenectomy to increase the platelet count, biopsy of the mediastinal mass revealed T-cell large cell lymphoma. Studies of biopsied tissue for the presence of Epstein-Barr virus and cytomegalovirus were negative, as were studies of blood, including the polymerase chain reaction, for the presence of the human immunodeficiency virus (HIV). This is the first report of Kaposi sarcoma arising in a patient with a congenital immunodeficiency syndrome. Although Kaposi sarcoma can arise in the face of the severe immunosuppression that follows allograft transplantation and in patients infected with HIV, we postulate that longevity in the face of mild immunosuppression was the major factor in the development of Kaposi sarcoma in this patient.. 1316718 11 25 Kaposi sarcoma SpecificDisease D012514 1316718 30 45 T-cell lymphoma SpecificDisease D016399 1316718 68 92 Wiskott-Aldrich syndrome SpecificDisease D014923 1316718 131 137 eczema DiseaseClass D004485 1316718 170 186 thrombocytopenia SpecificDisease D013921 1316718 207 231 Wiskott-Aldrich syndrome SpecificDisease D014923 1316718 233 236 WAS SpecificDisease D014923 1316718 285 300 lymphadenopathy SpecificDisease D008206 1316718 302 314 splenomegaly SpecificDisease D013163 1316718 327 343 thrombocytopenia SpecificDisease D013921 1316718 730 733 WAS SpecificDisease D014923 1316718 795 809 Kaposi sarcoma SpecificDisease D012514 1316718 914 940 T-cell large cell lymphoma SpecificDisease D016399 1316718 989 1007 Epstein-Barr virus SpecificDisease D020031 1316718 1012 1027 cytomegalovirus SpecificDisease D003586 1316718 1134 1162 human immunodeficiency virus SpecificDisease D015658 1316718 1164 1167 HIV SpecificDisease D015658 1316718 1198 1212 Kaposi sarcoma SpecificDisease D012514 1316718 1241 1277 congenital immunodeficiency syndrome DiseaseClass D007153 1316718 1288 1302 Kaposi sarcoma SpecificDisease D012514 1316718 1426 1429 HIV SpecificDisease D015658 1316718 1540 1554 Kaposi sarcoma SpecificDisease D012514 7964884|t|Treatment of cerebrotendinous xanthomatosis: effects of chenodeoxycholic acid, pravastatin, and combined use. 7964884|a|Treatments by oral administration of chenodeoxycholic acid (CDCA) alone, 3-hydroxy-3-methylglutaryl (HMG) CoA reductase inhibitor (pravastatin) alone, and combination of the two drugs were attempted for 7 patients with cerebrotendinous xanthomatosis (CTX). CDCA treatment at a dose of 300 mg/day reduced serum cholestanol (67. 3% reduction), lathosterol (50. 8%), campesterol (61. 7%) and sitosterol (12. 7%). However, the sera of the patients changed to be " atherogenic "; total cholesterol, triglyceride and low-density lipoprotein (LDL) -cholesterol were increased, while high-density lipoprotein (HDL) -cholesterol was decreased. Contrarily, pravastatin at a dose of 10 mg/day improved the sera of the patients to be markedly " anti-atherogenic ", but the reductions of cholestanol (30. 4%), lathosterol (44. 0%), campesterol (22. 9%) and sitosterol (9. 6%) were inadequate. Combined treatment with CDCA and pravastatin showed good overlapping of the effects of each drug alone. The sera of the patients were apparently more " anti-atherogenic " than those after CDCA treatment. Serum cholestanol concentration was still 2. 7 times higher than in controls, but the serum lathosterol level was within the normal range, indicating that the enhancement of overall cholesterol synthesis in the patients was sufficiently suppressed. Plant sterol levels were also within the normal range. The combination of CDCA and pravastatin was a good treatment for CTX, based on the improvement of serum lipoprotein metabolism, the suppression of cholesterol synthesis, and reductions of cholestanol and plant sterol levels. In all of 7 patients, the progression of disease was arrested, but dramatic effects on clinical manifestations, xanthoma, and electrophysiological findings could not be found after the treatment of these drugs 7964884 13 43 cerebrotendinous xanthomatosis SpecificDisease D019294 7964884 329 359 cerebrotendinous xanthomatosis SpecificDisease D019294 7964884 361 364 CTX SpecificDisease D019294 7964884 1563 1566 CTX SpecificDisease D019294 7964884 1835 1843 xanthoma DiseaseClass D014973 102474|t|Combined genetic deficiency of C6 and C7 in man. 102474|a|By routine screening of sera, a subject was discovered who showed a sub-total deficiency of C6 and C7. No clinical disease was associated with this deficiency which was transmitted through the subjects family as a single genetic characteristic, the C6 deficiency being associated with a silent allele at the structural locus. The propositus was found to have low quantities of an abnormal C6 which was both antigenically deficient and smaller in size than normal C6 (110, 000 daltons compared with 140, 000 daltons) and small quantities of apparently normal C7. It is concluded that the most likely explanation for this defect is that the subject has a structural mutation in his C6 gene which produces hyopsynthesis not only of C6 but also of the closely linked gene for C7. These findings suggest the possibility that C6 and C7 may function as a single genetic unit and that the primary transcript copied from the genome includes information for both proteins.. 102474 0 40 Combined genetic deficiency of C6 and C7 CompositeMention OMIM:610102|OMIM:612446 102474 117 150 sub-total deficiency of C6 and C7 CompositeMention OMIM:610102|OMIM:612446 102474 298 311 C6 deficiency SpecificDisease OMIM:612446 2651669|t|Partial deletion 8q without Langer-Giedion syndrome: a recognisable syndrome. 2651669|a|We report two de novo cases of del (8) (pter----q24. 1 ) with breakpoints involving the distal part of band 8q24. 1 1. The clinical features were similar and there were no obvious stigmata of Langer-Giedion syndrome (LGS). There are three other cases reported with a deletion of chromosome 8 at approximately the same breakpoint, one without LGS and some similarities to our cases, the other two with LGS. Our findings would support the observation that the critical segment for the assignment of LGS is proximal to or involves the proximal part of 8q24. 1, but a review of published reports suggests that the aetiology of LGS may be a more complex issue 2651669 28 51 Langer-Giedion syndrome SpecificDisease D015826 2651669 270 293 Langer-Giedion syndrome SpecificDisease D015826 2651669 295 298 LGS SpecificDisease D015826 2651669 420 423 LGS SpecificDisease D015826 2651669 479 482 LGS SpecificDisease D015826 2651669 575 578 LGS SpecificDisease D015826 2651669 701 704 LGS SpecificDisease D015826 10930361|t|Iron-dependent self-assembly of recombinant yeast frataxin: implications for Friedreich ataxia. 10930361|a|Frataxin deficiency is the primary cause of Friedreich ataxia (FRDA), an autosomal recessive cardiodegenerative and neurodegenerative disease. Frataxin is a nuclear-encoded mitochondrial protein that is widely conserved among eukaryotes. Genetic inactivation of the yeast frataxin homologue (Yfh1p) results in mitochondrial iron accumulation and hypersensitivity to oxidative stress. Increased iron deposition and evidence of oxidative damage have also been observed in cardiac tissue and cultured fibroblasts from patients with FRDA. These findings indicate that frataxin is essential for mitochondrial iron homeostasis and protection from iron-induced formation of free radicals. The functional mechanism of frataxin, however, is still unknown. We have expressed the mature form of Yfh1p (mYfh1p) in Escherichia coli and have analyzed its function in vitro. Isolated mYfh1p is a soluble monomer (13, 783 Da) that contains no iron and shows no significant tendency to self-associate. Aerobic addition of ferrous iron to mYfh1p results in assembly of regular spherical multimers with a molecular mass of approximately 1. 1 MDa (megadaltons) and a diameter of 13 +/-2 nm. Each multimer consists of approximately 60 subunits and can sequester > 3, 000 atoms of iron. Titration of mYfh1p with increasing iron concentrations supports a stepwise mechanism of multimer assembly. Sequential addition of an iron chelator and a reducing agent results in quantitative iron release with concomitant disassembly of the multimer, indicating that mYfh1p sequesters iron in an available form. In yeast mitochondria, native mYfh1p exists as monomer and a higher-order species with a molecular weight > 600, 000. After addition of (55) Fe to the medium, immunoprecipitates of this species contain > 16 atoms of (55) Fe per molecule of mYfh1p. We propose that iron-dependent self-assembly of recombinant mYfh1p reflects a physiological role for frataxin in mitochondrial iron sequestration and bioavailability.. 10930361 77 94 Friedreich ataxia SpecificDisease D005621 10930361 96 115 Frataxin deficiency SpecificDisease D005621 10930361 140 157 Friedreich ataxia SpecificDisease D005621 10930361 159 163 FRDA SpecificDisease D005621 10930361 169 237 autosomal recessive cardiodegenerative and neurodegenerative disease DiseaseClass D030342+D019636 10930361 406 437 mitochondrial iron accumulation SpecificDisease D028361 10930361 442 478 hypersensitivity to oxidative stress SpecificDisease D004194 10930361 625 629 FRDA SpecificDisease D005621 10698963|t|Human mutations in glucose 6-phosphate dehydrogenase reflect evolutionary history. 10698963|a|Glucose 6-phosphate dehydrogenase (G6PD) is a cytosolic enzyme encoded by a housekeeping X-linked gene whose main function is to produce NADPH, a key electron donor in the defense against oxidizing agents and in reductive biosynthetic reactions. Inherited G6PD deficiency is associated with either episodic hemolytic anemia (triggered by fava beans or other agents) or life-long hemolytic anemia. We show here that an evolutionary analysis is a key to understanding the biology of a housekeeping gene. From the alignment of the amino acid (aa) sequence of 52 glucose 6-phosphate dehydrogenase (G6PD) species from 42 different organisms, we found a striking correlation between the aa replacements that cause G6PD deficiency in humans and the sequence conservation of G6PD two-thirds of such replacements are in highly and moderately conserved (50-99%) aa; relatively few are in fully conserved aa (where they might be lethal) or in poorly conserved aa, where presumably they simply would not cause G6PD deficiency. This is consistent with the notion that all human mutants have residual enzyme activity and that null mutations are lethal at some stage of development. Comparing the distribution of mutations in a human housekeeping gene with evolutionary conservation is a useful tool for pinpointing amino acid residues important for the stability or the function of the corresponding protein. In view of the current explosive increase in full genome sequencing projects, this tool will become rapidly available for numerous other genes.. 10698963 339 354 G6PD deficiency SpecificDisease D005955 10698963 381 406 episodic hemolytic anemia SpecificDisease D000743 10698963 452 478 life-long hemolytic anemia SpecificDisease D000745 10698963 791 806 G6PD deficiency SpecificDisease D005955 10698963 1082 1097 G6PD deficiency SpecificDisease D005955 8162051|t|Regionally clustered APC mutations are associated with a severe phenotype and occur at a high frequency in new mutation cases of adenomatous polyposis coli. 8162051|a|Germline mutation in APC at 5q21-22 results in the dominantly inherited syndrome adenomatous polyposis coli (APC). Somatic mutation in this gene is an early event in colorectal tumourigenesis. Both types of mutation are concentrated in the 5 half of exon 15. We have used single strand conformational polymorphism (SSCP) and heteroduplex analysis to screen for variants in this region of the gene in a total of 45 affected but unrelated individuals. Eighteen patients had no family history of the disease; of these 11 were classified as having a severe phenotype, based on an early age at presentation or cancer development. This compared with 6 of 27 familial cases. A 5 bp deletion at codon 1309 reported to occur in 10-15% of unselected APC patients worldwide, was found in 5 of the 18 new mutation cases and 4 of the 27 familial cases all nine were classed as severe. A further 3 new mutations and 1 familial mutation were located downstream from codon 1309, these individuals similarly being classed as phenotypically severe. In contrast all of the APC mutations detected in affected individuals with an average phenotype were located prior to codon 1309. The frequent association of a severe phenotype with fresh mutation may explain the apparent conflict of a high mutation rate (20-30%) in a condition, which on average, is lethal at a post-reproductive age.. 8162051 21 24 APC Modifier D011125 8162051 129 155 adenomatous polyposis coli SpecificDisease D011125 8162051 238 264 adenomatous polyposis coli SpecificDisease D011125 8162051 266 269 APC SpecificDisease D011125 8162051 762 768 cancer Modifier D009369 8162051 897 900 APC Modifier D011125 8162051 1212 1215 APC Modifier D011125 10205262|t|Analysis of alkaptonuria (AKU) mutations and polymorphisms reveals that the CCC sequence motif is a mutational hot spot in the homogentisate 1,2 dioxygenase gene (HGO). 10205262|a|We recently showed that alkaptonuria (AKU) is caused by loss-of-function mutations in the homogentisate 1, 2 dioxygenase gene (HGO). Herein we describe haplotype and mutational analyses of HGO in seven new AKU pedigrees. These analyses identified two novel single-nucleotide polymorphisms (INV4 + 31A-- > G and INV11 + 18A-- > G) and six novel AKU mutations (INV1-1G-- > A, W60G, Y62C, A122D, P230T, and D291E), which further illustrates the remarkable allelic heterogeneity found in AKU. Reexamination of all 29 mutations and polymorphisms thus far described in HGO shows that these nucleotide changes are not randomly distributed; the CCC sequence motif and its inverted complement, GGG, are preferentially mutated. These analyses also demonstrated that the nucleotide substitutions in HGO do not involve CpG dinucleotides, which illustrates important differences between HGO and other genes for the occurrence of mutation at specific short-sequence motifs. Because the CCC sequence motifs comprise a significant proportion (34. 5%) of all mutated bases that have been observed in HGO, we conclude that the CCC triplet is a mutational hot spot in HGO. 10205262 12 24 alkaptonuria Modifier D000474 10205262 26 29 AKU Modifier D000474 10205262 193 205 alkaptonuria SpecificDisease D000474 10205262 207 210 AKU SpecificDisease D000474 10205262 375 378 AKU Modifier D000474 10205262 513 516 AKU Modifier D000474 10205262 653 656 AKU SpecificDisease D000474 7450778|t|Further evidence for heterogeneity of glucose-6-phosphate dehydrogenase deficiency in Papua New Guinea. 7450778|a|Four new G6PD variants have been characterized in individuals from Papua New Guinea. This study demonstrates that the previously reported Markham variant and the newly characterized Salata variant may be widely distributed in Papua New Guinea. Th data presented here together with those of previously published studies demonstrate a degree of heterogeneity of G6PD deficiency that is much higher than that in other regions of the world where G6PD deficiency is common.. 7450778 38 82 glucose-6-phosphate dehydrogenase deficiency SpecificDisease D005955 7450778 464 479 G6PD deficiency SpecificDisease D005955 7450778 546 561 G6PD deficiency SpecificDisease D005955 10353787|t|Overgrowth of oral mucosa and facial skin, a novel feature of aspartylglucosaminuria. 10353787|a|Aspartylglucosaminuria (AGU) is a lysosomal storage disorder caused by deficiency of aspartylglucosaminidase (AGA). The main symptom is progressive mental retardation. A spectrum of different mutations has been reported in this disease, one missense mutation (Cys163Ser) being responsible for the majority of Finnish cases. We were able to examine 66 Finnish AGU patients for changes in the oral mucosa and 44 of these for changes in facial skin. Biopsy specimens of 16 oral lesions, 12 of them associated with the teeth, plus two facial lesions were studied histologically. Immunohistochemical staining for AGA was performed on 15 oral specimens. Skin was seborrhoeic in adolescent and adult patients, with erythema of the facial skin already common in childhood. Of 44 patients, nine (20%) had facial angiofibromas, tumours primarily occurring in association with tuberous sclerosis. Oedemic buccal mucosa (leucoedema) and gingival overgrowths were more frequent in AGU patients than in controls (p < 0. 001). Of 16 oral mucosal lesions studied histologically, 15 represented fibroepithelial or epithelial hyperplasias and were reactive in nature. Cytoplasmic vacuolisation was evident in four. Immunohistochemically, expression of AGA in AGU patients mucosal lesions did not differ from that seen in corresponding lesions of normal subjects. Thus, the high frequency of mucosal overgrowth in AGU patients does not appear to be directly associated with lysosomal storage or with alterations in the level of AGA expression. 10353787 0 41 Overgrowth of oral mucosa and facial skin CompositeMention D006965 10353787 62 84 aspartylglucosaminuria SpecificDisease D054880 10353787 86 108 Aspartylglucosaminuria SpecificDisease D054880 10353787 110 113 AGU SpecificDisease D054880 10353787 120 146 lysosomal storage disorder DiseaseClass D016464 10353787 157 194 deficiency of aspartylglucosaminidase SpecificDisease D054880 10353787 234 252 mental retardation DiseaseClass D008607 10353787 445 448 AGU Modifier D054880 10353787 617 631 facial lesions DiseaseClass D013568 10353787 794 821 erythema of the facial skin SpecificDisease D017445 10353787 882 902 facial angiofibromas SpecificDisease D018322 10353787 904 911 tumours DiseaseClass D009369 10353787 952 970 tuberous sclerosis SpecificDisease D014402 10353787 972 993 Oedemic buccal mucosa SpecificDisease D007967 10353787 995 1005 leucoedema SpecificDisease D007967 10353787 1011 1031 gingival overgrowths SpecificDisease D019214 10353787 1054 1057 AGU Modifier D054880 10353787 1104 1124 oral mucosal lesions SpecificDisease D009059 10353787 1164 1206 fibroepithelial or epithelial hyperplasias CompositeMention D017573 10353787 1327 1330 AGU Modifier D054880 10353787 1340 1355 mucosal lesions SpecificDisease D009059 10353787 1459 1477 mucosal overgrowth SpecificDisease D009059 10353787 1481 1484 AGU Modifier D054880 7550229|t|Molecular characterization of galactosemia (type 1) mutations in Japanese. 7550229|a|We characterized two novel mutations of the galactose-1-phosphate uridyltransferase (GALT) gene in two Japanese patients with GALT deficiency and identified N314D and R333W mutations, previously found in Caucasians. One novel missense mutation was an G-to-A transition in exon 8, resulting in the substitution of arginine by histidine at the codon 231 (R231H). GALT activity of the R231H mutant construct was reduced to 15% of normal controls in a COS cell expression system. The other was a splicing mutation, an A-to-G transition at the 38th nucleotide in exon 3 (318A-- > G), resulting in a 38-bp deletion in the GALT cDNA by activating a cryptic splice acceptor site. In seven Japanese families (14 alleles for classic form and one allele for Duarte variant) with GALT deficiency, the R231H and 318A-- > G mutations were found only on both alleles of the proband. The N314D and R333W mutations were found on one allele each. The Q188R was prevalent in the United States but not in Japanese patients. The N314D mutation was associated with the Duarte variant in Japanese persons, as well as in the United States. We speculate that classic galactosemia mutations appear to differ between Japanese and Caucasian patients. Our limited data set on galactosemia mutations in Japanese suggests that the N314D GALT mutation encoding the Duarte variant arose before Asian and Caucasian people diverged and that classic galactosemia mutations arose and/or accumulated after the divergence of Asian and Caucasian populations.. 7550229 30 42 galactosemia Modifier D005693 7550229 201 216 GALT deficiency SpecificDisease D005693 7550229 843 858 GALT deficiency SpecificDisease D005693 7550229 1209 1229 classic galactosemia Modifier D005693 7550229 1322 1334 galactosemia Modifier D005693 7550229 1481 1501 classic galactosemia Modifier D005693 1345170|t|Linkage disequilibrium mapping in isolated founder populations: diastrophic dysplasia in Finland. 1345170|a|Linkage disequilibrium mapping in isolated populations provides a powerful tool for fine structure localization of disease genes. Here, Luria and Delbrucks classical methods for analysing bacterial cultures are adapted to the study of human isolated founder populations in order to estimate (i) the recombination fraction between a disease locus and a marker; (ii) the expected degree of allelic homogeneity in a population; and (iii) the mutation rate of marker loci. Using these methods, we report striking linkage disequilibrium for diastrophic dysplasia (DTD) in Finland indicating that the DTD gene should lie within 0. 06 centimorgans (or about 60 kilobases) of the CSF1R gene. Predictions about allelic homogeneity in Finland and mutation rates in simple sequence repeats are confirmed by independent observations. 1345170 64 85 diastrophic dysplasia SpecificDisease C536170 1345170 634 655 diastrophic dysplasia SpecificDisease C536170 1345170 657 660 DTD SpecificDisease C536170 1345170 693 696 DTD Modifier C536170 10615125|t|Mutations in TNFRSF11A, affecting the signal peptide of RANK, cause familial expansile osteolysis. 10615125|a|Familial expansile osteolysis (FEO, MIM 174810) is a rare, autosomal dominant bone disorder characterized by focal areas of increased bone remodelling. The osteolytic lesions, which develop usually in the long bones during early adulthood, show increased osteoblast and osteoclast activity. Our previous linkage studies mapped the gene responsible for FEO to an interval of less than 5 cM between D18S64 and D18S51 on chromosome 18q21. 2-21 2-21. 3 in a large Northern Irish family. The gene encoding receptor activator of nuclear factor-kappa B (RANK; ref. 5), TNFRSF11A, maps to this region. RANK is essential in osteoclast formation. We identified two heterozygous insertion mutations in exon 1 of TNFRSF11A in affected members of four families with FEO or familial Paget disease of bone (PDB). One was a duplication of 18 bases and the other a duplication of 27 bases, both of which affected the signal peptide region of the RANK molecule. Expression of recombinant forms of the mutant RANK proteins revealed perturbations in expression levels and lack of normal cleavage of the signal peptide. Both mutations caused an increase in RANK-mediated nuclear factor-kappaB (NF-kappaB) signalling in vitro, consistent with the presence of an activating mutation. 10615125 68 97 familial expansile osteolysis SpecificDisease OMIM:174810 10615125 99 128 Familial expansile osteolysis SpecificDisease OMIM:174810 10615125 130 133 FEO SpecificDisease OMIM:174810 10615125 158 190 autosomal dominant bone disorder DiseaseClass D001847 10615125 223 249 increased bone remodelling SpecificDisease D001847 10615125 255 273 osteolytic lesions SpecificDisease D030981 10615125 451 454 FEO SpecificDisease OMIM:174810 10615125 852 855 FEO SpecificDisease OMIM:174810 10615125 859 889 familial Paget disease of bone SpecificDisease C538098 10615125 891 894 PDB SpecificDisease C538098 10196381|t|Adrenoleukodystrophy-related protein can compensate functionally for adrenoleukodystrophy protein deficiency (X-ALD): implications for therapy. 10196381|a|Inherited defects in the peroxisomal ATP-binding cassette (ABC) transporter adrenoleukodystrophy protein (ALDP) lead to the lethal peroxisomal disorder X-linked adrenoleukodystrophy (X-ALD), for which no efficient treatment has been established so far. Three other peroxisomal ABC transporters currently are known adrenoleukodystrophy-related protein (ALDRP), 70 kDa peroxisomal membrane protein (PMP70) and PMP70- related protein. By using transient and stable overexpression of human cDNAs encoding ALDP and its closest relative ALDRP, we could restore the impaired peroxisomal beta-oxidation in fibroblasts of X-ALD patients. The pathognomonic accumulation of very long chain fatty acids could also be prevented by overexpression of ALDRP in immortalized X-ALD cells. Immunofluorescence analysis demonstrated that the functional replacement of ALDP by ALDRP was not due to stabilization of the mutated ALDP itself. Moreover, we were able to restore the peroxisomal beta-oxidation defect in the liver of ALDP-deficient mice by stimulation of ALDRP and PMP70 gene expression through a dietary treatment with the peroxisome proliferator fenofibrate. These results suggest that a correction of the biochemical defect in X-ALD could be possible by drug-induced overexpression or ectopic expression of ALDRP.. 10196381 69 108 adrenoleukodystrophy protein deficiency SpecificDisease D000326 10196381 110 115 X-ALD SpecificDisease D000326 10196381 144 161 Inherited defects DiseaseClass D030342 10196381 220 240 adrenoleukodystrophy Modifier D000326 10196381 275 295 peroxisomal disorder DiseaseClass D018901 10196381 296 325 X-linked adrenoleukodystrophy SpecificDisease D000326 10196381 327 332 X-ALD SpecificDisease D000326 10196381 758 763 X-ALD Modifier D000326 10196381 1364 1369 X-ALD SpecificDisease D000326 8317477|t|The normal Huntington disease (HD) allele, or a closely linked gene, influences age at onset of HD. 8317477|a|We evaluated the hypothesis that Huntington disease (HD) is influenced by the normal HD allele by comparing transmission patterns of genetically linked markers at the D4S10 locus in the normal parent against age at onset in the affected offspring. Analysis of information from 21 sibships in 14 kindreds showed a significant tendency for sibs who have similar onset ages to share the same D4S10 allele from the normal parent. Affected sibs who inherited different D4S10 alleles from the normal parent tended to have more variable ages at onset. These findings suggest that the expression of HD is modulated by the normal HD allele or by a closely linked locus.. 8317477 11 29 Huntington disease Modifier D006816 8317477 31 33 HD Modifier D006816 8317477 96 98 HD SpecificDisease D006816 8317477 133 151 Huntington disease SpecificDisease D006816 8317477 153 155 HD SpecificDisease D006816 8317477 185 187 HD Modifier D006816 8317477 691 693 HD SpecificDisease D006816 8317477 721 723 HD Modifier D006816 10545613|t|Synergistic effect of histone hyperacetylation and DNA demethylation in the reactivation of the FMR1 gene. 10545613|a|Most fragile X syndrome patients have expansion of a (CGG) (n) sequence with > 200 repeats (full mutation) in the FMR1 gene responsible for this condition. Hypermethylation of the expanded repeat and of the FMR1 promoter is almost always present and apparently suppresses transcription, resulting in absence of the FMR1 protein. We recently showed that transcriptional reactivation of FMR1 full mutations can be achieved by inducing DNA demethylation with 5-azadeoxycytidine (5-azadC). The level of histone acetylation is another important factor in regulating gene expression; therefore, we treated lymphoblastoid cell lines of non-mosaic full mutation patients with three drugs capable of inducing histone hyperacetylation. We observed a consistent, although modest, reactivation of the FMR1 gene with 4-phenylbutyrate, sodium butyrate and trichostatin A, as shown by RT-PCR. However, we report that combining these drugs with 5-azadC results in a 2- to 5-fold increase in FMR1 mRNA levels obtained with 5-azadC alone, thus showing a marked synergistic effect of histone hyperacetylation and DNA demethylation in the reactivation of FMR1 full mutations.. 10545613 112 130 fragile X syndrome Modifier D005600 1358807|t|Genetic heterogeneity in X-linked amelogenesis imperfecta. 1358807|a|The AMELX gene located at Xp22. 1-p22. 3 encodes for the enamel protein amelogenin and has been implicated as the gene responsible for the inherited dental abnormality X-linked amelogenesis imperfecta (XAI). Three families with XAI have been investigated using polymorphic DNA markers flanking the position of AMELX. Using two-point linkage analysis, linkage was established between XAI and several of these markers in two families, with a combined lod score of 6. 05 for DXS16 at theta = 0. 04 04. This supports the involvement of AMELX, located close to DXS16, in the XAI disease process (AIH1) in those families. Using multipoint linkage analysis, the combined maximum lod score for these two families was 7. 30 for a location of AIH1 at 2 cM distal to DXS16. The support interval around this location extended about 8 cM proximal to DXS92, and the AIH1 location could not be precisely defined by multipoint mapping. Study of recombination events indicated that AIH1 lies in the interval between DXS143 and DXS85. There was significant evidence against linkage to this region in the third family, indicating locus heterogeneity in XAI. Further analysis with markers on the long arm of the X chromosome showed evidence of linkage to DXS144E and F9 with no recombination with either of these markers. Two-point analysis gave a peak lod score at DXS144E with a maximum lod score of 2. 83 at theta = 0, with a peak lod score in multipoint linkage analysis of 2. 84 at theta = 0. The support interval extended 9 cM proximal to DXS144E and 14 cM distal to F9. (ABSTRACT TRUNCATED AT 250 WORDS). 1358807 25 57 X-linked amelogenesis imperfecta SpecificDisease C538243 1358807 198 226 inherited dental abnormality DiseaseClass D014071 1358807 227 259 X-linked amelogenesis imperfecta SpecificDisease C538243 1358807 261 264 XAI SpecificDisease C538243 1358807 287 290 XAI SpecificDisease C538243 1358807 442 445 XAI SpecificDisease C538243 1358807 629 640 XAI disease SpecificDisease C538243 1358807 1193 1196 XAI SpecificDisease C538243 8304342|t|Anonymous marker loci within 400 kb of HLA-A generate haplotypes in linkage disequilibrium with the hemochromatosis gene (HFE) 8304342|a|The hemochromatosis gene (HFE) maps to 6p21. 3 and is less than 1 cM from the HLA class I genes; however, the precise physical location of the gene has remained elusive and controversial. The unambiguous identification of a crossover event within hemochromatosis families is very difficult; it is particularly hampered by the variability of the phenotypic expression as well as by the sex- and age-related penetrance of the disease. For these practical considerations, traditional linkage analysis could prove of limited value in further refining the extrapolated physical position of HFE. We therefore embarked upon a linkage-disequilibrium analysis of HFE and normal chromosomes from the Brittany population. In the present report, 66 hemochromatosis families yielding 151 hemochromatosis chromosomes and 182 normal chromosomes were RFLP-typed with a battery of probes, including two newly derived polymorphic markers from the 6. 7 and HLA-F loci located 150 and 250 kb telomeric to HLA-A, respectively. The results suggest a strong peak of existing linkage disequilibrium focused within the i82-to-6. 7 interval (approximately 250 kb). The zone of linkage disequilibrium is flanked by the i97 locus, positioned 30 kb proximal to i82, and the HLA-F gene, found 250 kb distal to HLA-A, markers of which display no significant association with HFE. These data support the possibility that HFE resides within the 400-kb expanse of DNA between i97 and HLA-F. Alternatively, the very tight association of HLA-A3 and allele 1 of the 6. 7 locus, both of which are comprised by the major ancestral or founder HFE haplotype in Brittany, supports the possibility that the disease gene may reside immediately telomeric to the 6. 7 locus within the linkage-disequilibrium zone. Additionally, hemochromatosis haplotypes possessing HLA-A11 and the low-frequency HLA-F polymorphism (allele 2) are supportive of a separate founder chromosome containing a second, independently arising mutant allele. Overall, the establishment of a likely " hemochromatosis critical region " centromeric boundary and the identification of a linkage-disequilibrium zone both significantly contribute to a reduction in the amount of DNA required to be searched for novel coding sequences constituting the HFE defect 8304342 100 115 hemochromatosis Modifier D006432 8304342 131 146 hemochromatosis Modifier D006432 8304342 374 389 hemochromatosis Modifier D006432 8304342 864 879 hemochromatosis Modifier D006432 8304342 902 917 hemochromatosis Modifier D006432 8304342 1909 1924 hemochromatosis Modifier D006432 8304342 2154 2169 hemochromatosis Modifier D006432 8304342 2399 2409 HFE defect SpecificDisease D006432 7617034|t|Natural selection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. 7617034|a|Glucose-6-phosphate dehydrogenase (G6PD) deficiency, the most common enzymopathy of humans, affects over 400 million people. The geographical correlation of its distribution with the historical endemicity of malaria suggests that this disorder has risen in frequency through natural selection by malaria. However, attempts to confirm that G6PD deficiency is protective in case-control studies of malaria have yielded conflicting results. Hence, for this X-linked disorder, it is unclear whether both male hemizygotes and female heterozygotes are protected or, as frequently suggested, only females. Furthermore, how much protection may be afforded is unknown. Here we report that, in two large case-control studies of over 2, 000 African children, the common African form of G6PD deficiency (G6PD A-) is associated with a 46-58% reduction in risk of severe malaria for both female heterozygotes and male hemizygotes. A mathematical model incorporating the measured selective advantage against malaria suggests that a counterbalancing selective disadvantage, associated with this enzyme deficiency, has retarded its rise in frequency in malaria-endemic regions. Although G6PD deficiency is now regarded as a generally benign disorder, in earlier environmental conditions it could have been significantly disadvantageous.. 7617034 49 64 G6PD deficiency SpecificDisease D005955 7617034 99 106 malaria SpecificDisease D008288 7617034 108 159 Glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 7617034 177 188 enzymopathy DiseaseClass D008661 7617034 316 323 malaria SpecificDisease D008288 7617034 404 411 malaria SpecificDisease D008288 7617034 447 462 G6PD deficiency SpecificDisease D005955 7617034 504 511 malaria SpecificDisease D008288 7617034 562 579 X-linked disorder DiseaseClass D040181 7617034 883 898 G6PD deficiency SpecificDisease D005955 7617034 900 907 G6PD A- SpecificDisease D005955 7617034 965 972 malaria SpecificDisease D008288 7617034 1101 1108 malaria SpecificDisease D008288 7617034 1187 1204 enzyme deficiency DiseaseClass D008661 7617034 1244 1251 malaria Modifier D008288 7617034 1278 1293 G6PD deficiency SpecificDisease D005955 6618488|t|Prader-Willi syndrome and chromosome 15. A clinical discussion of 20 cases. 6618488|a|A chromosome 15 anomaly was observed in 12 of 20 patients, 17 of whom were clinically suspected of having Prader-Willi syndrome (PWS). The clinical features of eight cases with 15q11-12 deletion were very similar to those originally described in PWS. On the other hand, the group of normal karyotype patients is heterogeneous, and their features do not strictly correspond to the clinical definition of PWS. However, the hypothesis that PWS is associated with deletion of 15q11-12 can neither explain the apparently balanced translocations of chromosome 15 nor account for the small supernumerary metacentric chromosomes corresponding to an isochromosome 15 for band 15q11 observed in some cases.. 6618488 0 21 Prader-Willi syndrome SpecificDisease D011218 6618488 78 99 chromosome 15 anomaly SpecificDisease D002869 6618488 182 203 Prader-Willi syndrome SpecificDisease D011218 6618488 205 208 PWS SpecificDisease D011218 6618488 322 325 PWS SpecificDisease D011218 6618488 479 482 PWS SpecificDisease D011218 6618488 513 516 PWS SpecificDisease D011218 1301187|t|Molecular basis of phenylketonuria and related hyperphenylalaninemias: mutations and polymorphisms in the human phenylalanine hydroxylase gene. 1301187|a|Mutations in the human phenylalanine hydroxylase gene producing phenylketonuria or hyperphenylalaninemia have now been identified in many patients from various ethnic groups. These mutations all exhibit a high degree of association with specific restriction fragment-length polymorphism haplotypes at the PAH locus. About 50 of these mutations are single-base substitutions, including six nonsense mutations and eight splicing mutations, with the remainder being missense mutations. One splicing mutation results in a 3 amino acid in-frame insertion. Two or 3 large deletions, 2 single codon deletions, and 2 single base deletions have been found. Twelve of the missense mutations apparently result from the methylation and subsequent deamination of highly mutagenic CpG dinucleotides. Recurrent mutation has been observed at several of these sites, producing associations with different haplotypes in different populations. About half of all missense mutations have been examined by in vitro expression analysis, and a significant correlation has been observed between residual PAH activity and disease phenotype. Since continuing advances in molecular methodologies have dramatically accelerated the rate in which new mutations are being identified and characterized, this register of mutations will be updated periodically.. 1301187 19 34 phenylketonuria SpecificDisease D010661 1301187 47 69 hyperphenylalaninemias DiseaseClass D010661 1301187 208 223 phenylketonuria SpecificDisease D010661 1301187 227 248 hyperphenylalaninemia DiseaseClass D010661 1483696|t|The intron 7 donor splice site transition: a second Tay-Sachs disease mutation in French Canada. 1483696|a|Mutations at the hexosaminidase A (HEXA) gene which cause Tay-Sachs disease (TSD) have elevated frequency in the Ashkenazi Jewish and French-Canadian populations. We report a novel TSD allele in the French-Canadian population associated with the infantile form of the disease. The mutation, a G-- > A transition at the + 1 position of intron 7, abolishes the donor splice site. Cultured human fibroblasts from a compound heterozygote for this transition (and for a deletion mutation) produce no detectable HEXA mRNA. The intron 7 + 1 mutation occurs in the base adjacent to the site of the adult-onset TSD mutation (G805A). In both mutations a restriction site for the endonuclease EcoRII is abolished. Unambiguous diagnosis, therefore, requires allele-specific oligonucleotide hybridization to distinguish between these two mutant alleles. The intron 7 + 1 mutation has been detected in three unrelated families. Obligate heterozygotes for the intron 7 + 1 mutation were born in the Saguenay-Lac-St-Jean region of Quebec. The most recent ancestors common to obligate carriers of this mutation were from the Charlevoix region of the province of Quebec. This mutation thus has a different geographic centre of diffusion and is probably less common than the exon 1 deletion TSD mutation in French Canadians. Neither mutation has been detected in France, the ancestral homeland of French Canada.. 1483696 52 69 Tay-Sachs disease Modifier D013661 1483696 155 172 Tay-Sachs disease SpecificDisease D013661 1483696 174 177 TSD SpecificDisease D013661 1483696 278 281 TSD Modifier D013661 1483696 699 702 TSD Modifier D013661 1483696 1369 1372 TSD Modifier D013661 10465113|t|Recessively inherited multiple epiphyseal dysplasia with normal stature, club foot, and double layered patella caused by a DTDST mutation. 10465113|a|We have observed over 25 different mutations in the diastrophic dysplasia sulphate transporter gene (DTDST) in association with the recessive disorders achondrogenesis 1B, atelosteogenesis 2, and diastrophic dysplasia. The c862t (R279W) transition is the most common mutation in non-Finnish patients, but in these disorders it is usually combined with other DTDST mutations. We had not seen a case of homozygosity for c862t (R279W) until we analysed DNA from a 36 year old male with tall-normal stature (180 cm) who asked for genetic counselling for suspected multiple epiphyseal dysplasia. He was treated for club foot and hip dysplasia at birth. Skeletal changes consistent with multiple epiphyseal dysplasia, with the peculiar finding of a double layered patella, were recognised during childhood. Cleft palate, swelling of the ear pinna, and hitch hiker thumb were absent. He was found to be homozygous, and both healthy parents heterozygous, for the R279W mutation in DTDST, and his fibroblasts showed a sulphate incorporation defect typical of DTDST disorders. Counselling was given for a recessive disorder, thereby considerably reducing the probability of affected offspring. Multiple epiphyseal dysplasia is more frequently caused by dominant mutations in the COMP (EDM1, McKusick 132400) and COL9A2 genes (EDM2, McKusick 600204). A few other patients and families with features similar to our proband have been described previously and considered to have autosomal recessive MED (EDM4, McKusick 226900). This observation confirms the existence of this entity and assigns it to the phenotypic spectrum associated with mutations at the DTDST locus.. 10465113 22 51 multiple epiphyseal dysplasia SpecificDisease D010009 10465113 73 82 club foot SpecificDisease D003025 10465113 88 110 double layered patella DiseaseClass C535504 10465113 191 212 diastrophic dysplasia Modifier C536170 10465113 271 290 recessive disorders DiseaseClass D030342 10465113 291 309 achondrogenesis 1B SpecificDisease C536016 10465113 311 329 atelosteogenesis 2 SpecificDisease C535395 10465113 335 356 diastrophic dysplasia SpecificDisease C536170 10465113 699 728 multiple epiphyseal dysplasia SpecificDisease D010009 10465113 749 758 club foot SpecificDisease D003025 10465113 763 776 hip dysplasia SpecificDisease D006618 10465113 820 849 multiple epiphyseal dysplasia SpecificDisease D010009 10465113 882 904 double layered patella DiseaseClass C535504 10465113 940 952 Cleft palate SpecificDisease D002972 10465113 954 979 swelling of the ear pinna DiseaseClass D004427 10465113 985 1002 hitch hiker thumb DiseaseClass C536903 10465113 1189 1204 DTDST disorders SpecificDisease D030342 10465113 1234 1252 recessive disorder DiseaseClass D030342 10465113 1323 1352 Multiple epiphyseal dysplasia SpecificDisease D010009 10465113 1414 1418 EDM1 SpecificDisease OMIM:132400 10465113 1455 1459 EDM2 SpecificDisease OMIM:600204 10465113 1624 1627 MED SpecificDisease D010009 10465113 1629 1633 EDM4 SpecificDisease OMIM:226900 8098180|t|Myotonic dystrophy: size- and sex-dependent dynamics of CTG meiotic instability, and somatic mosaicism. 8098180|a|Myotonic dystrophy (DM) is a progressive neuromuscular disorder which results from elongations of an unstable (CTG) n repeat, located in the 3 untranslated region of the DM gene. A correlation has been demonstrated between the increase in the repeat number of this sequence and the severity of the disease. However, the clinical status of patients cannot be unambiguously ascertained solely on the basis of the number of CTG repeats. Moreover, the exclusive maternal inheritance of the congenital form remains unexplained. Our observation of differently sized repeats in various DM tissues from the same individual may explain why the size of the mutation observed in lymphocytes does not necessarily correlate with the severity and nature of symptoms. Through a molecular and genetic study of 142 families including 418 DM patients, we have investigated the dynamics of the CTG repeat meiotic instability. A positive correlation between the size of the repeat and the intergenerational enlargement was observed similarly through male and female meioses for < or = 0. 5-kb CTG sequences. Beyond 0. 5 kb, the intergenerational variation was more important through female meioses, whereas a tendency to compression was observed almost exclusively in male meioses, for > or = 1. 5-kb fragments. This implies a size- and sex-dependent meiotic instability. Moreover, segregation analysis supports the hypothesis of a maternal as well as a familial predisposition for the occurrence of the congenital form. Finally, this analysis reveals a significant excess of transmitting grandfathers partially accounted for by increased fertility in affected males 8098180 0 18 Myotonic dystrophy SpecificDisease D009223 8098180 104 122 Myotonic dystrophy SpecificDisease D009223 8098180 124 126 DM SpecificDisease D009223 8098180 145 167 neuromuscular disorder DiseaseClass D009468 8098180 274 276 DM Modifier D009223 8098180 683 685 DM Modifier D009223 8098180 925 927 DM Modifier D009223 1517503|t|Fatal pyoderma gangrenosum in association with C7 deficiency. 1517503|a|Although pyoderma gangrenosum (PG) is often associated with systemic diseases, it has not been reported in association with congenital complement deficiencies. We describe an aggressive and ultimately fatal case of PG in a patient with a congenital C7 deficiency. Deficiencies of C7 can be associated with decreased neutrophil chemotaxis, phagocytosis, and opsonization, similar to the immunologic abnormalities described in patients with PG. Our patients decreased complement level, if not directly related to the development of PG, may have contributed to the aggressive nature of her disease.. 1517503 6 26 pyoderma gangrenosum SpecificDisease D017511 1517503 47 60 C7 deficiency SpecificDisease OMIM:610102 1517503 71 91 pyoderma gangrenosum SpecificDisease D017511 1517503 93 95 PG SpecificDisease D017511 1517503 122 139 systemic diseases DiseaseClass D034721 1517503 186 220 congenital complement deficiencies DiseaseClass D007153 1517503 277 279 PG SpecificDisease D017511 1517503 311 324 C7 deficiency SpecificDisease OMIM:610102 1517503 326 344 Deficiencies of C7 SpecificDisease OMIM:610102 1517503 448 473 immunologic abnormalities DiseaseClass D007153 1517503 501 503 PG SpecificDisease D017511 1517503 592 594 PG SpecificDisease D017511 8589715|t|Emerin deficiency at the nuclear membrane in patients with Emery-Dreifuss muscular dystrophy. 8589715|a|Mutations in the STA gene at the Xq28 locus have been found in patients with X-linked Emery-Dreifuss muscular dystrophy (EDMD). This gene encodes a hitherto unknown protein named emerin. To elucidate the subcellular localization of emerin, we raised two antisera against synthetic peptide fragments predicted from emerin cDNA. Using both antisera, we found positive nuclear membrane staining in skeletal, cardiac and smooth muscles in the normal controls and in patients with neuromuscular diseases other than EDMD. In contrast, a deficiency in immunofluorescent staining of skeletal and cardiac muscle from EDMD patients was observed. A 34 kD protein is immunoreactive with the antisera--the protein is equivalent to that predicted for emerin. Together, our findings suggest the specific deficiency of emerin in the nuclear membrane of muscle cells in patients with EDMD.. 8589715 0 17 Emerin deficiency SpecificDisease D020389 8589715 59 92 Emery-Dreifuss muscular dystrophy SpecificDisease D020389 8589715 171 213 X-linked Emery-Dreifuss muscular dystrophy SpecificDisease D020389 8589715 215 219 EDMD SpecificDisease D020389 8589715 570 592 neuromuscular diseases DiseaseClass D009468 8589715 604 608 EDMD SpecificDisease D020389 8589715 702 706 EDMD Modifier D020389 8589715 883 903 deficiency of emerin SpecificDisease D020389 8589715 961 965 EDMD SpecificDisease D020389 7652577|t|A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. 7652577|a|A mutated cyclin-dependent kinase 4 (CDK4) was identified as a tumor-specific antigen recognized by HLA-A2. 1-restricted autologous cytolytic T lymphocytes (CTLs) in a human melanoma. The mutated CDK4 allele was present in autologous cultured melanoma cells and metastasis tissue, but not in the patients lymphocytes. The mutation, an arginine-to-cysteine exchange at residue 24, was part of the CDK4 peptide recognized by CTLs and prevented binding of the CDK4 inhibitor p16INK4a, but not of p21 or of p27KIP1. The same mutation was found in one additional melanoma among 28 melanomas analyzed. These results suggest that mutation of CDK4 can create a tumor-specific antigen and can disrupt the cell-cycle regulation exerted by the tumor suppressor p16INK4a.. 7652577 82 90 melanoma SpecificDisease D008545 7652577 155 160 tumor Modifier D009369 7652577 266 274 melanoma SpecificDisease D008545 7652577 335 343 melanoma Modifier D008545 7652577 650 658 melanoma SpecificDisease D008545 7652577 668 677 melanomas SpecificDisease D008545 7652577 745 750 tumor Modifier D009369 7652577 825 830 tumor Modifier D009369 10487710|t|Molecular analysis in familial neurohypophyseal diabetes insipidus: early diagnosis of an asymptomatic carrier. 10487710|a|Familial neurohypophyseal diabetes insipidus (FNDI) is an inherited deficiency of the hormone arginine vasopressin (AVP) and is transmitted as an autosomal dominant trait. In the present study we have analyzed the AVP-neurophysin II (AVP-NPII) gene in a Spanish kindred. Studies were performed on seven members (four clinically affected) of the family. Patients were diagnosed at the Hospital Universitario Gregorio Maranon (Madrid, Spain). The entire coding region of the AVP-NPII gene of all family members was amplified by PCR and sequenced. All affected individuals presented a missense mutation (G1757-- > A) that replaces glycine at position 23 with arginine within the NPII domain. The substitution was confirmed by restriction endonuclease analysis and was present in heterozygosis. Additionally, one of the asymptomatic relatives (a girl 8 months old at the time of study) was identified as carrier of the same mutation and developed the disease 3 months later. The alteration found in the second exon of the gene in this family seems to be responsible for the disease, as all individuals harboring the mutation had been previously diagnosed or have eventually developed FNDI. Identification of the molecular defect underlying FNDI in affected families is a powerful tool for early asymptomatic diagnosis in infants.. 10487710 22 66 familial neurohypophyseal diabetes insipidus SpecificDisease OMIM:125700 10487710 112 156 Familial neurohypophyseal diabetes insipidus SpecificDisease OMIM:125700 10487710 158 162 FNDI SpecificDisease OMIM:125700 10487710 180 226 deficiency of the hormone arginine vasopressin DiseaseClass OMIM:125700 10487710 1292 1296 FNDI SpecificDisease OMIM:125700 10487710 1348 1352 FNDI SpecificDisease OMIM:125700 3572301|t|Sialophorin, a surface sialoglycoprotein defective in the Wiskott-Aldrich syndrome, is involved in human T lymphocyte proliferation. 3572301|a|The mAb L10 was used to determine the distribution and the function of sialophorin, the heavily glycosylated surface molecule that is deficient/defective in lymphocytes of patients with the X-linked immunodeficiency Wiskott-Aldrich syndrome. Dual-parameter FACS analysis indicated that sialophorin is expressed on CD4 + and CD8 + lymphocytes, on a subpopulation of peripheral blood B lymphocytes, on all thymocytes, and on a subpopulation of bone marrow cells. Functional studies demonstrated that L10 mAb stimulates the proliferation of peripheral blood T lymphocytes as measured by stimulation of [3H] thymidine incorporation. The time course and magnitude of increased [3H] thymidine incorporation by T lymphocytes in response to L10 mAb paralleled that induced by anti-CD3 mAb. Effective stimulation was dependent on the presence of monocytes and the Fc portion of L10 mAb. Stimulation of lymphocytes by L10, like stimulation by anti-CD3 mAb, involves increased expression of 4F2, HLA-DR, and IL-2-R. These observations suggest that sialophorin functions in T cell activation.. 3572301 58 82 Wiskott-Aldrich syndrome SpecificDisease D014923 3572301 323 373 X-linked immunodeficiency Wiskott-Aldrich syndrome SpecificDisease D014923 10732811|t|Characterization of the rat spinocerebellar ataxia type 3 gene. 10732811|a|Machado-Joseph disease (MJD) belongs to a group of clinically and genetically heterogeneous neurodegenerative disorders characterized by progressive cerebellar ataxia. The disease-causing mutation has recently been identified as an unstable and expanded (CAG) n trinucleotide repeat in a novel gene of unknown function. In Caucasians, repeat expansions in the MJD1 gene have also been found in patients with the clinically distinct autosomal dominant spinocerebellar ataxia type 3 (SCA3). In order to gain insight into the biology of the MJD1/SCA3 gene we cloned the rat homologue and studied its expression. The rat and human ataxin-3 genes are highly homologous with an overall sequence identity of approximately 88%. However, the C-terminal end of the putative protein differs strongly from the published human sequence. The (CAG) n block in the rat cDNA consists of just three interrupted units suggesting that a long polyglutamine stretch is not essential for the normal function of the ataxin-3 protein in rodents. The expression pattern of the SCA3 gene in various rat and human tissues was investigated by Northern blot analyses. The mature transcript is approximately 6 kb in length. In rat testis, a smaller transcript of 1. 3 kb was identified. Transcription of rsca3 was detected in most rat tissues including brain. Analyzing the expression level of the SCA3 gene in several human brain sections revealed no significant higher mRNA level in regions predominantly affected in MJD. Thus additional molecules and/or regulatory events are necessary to explain the exclusive degeneration of certain brain areas. 10732811 64 86 Machado-Joseph disease SpecificDisease D017827 10732811 88 91 MJD SpecificDisease D017827 10732811 156 183 neurodegenerative disorders DiseaseClass D019636 10732811 201 230 progressive cerebellar ataxia SpecificDisease D002524 10732811 515 544 spinocerebellar ataxia type 3 SpecificDisease D017827 10732811 546 550 SCA3 SpecificDisease D017827 10732811 1552 1555 MJD SpecificDisease D017827 10732811 1647 1682 degeneration of certain brain areas DiseaseClass D001927 7630639|t|The alveolar rhabdomyosarcoma PAX3/FKHR fusion protein is a transcriptional activator. 7630639|a|Chimeric transcription factors, created by gene fusions as the result of chromosomal translocations, have been implicated in the pathogenesis of several pathologically disparate solid tumors. The PAX3/FKHR fusion gene, formed by a t (2; 13) (q35; q14) in alveolar rhabdomyosarcoma, encodes a hybrid protein that contains both PAX3 DNA binding domains, the paired box and homeodomain, linked to the bisected DNA binding domain of FKHR, a member of the forkhead family of transcription factors. Here we report that PAX3 and PAX3/FKHR display similar, but not identical transactivation activities when tested with model Pax recognition sequences. No functional role could be ascribed solely to the residual FKHR binding domain present in the fusion protein, but FKHR was found to contribute a strong carboxyl terminal activation domain replacing the one located in the unrearranged PAX3 gene. We show that the native PAX3/FKHR protein present in tumor cells with this translocation has transcriptional characteristics similar to the in vitro expressed protein. The ability of the PAX3/FKHR hybrid protein to bind DNA in a sequence specific manner and to transactivate the expression of artificial reporter genes suggests that its aberrant expression could subvert the transcriptional programs that normally control the growth, differentiation, and survival of primitive myogenic precursors in vivo.. 7630639 4 29 alveolar rhabdomyosarcoma Modifier D018232 7630639 265 277 solid tumors DiseaseClass D009369 7630639 342 367 alveolar rhabdomyosarcoma SpecificDisease D018232 7630639 1030 1035 tumor Modifier D009369 7581380|t|Myotonia levior is a chloride channel disorder. 7581380|a|The group of dominant non-dystrophic myotonias, comprising disorders characterized by clinically similar forms of myogenic muscle stiffness, is genetically inhomogeneous. Dominant myotonia congenita (Thomsens disease) is linked to CLCN1, the gene encoding the major muscle chloride channel, localized on chromosome 7q35. In contrast, dominant myotonias sensitive to potassium are caused by point mutations in SCN4A on chromosome 17q, the gene for the alpha subunit of the adult skeletal muscle sodium channel. No linkage or molecular genetic data are as yet available on myotonia levior characterized by milder symptoms and later onset of myotonia than in Thomsens disease, and absence of muscle hypertrophy. We report a CLCN1 Gln-552-Arg substitution for a family with dominant inheritance previously diagnosed to have myotonia levior. Thus, this disorder appears as a variant of Thomsens disease due to mutations leading to low clinical expressivity. In addition, we report a novel Ile-290-Met CLCN1 mutation for a typical Thomsen pedigree. In another family previously diagnosed as having Thomsens disease, we unexpectedly found a CLCN1 14 bp deletion known to cause recessive myotonia, and a rare Trp-118-Gly polymorphism.. 7581380 0 15 Myotonia levior SpecificDisease D009224 7581380 21 46 chloride channel disorder DiseaseClass OMIM:160800 7581380 61 94 dominant non-dystrophic myotonias DiseaseClass C536245 7581380 219 246 Dominant myotonia congenita SpecificDisease D009224 7581380 248 264 Thomsens disease SpecificDisease D009224 7581380 382 400 dominant myotonias DiseaseClass D009224 7581380 619 634 myotonia levior SpecificDisease D009224 7581380 687 695 myotonia DiseaseClass D009222 7581380 704 720 Thomsens disease SpecificDisease D009224 7581380 737 755 muscle hypertrophy SpecificDisease C536106 7581380 868 883 myotonia levior SpecificDisease D009224 7581380 929 945 Thomsens disease SpecificDisease D009224 7581380 1140 1156 Thomsens disease SpecificDisease D009224 7581380 1218 1236 recessive myotonia DiseaseClass D009224 8282802|t|Markedly accelerated catabolism of apolipoprotein A-II (ApoA-II) and high density lipoproteins containing ApoA-II in classic lecithin: cholesterol acyltransferase deficiency and fish-eye disease. 8282802|a|Classic (complete) lecithin cholesterol acyltransferase (LCAT) deficiency and Fish-eye disease (partial LCAT deficiency) are genetic syndromes associated with markedly decreased plasma levels of high density lipoprotein (HDL) cholesterol but not with an increased risk of atherosclerotic cardiovascular disease. We investigated the metabolism of the HDL apolipoproteins (apo) apoA-I and apoA-II in a total of five patients with LCAT deficiency, one with classic LCAT deficiency and four with Fish-eye disease. Plasma levels of apoA-II were decreased to a proportionately greater extent (23% of normal) than apoA-I (30% of normal). In addition, plasma concentrations of HDL particles containing both apoA-I and apoA-II (LpA-I A-II) were much lower (18% of normal) than those of particles containing only apoA-I (LpA-I) (51% of normal). The metabolic basis for the low levels of apoA-II and LpA-I A-II was investigated in all five patients using both exogenous radiotracer and endogenous stable isotope labeling techniques. The mean plasma residence time of apoA-I was decreased at 2. 08 +/- 0. 27 d (controls 4. 74 +/- 0. 65 days); however, the residence time of apoA-II was even shorter at 1. 66 +/- 0. 24 d (controls 5. 25 +/- 0. 61 d). In addition, the catabolism of apoA-I in LpA-I A-II was substantially faster than that of apoA-I in LpA-I. In summary, genetic syndromes of either complete or partial LCAT deficiency result in low levels of HDL through preferential hypercatabolism of apoA-II and HDL particles containing apoA-II. Because LpA-I has been proposed to be more protective than LpA-I A-II against atherosclerosis, this selective effect on the metabolism of LpA-I A-II may provide a potential explanation why patients with classic LCAT deficiency and Fish-eye disease are not at increased risk for premature atherosclerosis despite markedly decreased levels of HDL cholesterol and apoA-I 8282802 135 173 cholesterol acyltransferase deficiency SpecificDisease D007863 8282802 178 194 fish-eye disease SpecificDisease D007863 8282802 196 270 Classic (complete) lecithin cholesterol acyltransferase (LCAT) deficiency CompositeMention D007863 8282802 275 291 Fish-eye disease SpecificDisease D007863 8282802 293 316 partial LCAT deficiency SpecificDisease D007863 8282802 469 507 atherosclerotic cardiovascular disease SpecificDisease D050197 8282802 625 640 LCAT deficiency SpecificDisease D007863 8282802 651 674 classic LCAT deficiency SpecificDisease D007863 8282802 689 705 Fish-eye disease SpecificDisease D007863 8282802 1585 1620 complete or partial LCAT deficiency CompositeMention D007863 8282802 1814 1829 atherosclerosis SpecificDisease D050197 8282802 1940 1963 classic LCAT deficiency SpecificDisease D007863 8282802 1968 1984 Fish-eye disease SpecificDisease D007863 8282802 2025 2040 atherosclerosis SpecificDisease D050197 10598803|t|The identical 5' splice-site acceptor mutation in five attenuated APC families from Newfoundland demonstrates a founder effect. 10598803|a|Inherited mutations of the APC gene predispose carriers to multiple adenomatous polyps of the colon and rectum and to colorectal cancer. Mutations located at the extreme 5 end of the APC gene, however, are associated with a less severe disease known as attenuated adenomatous polyposis coli (AAPC). Many individuals with AAPC develop relatively few colorectal polyps but are still at high risk for colorectal cancer. We report here the identification of a 5 APC germline mutation in five separately ascertained AAPC families from Newfoundland, Canada. This disease-causing mutation is a single basepair change (G to A) in the splice-acceptor region of APC intron 3 that creates a mutant RNA without exon 4 of APC. The observation of the same APC mutation in five families from the same geographic area demonstrates a founder effect. Furthermore, the identification of this germline mutation strengthens the correlation between the 5 location of an APC disease-causing mutation and the attenuated polyposis phenotype.. 10598803 55 69 attenuated APC Modifier C538265 10598803 155 158 APC Modifier D011125 10598803 196 238 adenomatous polyps of the colon and rectum CompositeMention D011125 10598803 246 263 colorectal cancer SpecificDisease D015179 10598803 311 314 APC Modifier D011125 10598803 381 418 attenuated adenomatous polyposis coli SpecificDisease C538265 10598803 420 424 AAPC SpecificDisease C538265 10598803 449 453 AAPC SpecificDisease C538265 10598803 477 494 colorectal polyps SpecificDisease D003111 10598803 526 543 colorectal cancer SpecificDisease D015179 10598803 586 589 APC Modifier D011125 10598803 639 643 AAPC Modifier C538265 10598803 780 783 APC Modifier D011125 10598803 870 873 APC Modifier D011125 10598803 1076 1079 APC Modifier D011125 10598803 1113 1133 attenuated polyposis Modifier C538265 1973404|t|Linkage of aspartylglucosaminuria (AGU) to marker loci on the long arm of chromosome 4. 1973404|a|Aspartylglucosaminuria (AGU) is caused by deficient activity of the enzyme aspartylglucosaminidase (AGA). The structural gene for AGA has been assigned to the region 4q21-qter of chromosome 4. We have studied the map position of the AGU locus in relation to other marker loci on the long arm of chromosome 4 using linkage analyses. Restriction fragment length polymorphism alleles for the ADH2, ADH3, EGF, FG alpha and FG beta loci and blood group antigens for the MNS locus were determined in a panel of 12 Finnish AGU families. The heterozygous family members were identified by reduced activity of AGA in lymphocytes. Linkage studies were performed using both pairwise and multipoint analyses. Loose linkage of the AGU locus to the FG and MNS loci was observed (z = 1. 16, z = 1. 39, respectively). Multipoint analysis to the fixed map [ADH- (0. 03) -EGF- (0. 35) -FG- (0. 11) -MNS] suggests that the location of the AGU locus is 0. 05-0. 30 recombination units distal to MNS (z = 3. 03). The order cen-ADH-EGF-FG-MNS-AGU is 35 times more likely than the next best order cen-ADH-EGF-AGU-FG-MNS. 1973404 11 33 aspartylglucosaminuria SpecificDisease D054880 1973404 35 38 AGU SpecificDisease D054880 1973404 88 110 Aspartylglucosaminuria SpecificDisease D054880 1973404 112 115 AGU SpecificDisease D054880 1973404 604 607 AGU Modifier D054880 1973404 806 809 AGU Modifier D054880 1671851|t|Linkage analysis in X-linked adrenoleukodystrophy and application in post- and prenatal diagnosis. 1671851|a|We have performed linkage analysis with the DNA markers DXS52 and the clotting factor VIII gene (F8C), in several large families with X-linked adrenoleukodystrophy (ALD). The tight linkage to DXS52 could be extended giving a maximal LOD score of 22. 5 at 1 cM. F8C was also tightly linked to ALD with a maximal LOD score of 7. 8 without recombination. Multipoint linkage analysis with the markers DXS304, DXS52, and F8C indicated that both the gene for ALD and for F8C are distal to DXS52. In four patients with ALD, no major structural rearrangement in the Xqter region was observed; in particular, there were no abnormalities in the vision blindness genes. DNA analysis appeared to be of use in determination of the carrier status of females at risk, for the determination of the origin of the mutation in a particular family, and for prenatal diagnosis. 1671851 20 49 X-linked adrenoleukodystrophy SpecificDisease D000326 1671851 233 262 X-linked adrenoleukodystrophy SpecificDisease D000326 1671851 264 267 ALD SpecificDisease D000326 1671851 391 394 ALD SpecificDisease D000326 1671851 552 555 ALD SpecificDisease D000326 1671851 611 614 ALD SpecificDisease D000326 1671851 713 756 abnormalities in the vision blindness genes DiseaseClass D003117 10090890|t|Multicentric origin of hemochromatosis gene (HFE) mutations. 10090890|a|Genetic hemochromatosis (GH) is believed to be a disease restricted to those of European ancestry. In northwestern Europe, > 80% of GH patients are homozygous for one mutation, the substitution of tyrosine for cysteine at position 282 (C282Y) in the unprocessed protein. In a proportion of GH patients, two mutations are present, C282Y and H63D. The clinical significance of this second mutation is such that it appears to predispose 1% -2% of compound heterozygotes to expression of the disease. The distribution of the two mutations differ, C282Y being limited to those of northwestern European ancestry and H63D being found at allele frequencies > 5%, in Europe, in countries bordering the Mediterranean, in the Middle East, and in the Indian subcontinent. The C282Y mutation occurs on a haplotype that extends T) introduces a premature stop codon at amino acid 286. Three exon 2 mutations were identified, including a single nucleotide deletion (2692delA) of codon 349 introducing a frameshift and premature termination codon, a 2 bp deletion (2673-2674delCT) that results in introduction of a stop codon at amino acid 343, and a G-- > A substitution in codon 429 (2931G-- > A) introducing a premature termination codon. All PLS patients were homozygous for cathepsin C mutations inherited from a common ancestor. Parents and sibs heterozygous for cathepsin C mutations do not show either the palmoplantar hyperkeratosis or severe early onset periodontitis characteristic of PLS. A more complete understanding of the functional physiology of cathepsin C carries significant implications for understanding normal and abnormal skin development and periodontal disease susceptibility 10593994 54 79 Papillon-Lefevre syndrome SpecificDisease D010214 10593994 81 106 Papillon-Lefevre syndrome SpecificDisease D010214 10593994 108 111 PLS SpecificDisease D010214 10593994 119 147 autosomal recessive disorder DiseaseClass D030342 10593994 165 192 palmoplantar hyperkeratosis SpecificDisease D007645 10593994 216 229 periodontitis SpecificDisease D010518 10593994 329 332 PLS SpecificDisease D010214 10593994 801 804 PLS SpecificDisease D010214 10593994 970 973 PLS SpecificDisease D010214 10593994 1506 1509 PLS Modifier D010214 10593994 1674 1701 palmoplantar hyperkeratosis SpecificDisease D007645 10593994 1724 1737 periodontitis SpecificDisease D010518 10593994 1756 1759 PLS SpecificDisease D010214 10593994 1927 1946 periodontal disease Modifier D010510 2910902|t|Identification of a single nucleotide change in the hypoxanthine-guanine phosphoribosyltransferase gene (HPRTYale) responsible for Lesch-Nyhan syndrome. 2910902|a|Complete deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT) causes the Lesch-Nyhan syndrome. Previous characterization of a mutant form of HPRT, HPRTYale, from a subject with the Lesch-Nyhan syndrome revealed normal mRNA and protein concentrations, no residual catalytic activity, and cathodal migration upon PAGE. We have cloned and sequenced HPRTYale cDNA. The nucleotide sequence of full-length HPRTYale cDNA revealed a single nucleotide substitution compared with normal HPRT cDNA G----C at nucleotide position 211. This transversion predicts substitution of arginine for glycine at amino acid position 71, explaining the cathodal migration of HPRTYale. Chou-Fasman secondary structure analysis predicts a change in the probability of beta-turn formation in the region containing the mutation. Inclusion of the bulky arginine side chain in place of glycine probably disrupts protein folding as well. Cloning mutant forms of cDNA allows identification of specific mutations, provides insight into mutational mechanisms, and facilitates structure-function analysis of mutant proteins.. 2910902 131 151 Lesch-Nyhan syndrome SpecificDisease D007926 2910902 162 222 deficiency of hypoxanthine-guanine phosphoribosyltransferase SpecificDisease D007926 2910902 241 261 Lesch-Nyhan syndrome SpecificDisease D007926 2910902 349 369 Lesch-Nyhan syndrome SpecificDisease D007926 2927388|t|Molecular detection of chromosomal translocations that disrupt the putative retinoblastoma susceptibility locus. 2927388|a|A candidate DNA sequence with many of the properties predicted for the retinoblastoma susceptibility (RB1) locus has been cloned (S. H. Friend, R. Bernards, S. Rogelj, R. A. Weinberg, J. M. Rapaport, D. M. Albert, and T. P. Dryja, Nature [London] 323 643-645, 1986). The large size of this gene (ca. 200 kilobases [kb]) and its multiple dispersed exons (Wiggs et al., N. Engl. J. Med. 318 151-157, 1988) complicate molecular screening strategies important in prenatal and presymptomatic diagnosis and in carrier detection. Here we used field inversion gel electrophoresis (FIGE) to construct a restriction map of approximately 1, 000 kb of DNA surrounding the RB1 locus and to detect the translocation breakpoints in three retinoblastoma patients. DNA probes from either the 5 or 3 end of the gene were used to detect a 250-kb EagI restriction fragment in DNA from unaffected individuals. Both probes identified an additional hybridizing fragment in the DNA from each patient, permitting the breakpoints in all three to be mapped within the cloned RB1 gene. Analysis of the breakpoint in one translocation cell line allowed the RB1 gene to be oriented with its 5 end toward the centromere. The 5 end of the gene also appeared to be associated with a clustering of sites for several infrequently cleaving restriction enzymes, indicating the presence of an HpaII tiny fragment island. The detection and mapping of the translocation breakpoints of all three retinoblastoma patients to within the putative RB1 gene substantiated the authenticity of this candidate sequence and demonstrated the utility of FIGE in detecting chromosomal rearrangements affecting this locus. 2927388 76 90 retinoblastoma Modifier D012175 2927388 184 198 retinoblastoma Modifier D012175 2927388 838 852 retinoblastoma Modifier D012175 2927388 1570 1584 retinoblastoma Modifier D012175 1338906|t|Constitutional mutations in the WT1 gene in patients with Denys-Drash syndrome. 1338906|a|The Denys-Drash syndrome is characterised by a typical nephropathy, genital abnormalities and also predisposes to the development of Wilms tumor. These patients eventually go into end stage renal failure. A candidate Wilms tumor gene, WT1, from the 11p13 chromosome region has recently been cloned. We have analysed the DNA sequence in constitutional cells from eight patients and have shown heterozygous mutations in six of them. Four of the mutations were in exon 9, all resulting in missense mutations. Three were at nucleotide position 1180 resulting in an arg > trp amino acid change. The other was at position 1186 converting an asp > asn in the predicted resultant protein. One patient had a missense mutation in exon 8, converting an arg > his. A single base pair insertion at nucleotide position 821 in exon 6 resulted in the generation of a premature stop codon in the last patient. We were unable to find a mutation in one patient despite complete sequencing of the genomic sequence of the gene. The last patient carried a constitutional deletion of the 11p13 region and no additional mutation was found. There was no obvious correlation between the type of mutation and phenotypic expression. These results further demonstrate that the WT1 gene is important in both the development of the kidney and the genito-urinary system.. 1338906 58 78 Denys-Drash syndrome SpecificDisease D030321 1338906 84 104 Denys-Drash syndrome SpecificDisease D030321 1338906 135 146 nephropathy DiseaseClass D007674 1338906 148 169 genital abnormalities DiseaseClass D014564 1338906 213 224 Wilms tumor SpecificDisease D009396 1338906 270 283 renal failure SpecificDisease D051437 1338906 297 308 Wilms tumor Modifier D009396 8198124|t|Four novel PEPD alleles causing prolidase deficiency. 8198124|a|Mutations at the PEPD locus cause prolidase deficiency (McKusick 170100), a rare autosomal recessive disorder characterized by iminodipeptiduria, skin ulcers, mental retardation, and recurrent infections. Four PEPD mutations from five severely affected individuals were characterized by analysis of reverse-transcribed, PCR-amplified (RT-PCR) cDNA. We used SSCP analysis on four overlapping cDNA fragments covering the entire coding region of the PEPD gene and detected abnormal SSCP bands for the fragment spanning all or part of exons 13-15 in three of the probands. Direct sequencing of the mutant cDNAs showed a G-- > A, 1342 substitution (G448R) in two patients and a 3-bp deletion (delta E452 or delta E453) in another. In the other two probands the amplified products were of reduced size. Direct sequencing of these mutant cDNAs revealed a deletion of exon 5 in one patient and of exon 7 in the other. Intronic sequences flanking exons 5 and 7 were identified using inverse PCR followed by direct sequencing. Conventional PCR and direct sequencing then established the intron-exon borders of the mutant genomic DNA revealing two splice acceptor mutations a G-- > C substitution at position -1 of intron 4 and an A-- > G substitution at position -2 of intron 6. Our results indicate that the severe form of prolidase deficiency is caused by multiple PEPD alleles. In this report we attempt to begin the process of describing these alleles and cataloging their phenotypic expression.. 8198124 32 52 prolidase deficiency SpecificDisease D056732 8198124 88 108 prolidase deficiency SpecificDisease D056732 8198124 135 163 autosomal recessive disorder DiseaseClass D030342 8198124 181 198 iminodipeptiduria SpecificDisease D000592 8198124 200 211 skin ulcers SpecificDisease D012883 8198124 213 231 mental retardation DiseaseClass D008607 8198124 1369 1389 prolidase deficiency SpecificDisease D056732 2817003|t|Translocation t(5;11)(q13.1;p13) associated with familial isolated aniridia. 2817003|a|A father and daughter with isolated aniridia were observed to have an apparently balanced, reciprocal translocation involving chromosomes 5 and 11 [t (5; 11) (q13. 1; p13)]. No other clinical characteristics often associated with the deletion of 11p13 were observed in this family. This finding, in association with 3 other instances of single breaks at 11p13 and aniridia, supports the assignment of AN2 to 11p13. 2817003 58 75 isolated aniridia SpecificDisease D015783 2817003 104 121 isolated aniridia SpecificDisease D015783 2817003 441 449 aniridia SpecificDisease D015783 10382909|t|Genotype-phenotype analysis in X-linked Emery-Dreifuss muscular dystrophy and identification of a missense mutation associated with a milder phenotype. 10382909|a|Direct sequencing of the emerin gene in 22 families with Emery-Dreifuss muscular dystrophy (EMD) revealed mutations in 21 (95%), confirming that emerin mutations can be identified in the majority of families with X-linked EMD. Most emerin mutations result in absence of the protein. In this study three mutations (a missense mutation Pro183Thr and two in-frame deletions removing residues 95-99 and 236-241, respectively) were unusual in being associated with expression of mutant protein. The phenotype in these families was compared in detail with the clinical features in cases with typical null mutations. For the in-frame deletions there were no significant differences. In the family with the missense mutation the phenotype was milder. Age at onset was later for first symptoms and for development of ankle contractures and muscle weakness. These findings have diagnostic implications as well as pointing to functionally important regions of the emerin protein.. 10382909 31 73 X-linked Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10382909 209 242 Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10382909 244 247 EMD SpecificDisease D020389 10382909 365 377 X-linked EMD SpecificDisease D020389 10382909 960 978 ankle contractures DiseaseClass D003286 10382909 983 998 muscle weakness DiseaseClass D018908 10947987|t|Asef, a link between the tumor suppressor APC and G-protein signaling. 10947987|a|The adenomatous polyposis coli gene (APC) is mutated in familial adenomatous polyposis and in sporadic colorectal tumors. Here the APC gene product is shown to bind through its armadillo repeat domain to a Rac-specific guanine nucleotide exchange factor (GEF), termed Asef. Endogenous APC colocalized with Asef in mouse colon epithelial cells and neuronal cells. Furthermore, APC enhanced the GEF activity of Asef and stimulated Asef-mediated cell flattening, membrane ruffling, and lamellipodia formation in MDCK cells. These results suggest that the APC-Asef complex may regulate the actin cytoskeletal network, cell morphology and migration, and neuronal function.. 10947987 25 30 tumor Modifier D009369 10947987 75 101 adenomatous polyposis coli Modifier D011125 10947987 127 157 familial adenomatous polyposis SpecificDisease D011125 10947987 174 191 colorectal tumors SpecificDisease D015179 10947987 202 205 APC Modifier D011125 3600794|t|Localization of the region homologous to the Duchenne muscular dystrophy locus on the mouse X chromosome. 3600794|a|Recent progress has resulted in part of the gene mutated in Duchenne and the milder Becker muscular dystrophies being cloned and has suggested that the gene itself extends over 1, 000 to 2, 000 kilobases (kb). To study how mutations in this gene affect muscle development and integrity, it would be of interest to have available a mouse model of the human disease. The mouse mdx mutation affects muscle and confers a mild dystrophic syndrome, but it is not clear whether this mutation is equivalent to Duchenne/Becker muscular dystrophy in man. Here we describe the use of two sequences from the human Duchenne muscular dystrophy (DMD) gene that cross-hybridize to mouse X-linked sequences to localize the gene homologous to DMD in the mouse. Both sequences map to the region of 10 centimorgan lying between the Tabby (Ta) and St14-1 (DxPas8) loci, close to the phosphorylase b kinase locus (Phk). By analogy with the human X-chromosome, we conclude that the region in the mouse around the G6pd and St14-1 loci may contain two genes corresponding to distinct human myopathies Emery Dreifuss muscular dystrophy which is known to be closely linked to St14-1 in man and the DMD homologue described here.. 3600794 45 72 Duchenne muscular dystrophy Modifier D020388 3600794 166 217 Duchenne and the milder Becker muscular dystrophies CompositeMention D020388|C537666 3600794 528 547 dystrophic syndrome SpecificDisease D009136 3600794 608 642 Duchenne/Becker muscular dystrophy CompositeMention D020388|C537666 3600794 708 735 Duchenne muscular dystrophy Modifier D020388 3600794 737 740 DMD Modifier D020388 3600794 831 834 DMD SpecificDisease D020388 3600794 1171 1181 myopathies DiseaseClass D009135 3600794 1183 1216 Emery Dreifuss muscular dystrophy SpecificDisease D020389 3600794 1278 1281 DMD Modifier D020388 1301937|t|Molecular basis of hexosaminidase A deficiency and pseudodeficiency in the Berks County Pennsylvania Dutch. 1301937|a|Following the birth of two infants with Tay-Sachs disease (TSD), a non-Jewish, Pennsylvania Dutch kindred was screened for TSD carriers using the biochemical assay. A high frequency of individuals who appeared to be TSD heterozygotes was detected (Kelly et al., 1975). Clinical and biochemical evidence suggested that the increased carrier frequency was due to at least two altered alleles for the hexosaminidase A alpha-subunit. We now report two mutant alleles in this Pennsylvania Dutch kindred, and one polymorphism. One allele, reported originally in a French TSD patient (Akli et al., 1991), is a GT-- > AT transition at the donor splice-site of intron 9. The second, a C-- > T transition at nucleotide 739 (Arg247Trp), has been shown by Triggs-Raine et al. (1992) to be a clinically benign " pseudodeficient " allele associated with reduced enzyme activity against artificial substrate. Finally, a polymorphism [G-- > A (759)], which leaves valine at codon 253 unchanged, is described . 1301937 19 46 hexosaminidase A deficiency SpecificDisease D013661 1301937 148 165 Tay-Sachs disease SpecificDisease D013661 1301937 167 170 TSD SpecificDisease D013661 1301937 231 234 TSD Modifier D013661 1301937 324 327 TSD Modifier D013661 1301937 673 676 TSD Modifier D013661 3856322|t|Gene transfer and expression of human phenylalanine hydroxylase. 3856322|a|Phenylketonuria (PKU) is caused by a genetic deficiency of the enzyme phenylalanine hydroxylase (PAH). A full-length complementary DNA clone of human PAH was inserted into a eukaryotic expression vector and transferred into mouse NIH3T3 cells which do not normally express PAH. The transformed mouse cells expressed PAH messenger RNA, immunoreactive protein, and enzymatic activity that are characteristic of the normal human liver products, demonstrating that a single gene contains all of the necessary genetic information to code for functional PAH. These results support the use of the human PAH probe in prenatal diagnosis and detection of carriers, to provide new opportunities for the biochemical characterization of normal and mutant enzymes, and in the investigation of alternative genetic therapies for PKU.. 3856322 65 80 Phenylketonuria SpecificDisease D010661 3856322 82 85 PKU SpecificDisease D010661 3856322 110 160 deficiency of the enzyme phenylalanine hydroxylase SpecificDisease OMIM:261600 3856322 878 881 PKU SpecificDisease D010661 8302543|t|Macular dystrophy associated with mutations at codon 172 in the human retinal degeneration slow gene. 8302543|a|BACKGROUND Recently, mutations in the retinal degeneration slow (rds) gene which codes for peripherin-rds have been implicated as a cause of autosomal dominant retinitis pigmentosa. Because this gene is expressed in both rods and cones, mutations in the rds gene might be expected to cause degeneration affecting either the scotopic or photopic systems. Mutations at codon 172 of the rds gene have been identified in three families with autosomal dominantly inherited, progressive macular dystrophy. METHODS Affected individuals underwent ophthalmic examination, scotopic perimetry, dark adaptometry, measurement of color-contrast sensitivity, and electroretinography to characterize the photoreceptor dysfunction. RESULTS In all but one affected member, symptoms of progressive central visual loss developed in the third or fourth decade of life accompanied by central scotoma and well-demarcated atrophy of the retinal pigment epithelium and choriocapillaris of the macula. In general, cone and rod thresholds were elevated, and color-contrast sensitivity was absent in the central visual field. Peripherally, the scotopic sensitivities were normal, as was the recovery from bleach. Cone electroretinograms were diminished in amplitude, and delayed in all affected adults except one. Rod electroretinograms were normal or near normal in amplitude, and had normal implicit times. Affected asymptomatic children had macular changes, abnormal color-contrast sensitivity, and reduced pattern and cone electroretinograms. CONCLUSION These results indicate that mutations in the rds gene can be expressed as a macular dystrophy, with evidence of primary cone dysfunction and preservation of peripheral rod function.. 8302543 0 17 Macular dystrophy SpecificDisease D008268 8302543 70 90 retinal degeneration Modifier D012162 8302543 141 161 retinal degeneration Modifier D012162 8302543 244 283 autosomal dominant retinitis pigmentosa SpecificDisease D012174 8302543 584 601 macular dystrophy SpecificDisease D008268 8302543 892 903 visual loss SpecificDisease C531604 8302543 967 982 central scotoma SpecificDisease D012607 8302543 1003 1044 atrophy of the retinal pigment epithelium SpecificDisease C536309 8302543 1049 1079 choriocapillaris of the macula SpecificDisease D008268 8302543 1712 1729 macular dystrophy SpecificDisease D008268 8302543 1756 1772 cone dysfunction DiseaseClass OMIM:180020 23402|t|Glucose 6-phosphate dehydrogenase variants: Gd (+) Alexandra associated with neonatal jaundice and Gd (-) Camperdown in a young man with lamellar cataracts. 23402|a|Two male subjects are described, with unusual clinical presentations and with hitherto undescribed G6PD variants. The first, of Italian extraction, suffered from severe neonatal jaundice following maternal ingestion of fresh broad beans (Vicia fava) both prenatally and postnatally the expression of the enzymatic defect was much more severe in the neonatal period than on retesting in adolescence, when biochemical characterization showed unique features which justify designation as a new variant Gd (+) Alexandra. The second patient, a boy of Maltese extraction who was found to have bilateral lamellar cataracts at the age of 4 years, was identified as G6PD deficient only as a result of a survey of children of Mediterranean origin with unexplained cataract formation; he has approximately 15% of normal enzyme activity, with another unique combination of biochemical characteristics which has led to its designation as Gd (-) Camperdown. Although this association may be coincidental, it prompts further attention to the possibility that under certain circumstances G6PD deficiency may favor cataract formation. The two cases illustrate the value of characterization of the mutant enzyme whenever unexpected clinical or laboratory results are obtained.. 23402 77 94 neonatal jaundice SpecificDisease D007567 23402 137 155 lamellar cataracts SpecificDisease C535342|OMIM:116800 23402 326 343 neonatal jaundice SpecificDisease D007567 23402 745 773 bilateral lamellar cataracts SpecificDisease C535342|OMIM:116800 23402 815 829 G6PD deficient SpecificDisease D005955 23402 912 920 cataract Modifier D002386 23402 1230 1245 G6PD deficiency SpecificDisease D005955 23402 1256 1264 cataract Modifier D002386 2989709|t|DNA restriction fragments associated with alpha 1-antitrypsin indicate a single origin for deficiency allele PI Z. 2989709|a|The alpha 1-protease inhibitor, or alpha-antitrypsin (AAT), a major plasma inhibitor of leukocyte elastase and bacterial proteases, is encoded at the PI locus on chromosome 14 (14q24. 3-q32. 1). A deficiency of AAT in individuals homozygous for the PI Z allele occurs in about 1 in 2, 000-8, 000 caucasians and is associated with an increased risk of early adult onset emphysema and liver disease in childhood. We have now used DNA polymorphisms associated with the AAT gene to investigate the origin of the PI Z allele. Using two genomic probes extending into the 5 and 3 flanking regions, respectively, we have identified eight polymorphic restriction sites. Extensive linkage disequilibrium occurs throughout the probed region with the PI Z allele, but not with normal PI M alleles. The Z allele occurs mainly with one haplotype, indicating a single, relatively recent, origin in caucasians 2989709 312 329 deficiency of AAT SpecificDisease C531610 2989709 484 493 emphysema DiseaseClass D004646 2989709 498 511 liver disease DiseaseClass D008107 10976074|t|Myotonic dystrophy in transgenic mice expressing an expanded CUG repeat. 10976074|a|Myotonic dystrophy (DM), the most common form of muscular dystrophy in adult humans, results from expansion of a CTG repeat in the 3 untranslated region of the DMPK gene. The mutant DMPK messenger RNA (mRNA) contains an expanded CUG repeat and is retained in the nucleus. We have expressed an untranslated CUG repeat in an unrelated mRNA in transgenic mice. Mice that expressed expanded CUG repeats developed myotonia and myopathy, whereas mice expressing a nonexpanded repeat did not. Thus, transcripts with expanded CUG repeats are sufficient to generate a DM phenotype. This result supports a role for RNA gain of function in disease pathogenesis.. 10976074 0 18 Myotonic dystrophy SpecificDisease D009223 10976074 73 91 Myotonic dystrophy SpecificDisease D009223 10976074 93 95 DM SpecificDisease D009223 10976074 122 140 muscular dystrophy SpecificDisease D009136 10976074 482 490 myotonia DiseaseClass D009222 10976074 495 503 myopathy DiseaseClass D009135 10976074 632 634 DM Modifier D009223 10797418|t|Submicroscopic deletion in cousins with Prader-Willi syndrome causes a grandmatrilineal inheritance pattern: effects of imprinting. 10797418|a|The Prader-Willi syndrome (PWS) critical region on 15q11-q13 is subject to imprinting. PWS becomes apparent when genes on the paternally inherited chromosome are not expressed. Familial PWS is rare. We report on a family in which a male and a female paternal first cousin both have PWS with cytogenetically normal karyotypes. Fluorescence in situ hybridization (FISH) analysis shows a submicroscopic deletion of SNRPN, but not the closely associated loci D15S10, D15S11, D15S63, and GABRB3. The cousins fathers and two paternal aunts have the same deletion and are clinically normal. The grandmother of the cousins is deceased and not available for study, and their grandfather is not deleted for SNRPN. DNA methylation analysis of D15S63 is consistent with an abnormality of the imprinting center associated with PWS. " Grandmatrilineal " inheritance occurs when a woman with deletion of an imprinted, paternally expressed gene is at risk of having affected grandchildren through her sons. In this case, PWS does not become evident as long as the deletion is passed through the matrilineal line. This represents a unique inheritance pattern due to imprinting.. 10797418 40 61 Prader-Willi syndrome SpecificDisease D011218 10797418 136 157 Prader-Willi syndrome SpecificDisease D011218 10797418 159 162 PWS SpecificDisease D011218 10797418 219 222 PWS SpecificDisease D011218 10797418 309 321 Familial PWS SpecificDisease D011218 10797418 414 417 PWS SpecificDisease D011218 10797418 946 949 PWS SpecificDisease D011218 10797418 1137 1140 PWS SpecificDisease D011218 10915770|t|Expression and imprinting of MAGEL2 suggest a role in Prader-willi syndrome and the homologous murine imprinting phenotype. 10915770|a|Prader-Willi syndrome (PWS) is caused by the loss of expression of imprinted genes in chromosome 15q11-q13. Affected individuals exhibit neonatal hypotonia, developmental delay and childhood-onset obesity. Necdin, a protein implicated in the terminal differentiation of neurons, is the only PWS candidate gene to reduce viability when disrupted in a mouse model. In this study, we have characterized MAGEL2 (also known as NDNL1), a gene with 51% amino acid sequence similarity to necdin and located 41 kb distal to NDN in the PWS deletion region. MAGEL2 is expressed predominantly in brain, the primary tissue affected in PWS and in several fetal tissues as shown by northern blot analysis. MAGEL2 is imprinted with monoallelic expression in control brain, and paternal-only expression in the central nervous system as demonstrated by its lack of expression in brain from a PWS-affected individual. The orthologous mouse gene (Magel2) is located within 150 kb of NDN , is imprinted with paternal-only expression and is expressed predominantly in late developmental stages and adult brain as shown by northern blotting, RT-PCR and whole-mount RNA in situ hybridization. Magel2 distribution partially overlaps that of NDN , with strong expression being detected in the central nervous system in mid-gestation mouse embryos by in situ hybridization. We hypothesize that, although loss of necdin expression may be important in the neonatal presentation of PWS, loss of MAGEL2 may be critical to abnormalities in brain development and dysmorphic features in individuals with PWS.. 10915770 54 75 Prader-willi syndrome SpecificDisease D011218 10915770 124 145 Prader-Willi syndrome SpecificDisease D011218 10915770 147 150 PWS SpecificDisease D011218 10915770 261 279 neonatal hypotonia SpecificDisease D009123 10915770 281 300 developmental delay DiseaseClass D002658 10915770 305 328 childhood-onset obesity DiseaseClass D009765 10915770 415 418 PWS Modifier D011218 10915770 650 653 PWS Modifier D011218 10915770 746 749 PWS SpecificDisease D011218 10915770 998 1001 PWS Modifier D011218 10915770 1576 1579 PWS SpecificDisease D011218 10915770 1615 1649 abnormalities in brain development DiseaseClass D002658 10915770 1654 1673 dysmorphic features DiseaseClass D057215 10915770 1694 1697 PWS SpecificDisease D011218 1968617|t|A single origin of phenylketonuria in Yemenite Jews. 1968617|a|Phenylketonuria (PKU) is a metabolic disease caused by recessive mutations of the gene encoding the hepatic enzyme phenylalanine hydroxylase (PAH). The incidence of PKU varies widely across different geographic areas, and is highest (about 1 in 5, 000 live births) in Ireland and western Scotland, and among Yemenite Jews. A limited number of point mutations account for most of the PKU cases in the European population. Here we report that a single molecular defect--a deletion spanning the third exon of the PAH gene--is responsible for all the PKU cases among the Yemenite Jews. Examination of a random sample of Yemenite Jews using a molecular probe that detects the carriers of this deletion indicated a high frequency of the defective gene in this community. Although the deleted PAH gene was traced to 25 different locations throughout Yemen, family histories and official documents of the Yemenite Jewish community showed that the common ancestor of all the carriers of this genetic defect lived in Sana, the capital of Yemen, before the eighteenth century.. 1968617 19 34 phenylketonuria SpecificDisease D010661 1968617 53 68 Phenylketonuria SpecificDisease D010661 1968617 70 73 PKU SpecificDisease D010661 1968617 80 97 metabolic disease DiseaseClass D008659 1968617 218 221 PKU SpecificDisease D010661 1968617 436 439 PKU Modifier D010661 1968617 600 603 PKU Modifier D010661 1968617 1036 1050 genetic defect DiseaseClass D030342 7909252|t|High resolution genetic analysis suggests one ancestral predisposing haplotype for the origin of the myotonic dystrophy mutation. 7909252|a|The mutation causing myotonic dystrophy (DM) has been identified as an amplification of an unstable trinucleotide (CTG) n repeat in over 99% of the global DM population. It is in complete linkage disequilibrium with an Alu element polymorphism within the DM kinase gene, suggesting that DM is a consequence of one or few ancestral mutations. A recent analysis utilizing this polymorphism as well as a flanking dinucleotide marker, suggested that similar to Fragile X syndrome, DM exhibited a founder effect (Imbert et al., 1993 Nature Genet. 4, 72-76). In contrast, the low reproductive fitness of individuals with congenital DM (the endpoint of genetic anticipation in myotonic dystrophy) suggests a higher rate of new mutations. We present a high resolution genetic analysis of the DM locus using PCR based assays of nine polymorphisms, spanning a physical distance of 30 kb, within and immediately flanking the DM kinase gene. The persistent complete allelic association of the DM mutation with all these polymorphisms provides further support to previous observations and suggests more strongly that the DM mutation occurred on the background of a particular haplotype in which the (CTG) n repeat became inherently unstable and therefore predisposed to amplification. 7909252 101 119 myotonic dystrophy Modifier D009223 7909252 151 169 myotonic dystrophy SpecificDisease D009223 7909252 171 173 DM SpecificDisease D009223 7909252 285 287 DM Modifier D009223 7909252 385 387 DM Modifier D009223 7909252 417 419 DM SpecificDisease D009223 7909252 587 605 Fragile X syndrome SpecificDisease D005600 7909252 607 609 DM SpecificDisease D009223 7909252 756 758 DM SpecificDisease D009223 7909252 800 818 myotonic dystrophy SpecificDisease D009223 7909252 914 916 DM Modifier D009223 7909252 1044 1046 DM Modifier D009223 7909252 1111 1113 DM Modifier D009223 7909252 1238 1240 DM Modifier D009223 1322637|t|Identification and rapid detection of three Tay-Sachs mutations in the Moroccan Jewish population. 1322637|a|Infantile Tay-Sachs disease (TSD) is caused by mutations in the HEXA gene that result in the complete absence of beta-hexosaminidase A activity. It is well known that an elevated frequency of TSD mutations exists among Ashkenazi Jews. More recently it has become apparent that elevated carrier frequencies for TSD also occur in several other ethnic groups, including Moroccan Jews, a subgroup of Sephardic Jews. Elsewhere we reported an in-frame deletion of one of the two adjacent phenylalanine codons at position 304 or 305 (delta F304/305) in one HEXA allele of a Moroccan Jewish TSD patient and in three obligate carriers from six unrelated Moroccan Jewish families. We have now identified two additional mutations within exon 5 of the HEXA gene that account for the remaining TSD alleles in the patient and carriers. One of the mutations is a novel C-to-G transversion, resulting in a replacement of Tyr180 by a stop codon. The other mutation is a G-to-A transition resulting in an Arg170-to-Gln substitution. This mutation is at a CpG site in a Japanese infant with Tay-Sachs disease and was described elsewhere. Analysis of nine obligate carriers from seven unrelated families showed that four harbor the delta F304/305 mutation, two the Arg170----Gln mutation, and one the Tyr180----Stop mutation. We also have developed rapid, nonradioactive assays for the detection of each mutation, which should be helpful for carrier screening.. 1322637 44 53 Tay-Sachs Modifier D013661 1322637 109 126 Tay-Sachs disease SpecificDisease D013661 1322637 128 131 TSD SpecificDisease D013661 1322637 291 294 TSD Modifier D013661 1322637 409 412 TSD SpecificDisease D013661 1322637 682 685 TSD Modifier D013661 1322637 880 883 TSD Modifier D013661 1322637 1171 1188 Tay-Sachs disease SpecificDisease D013661 7490097|t|Mapping of the mouse homologue of the Wilson disease gene to mouse chromosome 8. 7490097|a|ATP7B, the gene altered in Wilson disease (WD) patients, lies in a block of homology shared between human chromosome 13q14 and the central region of mouse chromosome 14. However, we have mapped the murine homologue of ATP7B (Atp7b) to mouse chromosome 8 by somatic cell hybrid analysis. Analysis of 80 interspecific backcross offspring was used to position Atp7b close to D8Mit3 and another ATPase locus, Atp4b, on mouse chromosome 8. ATP4B lies in 13q34 and is separated from ATP7B by several loci whose mouse homologues map to mouse chromosome 14. The assignment of Atp7b to mouse chromosome 8 identifies a previously unrecognized region of homology between this chromosome and human chromosome 13. This assignment suggests a possible location for the toxic milk mutation in the mouse, which has been proposed as a homologue of WD.. 7490097 38 52 Wilson disease Modifier D006527 7490097 108 122 Wilson disease Modifier D006527 7490097 124 126 WD Modifier D006527 7490097 911 913 WD SpecificDisease D006527 8116611|t|Characteristics of intergenerational contractions of the CTG repeat in myotonic dystrophy. 8116611|a|In myotonic dystrophy (DM), the size of a CTG repeat in the DM kinase gene generally increases in successive generations with clinical evidence of anticipation. However, there have also been cases with an intergenerational contraction of the repeat. We examined 1, 489 DM parent-offspring pairs, of which 95 (6. 4%) showed such contractions in peripheral blood leukocytes (PBL). In 56 of the 95 pairs, clinical data allowed an analysis of their anticipation status. It is surprising that anticipation occurred in 27 (48%) of these 56 pairs, while none clearly showed a later onset of DM in the symptomatic offspring. The contraction occurred in 76 (10%) of 753 paternal transmissions and in 19 (3%) of 736 maternal transmissions. Anticipation was observed more frequently in maternal (85%) than in paternal (37%) transmissions (P <. 001). The parental repeat size correlated with the size of intergenerational contraction (r2 =. 50, P < <. 001), and the slope of linear regression was steeper in paternal (-. 62) than in maternal (-. 30) transmissions (P < <. 001). Sixteen DM parents had multiple DM offspring with the CTG repeat contractions. This frequency was higher than the frequency expected from the probability of the repeat contractions (6. 4%) and the size of DM sib population (1. 54 DM offspring per DM parent, in 968 DM parents). We conclude that (1) intergenerational contractions of the CTG repeat in leukocyte DNA frequently accompanies apparent anticipation, especially when DM is maternally transmitted, and (2) the paternal origin of the repeat and the presence of the repeat contraction in a sibling increase the probability of the CTG repeat contraction 8116611 71 89 myotonic dystrophy SpecificDisease D009223 8116611 94 112 myotonic dystrophy SpecificDisease D009223 8116611 114 116 DM SpecificDisease D009223 8116611 151 153 DM Modifier D009223 8116611 360 362 DM Modifier D009223 8116611 675 677 DM SpecificDisease D009223 8116611 1165 1167 DM Modifier D009223 8116611 1189 1191 DM Modifier D009223 8116611 1362 1364 DM Modifier D009223 8116611 1387 1389 DM Modifier D009223 8116611 1404 1406 DM Modifier D009223 8116611 1422 1424 DM Modifier D009223 8116611 1584 1586 DM SpecificDisease D009223 10571950|t|Splice-site mutation in the PDS gene may result in intrafamilial variability for deafness in Pendred syndrome. 10571950|a|Pendred syndrome is a recessive inherited disorder that consists of developmental abnormalities of the cochlea, sensorineural hearing loss, and diffuse thyroid enlargement (goiter). This disorder may account for up to 10% of cases of hereditary deafness. The disease gene (PDS) has been mapped to chromosome 7q22-q31, and encodes a chloride-iodide transport protein. We performed mutation analysis of individual exons of the PDS gene in one Spanish family that shows intrafamilial variability of the deafness phenotype (two patients with profound and one with moderate-severe deafness). We identified a new splice-site mutation affecting intron 4 of the PDS gene, at nucleotide position 639 + 7. RNA analysis from lymphocytes of the affected patients showed that mutation 639 + 7A-- > G generates a new donor splice site, leading to an mRNA with an insertion of six nucleotides from intron 4 of PDS. Since the newly created donor splice site is likely to compete with the normal one, variations of the levels of normal and aberrant transcripts of the PDS gene in the cochlea may explain the variability in the deafness presentation.. 10571950 28 31 PDS Modifier C536648 10571950 81 89 deafness SpecificDisease D003638 10571950 93 109 Pendred syndrome SpecificDisease C536648 10571950 111 127 Pendred syndrome SpecificDisease C536648 10571950 133 161 recessive inherited disorder DiseaseClass D030342 10571950 179 221 developmental abnormalities of the cochlea DiseaseClass D015834 10571950 223 249 sensorineural hearing loss SpecificDisease D006319 10571950 255 282 diffuse thyroid enlargement SpecificDisease D013959 10571950 284 290 goiter SpecificDisease D006042 10571950 345 364 hereditary deafness SpecificDisease D030342+D003638 10571950 384 387 PDS SpecificDisease C536648 10571950 536 539 PDS Modifier C536648 10571950 611 619 deafness Modifier D003638 10571950 687 695 deafness SpecificDisease D003638 10571950 765 768 PDS Modifier C536648 10571950 1162 1165 PDS Modifier C536648 10571950 1221 1229 deafness Modifier D003638 10802668|t|Heterozygous loss of Six5 in mice is sufficient to cause ocular cataracts. 10802668|a|Myotonic dystrophy (DM) is an autosomal dominant disorder characterized by skeletal muscle wasting, myotonia, cardiac arrhythmia, hyperinsulinaemia, mental retardation and ocular cataracts. The genetic defect in DM is a CTG repeat expansion located in the 3 untranslated region of DMPK and 5 of a homeodomain-encoding gene, SIX5 (formerly DMAHP; refs 2-5). There are three mechanisms by which CTG expansion can result in DM. First, repeat expansion may alter the processing or transport of the mutant DMPK mRNA and consequently reduce DMPK levels. Second, CTG expansion may establish a region of heterochromatin 3 of the repeat sequence and decrease SIX5 transcription. Third, toxic effects of the repeat expansion may be intrinsic to the repeated elements at the level of DNA or RNA (refs 10, 11). Previous studies have demonstrated that a dose-dependent loss of Dm15 (the mouse DMPK homologue) in mice produces a partial DM phenotype characterized by decreased development of skeletal muscle force and cardiac conduction disorders. To test the role of Six5 loss in DM, we have analysed a strain of mice in which Six5 was deleted. Our results demonstrate that the rate and severity of cataract formation is inversely related to Six5 dosage and is temporally progressive. Six5 +/- and Six5-/- mice show increased steady-state levels of the Na +/K + -ATPase alpha-1 subunit and decreased Dm15 mRNA levels. Thus, altered ion homeostasis within the lens may contribute to cataract formation. As ocular cataracts are a characteristic feature of DM, these results demonstrate that decreased SIX5 transcription is important in the aetiology of DM. Our data support the hypothesis that DM is a contiguous gene syndrome associated with the partial loss of both DMPK and SIX5.. 10802668 64 73 cataracts SpecificDisease D002386 10802668 75 93 Myotonic dystrophy SpecificDisease D009223 10802668 95 97 DM SpecificDisease D009223 10802668 105 131 autosomal dominant disorde DiseaseClass D030342 10802668 159 173 muscle wasting SpecificDisease D009133 10802668 175 183 myotonia DiseaseClass D009222 10802668 185 203 cardiac arrhythmia DiseaseClass D001145 10802668 205 222 hyperinsulinaemia SpecificDisease D006946 10802668 224 242 mental retardation DiseaseClass D008607 10802668 254 263 cataracts SpecificDisease D002386 10802668 269 283 genetic defect DiseaseClass D030342 10802668 287 289 DM SpecificDisease D009223 10802668 496 498 DM SpecificDisease D009223 10802668 998 1000 DM Modifier D009223 10802668 1079 1107 cardiac conduction disorders DiseaseClass D001145 10802668 1142 1144 DM SpecificDisease D009223 10802668 1261 1269 cataract Modifier D002386 10802668 1544 1552 cataract Modifier D002386 10802668 1574 1583 cataracts SpecificDisease D002386 10802668 1616 1618 DM SpecificDisease D009223 10802668 1713 1715 DM SpecificDisease D009223 10802668 1754 1756 DM SpecificDisease D009223 10484765|t|Cis and trans effects of the myotonic dystrophy (DM) mutation in a cell culture model. 10484765|a|The mutation causing myotonic dystrophy (DM) has been identified as a CTG expansion in the 3-untranslated region (3-UTR) of the DM protein kinase gene (DMPK), but the mechanism (s) of pathogenesis remain unknown. Studies using DM patient materials have often produced confusing results. Therefore, to study the effects of the DM mutation in a controlled environment, we have established a cell culture model system using C2C12 mouse myoblasts. By expressing chimeric reporter constructs containing a reporter gene fused to a human DMPK 3-UTR, we identified both cis and trans effects that are mediated by the DM mutation. Our data show that a mutant DMPK 3-UTR, with as few as 57 CTGs, had a negative cis effect on protein expression and resulted in the aggregation of reporter transcripts into discrete nuclear foci. We determined by deletion analysis that an expanded (CTG) (n) tract alone was sufficient to mediate these cis effects. Furthermore, in contrast to the normal DMPK 3-UTR mRNA, a mutant DMPK 3-UTR mRNA with (CUG) (200) selectively inhibited myogenic differentiation of C2C12 myoblasts. Genetic analysis and the Cre- loxP system were used to clearly demonstrate that the myoblast fusion defect could be rescued by eliminating the expression of the mutant DMPK 3-UTR transcript. Characterization of spontaneous deletion events mapped the inhibitory effect to the (CTG) (n) expansion and/or the 3 end of the DMPK 3-UTR. These results provide evidence that the DM mutation acts in cis to reduce protein production (consistent with DMPK haploinsufficiency) and in trans as a riboregulator to inhibit myogenesis.. 10484765 29 47 myotonic dystrophy Modifier D009223 10484765 49 51 DM Modifier D009223 10484765 108 126 myotonic dystrophy SpecificDisease D009223 10484765 128 130 DM SpecificDisease D009223 10484765 215 217 DM Modifier D009223 10484765 314 316 DM Modifier D009223 10484765 413 415 DM Modifier D009223 10484765 696 698 DM Modifier D009223 10484765 1560 1562 DM Modifier D009223 10484765 1630 1653 DMPK haploinsufficiency SpecificDisease D058495 8071955|t|Intelligence quotient profile in myotonic dystrophy, intergenerational deficit, and correlation with CTG amplification. 8071955|a|An abbreviated Wechsler Adult Intelligence Scale Revised (WAIS-R) was used to assess verbal and arithmetical cognitive performance in 55 subjects with myotonic dystrophy (DM), covering all grades of disease severity, and 31 controls at 50% risk of inheriting DM. Scaled scores from the assessment were converted into an intelligence quotient (IQ) estimation on each person. Significant IQ differences were found between (1) all 55 DM subjects (mean 90. 2, SD 16. 1) and 31 controls (102. 6, SD 9. 4), with no sex differences in either group; (2) 15 affected parents (99. 3, SD 12. 2) and their affected children (88. 1, SD 17. 2), where significance was dependent on parental sex being female; and (3) 15 pairs of affected sibs (89. 6, SD 13. 2) and their normal sibs (100. 2, SD 7. 6). IQ steadily declined as (1) the age of onset of signs and symptoms decreased, and (2) the CTG expansion size increased. The correlation appeared to be more linear with age of onset. The correlation of IQ difference and CTG expansion difference in both the DM parent-child pairs and normal sib-affected sib pairs was poor, indicating that CTG expansion is not a reliable predictor of IQ either in individual persons or families. Further analysis of cognitive function in DM is required to clarify specific deficits characteristic of this patient group 8071955 33 51 myotonic dystrophy SpecificDisease D009223 8071955 271 289 myotonic dystrophy SpecificDisease D009223 8071955 291 293 DM SpecificDisease D009223 8071955 379 381 DM SpecificDisease D009223 8071955 552 554 DM Modifier D009223 8071955 1164 1166 DM Modifier D009223 8071955 1378 1380 DM SpecificDisease D009223 1361318|t|Complement factor 2 deficiency: a clinical and serological family study. 1361318|a|Inherited complement deficiencies are associated with a variety of connective tissue diseases. A family with inherited deficiency of complement factor 2 (C2) is described in which two family members with homozygous C2 deficiency developed cutaneous vasculitis and sicca syndrome. The other family members had heterozygous C2 deficiency and each member had the HLA-A25, B18, DR2 (w15) haplotype. The mother had seropositive rheumatoid arthritis. Further studies showed the presence of cryoglobulins, antibodies against endothelial cells, and anticardiolipin antibodies.. 1361318 0 30 Complement factor 2 deficiency SpecificDisease OMIM:217000 1361318 73 106 Inherited complement deficiencies DiseaseClass D007153 1361318 182 225 inherited deficiency of complement factor 2 SpecificDisease OMIM:217000 1361318 288 301 C2 deficiency SpecificDisease OMIM:217000 1361318 312 332 cutaneous vasculitis SpecificDisease D018366 1361318 337 351 sicca syndrome SpecificDisease D012859 1361318 395 408 C2 deficiency SpecificDisease OMIM:217000 1361318 483 516 seropositive rheumatoid arthritis SpecificDisease D001172 1352883|t|Oncogenic point mutations in exon 20 of the RB1 gene in families showing incomplete penetrance and mild expression of the retinoblastoma phenotype. 1352883|a|The retinoblastoma-predisposition gene, RB1, segregates as an autosomal dominant trait with high (90%) penetrance. Certain families, however, show an unusual low-penetrance phenotype with many individuals being unaffected, unilaterally affected, or with evidence of spontaneously regressed tumors. We have used single-strand conformation polymorphism analysis and PCR sequencing to study two such families. Mutations were found in exon 20 of RB1 in both cases. In one family a C----T transition in codon 661 converts an arginine (CGG) to a tryptophan (TGG) codon. In this family, incomplete penetrance and mild phenotypic expression were observed in virtually all patients, possibly indicating that single amino acid changes may modify protein structure/function such that tumorigenesis is not inevitable. In the second family the mutation in codon 675 is a G----T transversion that converts a glutamine (GAA) to a stop (TAA) codon. However, this mutation also occurs near a potential cryptic splice acceptor site, raising the possibility of alternative splicing resulting in a less severely disrupted protein.. 1352883 122 136 retinoblastoma Modifier D012175 1352883 152 166 retinoblastoma Modifier D012175 1352883 438 444 tumors DiseaseClass D009369 10502833|t|Identification of a novel R21X mutation in the liver-type arginase gene (ARG1) in four Portuguese patients with argininemia. 10502833|a|Argininemia is a rare autossomal recessive disorder caused by deficiency in the cytosolic liver-type arginase enzyme (L-arginine urea-hydrolase; E. C. 3. 5. 3. 1). In order to investigate the molecular basis for argininemia in four unrelated Portuguese patients (two from northern Portugal and two from Madeira Island) we performed a DNA sequence analysis of all the exons and exon/intron boundaries of the liver-type arginase gene (ARG1). All patients were found to be homozygous for a newly identified C - > T transition in codon 21 (exon 2) substituting arginine for a premature stop codon (R21X CGA to TGA) and generating a NlaIII restriction site. Restriction digestion following PCR amplification of ARG1 exon 2 confirmed the presence of the mutation. 10502833 112 123 argininemia SpecificDisease D020162 10502833 125 136 Argininemia SpecificDisease D020162 10502833 147 176 autossomal recessive disorder DiseaseClass D030342 10502833 187 241 deficiency in the cytosolic liver-type arginase enzyme DiseaseClass D020162 10502833 337 348 argininemia SpecificDisease D020162 10930571|t|A mutation in the pleckstrin homology (PH) domain of the FGD1 gene in an Italian family with faciogenital dysplasia (Aarskog-Scott syndrome). 10930571|a|Aarskog-Scott Syndrome (AAS) is an X-linked disorder characterised by short stature and multiple facial, limb and genital abnormalities. A gene, FGD1, altered in a patient with AAS phenotype, has been identified and found to encode a protein with homology to Rho/Rac guanine nucleotide exchange factors (Rho/Rac GEF). However, since this original report on identification of a mutated FGD1 gene in an AAS patient, no additional mutations in the FGD1 gene have been described. We analysed 13 independent patients with clinical diagnosis of AAS. One patient presented a mutation that results in a nucleotide change in exon 10 of the FGD1 gene (G2559 > A) substituting a Gln for Arg in position 610. The mutation was found to segregate with the AAS phenotype in affected males and carrier females in the family of this patient. Interestingly, Arg-610 is located within one of the two pleckstrin homology (PH) domains of the FGD1 gene and it corresponds to a highly conserved residue which has been involved in InsP binding in PH domains of other proteins. The same residue is often mutated in the Brutons tyrosine kinase (Btk) gene in patients with an X-linked agammaglobulinemia. The Arg610Gln mutation represents the first case of a mutation in the PH domain of the FGD1 gene and additional evidence that mutations in PH domains can be associated to human diseases.. 10930571 93 115 faciogenital dysplasia SpecificDisease C535331 10930571 117 139 Aarskog-Scott syndrome SpecificDisease C535331 10930571 142 164 Aarskog-Scott Syndrome SpecificDisease C535331 10930571 166 169 AAS SpecificDisease C535331 10930571 177 194 X-linked disorder DiseaseClass D040181 10930571 212 225 short stature DiseaseClass D006130 10930571 239 277 facial, limb and genital abnormalities CompositeMention D019465|D017880|D014564 10930571 319 322 AAS Modifier C535331 10930571 543 546 AAS Modifier C535331 10930571 681 684 AAS SpecificDisease C535331 10930571 884 887 AAS Modifier C535331 10930571 1291 1318 X-linked agammaglobulinemia SpecificDisease OMIM:300755 2055114|t|Localization of histidase to human chromosome region 12q22----q24.1 and mouse chromosome region 10C2----D1. 2055114|a|The human gene for histidase (histidine ammonia-lyase; HAL), the enzyme deficient in histidinemia, was assigned to human chromosome 12 by Southern blot analysis of human X mouse somatic cell hybrid DNA. The gene was sublocalized to region 12q22----q24. 1 by in situ hybridization, using a human histidase cDNA. The homologous locus in the mouse (Hal) was mapped to region 10C2----D1 by in situ hybridization, using a cell line from a mouse homozygous for a 1. 10 Robertsonian translocation. These assignments extend the conserved syntenic region between human chromosome 12 and mouse chromosome 10 that includes the genes for phenylalanine hydroxylase, gamma interferon, peptidase, and citrate synthase. The localization of histidase to mouse chromosome 10 suggests that the histidase regulatory locus (Hsd) and the histidinemia mutation (his), which are both known to be on chromosome 10, may be alleles of the histidase structural gene locus. 2055114 193 205 histidinemia SpecificDisease C538320 10364521|t|Noninvasive test for fragile X syndrome, using hair root analysis. 10364521|a|Identification of the FMR1 gene and the repeat-amplification mechanism causing fragile X syndrome led to development of reliable DNA-based diagnostic methods, including Southern blot hybridization and PCR. Both methods are performed on DNA isolated from peripheral blood cells and measure the repeat size in FMR1. Using an immunocytochemical technique on blood smears, we recently developed a novel test for identification of patients with fragile X syndrome. This method, also called " antibody test, " uses monoclonal antibodies against the FMR1 gene product (FMRP) and is based on absence of FMRP in patients cells. Here we describe a new diagnostic test to identify male patients with fragile X syndrome, on the basis of lack of FMRP in their hair roots. Expression of FMRP in hair roots was studied by use of an FMRP-specific antibody test, and the percentage of FMRP-expressing hair roots in controls and in male fragile X patients was determined. Control individuals showed clear expression of FMRP in nearly every hair root, whereas male fragile X patients lacked expression of FMRP in almost all their hair roots. Mentally retarded female patients with a full mutation showed FMRP expression in only some of their hair roots (< 55%), and no overlap with normal female controls was observed. The advantages of this test are (1) plucking of hair follicles does no appreciable harm to the mentally retarded patient, (2) hairs can be sent in a simple envelope to a diagnostic center, and (3) the result of the test is available within 5 h of plucking. In addition, this test enabled us to identify two fragile X patients who did not show the full mutation by analysis of DNA isolated from blood cells.. 10364521 21 39 fragile X syndrome SpecificDisease D005600 10364521 146 164 fragile X syndrome SpecificDisease D005600 10364521 507 525 fragile X syndrome SpecificDisease D005600 10364521 756 774 fragile X syndrome SpecificDisease D005600 10364521 986 995 fragile X Modifier D005600 10364521 1113 1122 fragile X Modifier D005600 10364521 1190 1207 Mentally retarded Modifier D008607 10364521 1462 1479 mentally retarded Modifier D008607 10364521 1674 1683 fragile X Modifier D005600 1384324|t|Pelizaeus-Merzbacher disease: detection of mutations Thr181----Pro and Leu223----Pro in the proteolipid protein gene, and prenatal diagnosis. 1384324|a|A family with an apparent history of X-linked Pelizaeus-Merzbacher disease presented for genetic counseling, requesting carrier detection and prenatal diagnosis. RFLP analysis using the proteolipid protein (PLP) gene probe was uninformative in this family. A prenatal diagnosis on a chorionic villus sample (CVS) was carried out using single-strand conformation polymorphism (SSCP) analysis of a variant in exon 4 of the PLP gene. The fetus was predicted to be unaffected. Sequencing of the exon from the CVS, the predicted-carrier mother, and the obligate-carrier grandmother revealed an A-to-C change at nucleotide 541 in the two women but not in the fetus. As this change results in a Thr-to-Pro change at amino acid 181 in a region of the gene predicted to be part of a transmembrane segment, it was concluded that this was the mutation causing the disease in this family. In addition, in a second family, an exon 5 variant band pattern on SSCP analysis was shown by sequencing to be due to a T-to-C change at nucleotide 668. This results in a Leu-to-Pro change in a carrier mother and in her two affected sons. These results provide further examples of mutations in PLP that cause Pelizaeus-Merzbacher disease and illustrate the value of SSCP in genetic analysis.. 1384324 0 28 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 1384324 188 216 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 1384324 1328 1356 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 2404853|t|A normal male with an inherited deletion of one exon within the DMD gene. 2404853|a|We describe two brothers with identical inherited deletions of one single exon within the middle of the DMD gene; one brother has Becker muscular dystrophy diagnosed at 11 years of age, whereas the older brother is normal at 18. These results have implications for genetic counselling and prenatal diagnosis in families with Becker muscular dystrophy.. 2404853 64 67 DMD Modifier D020388 2404853 178 181 DMD Modifier D020388 2404853 204 229 Becker muscular dystrophy SpecificDisease C537666 2404853 399 424 Becker muscular dystrophy SpecificDisease C537666 10406661|t|Small deletions in the type II collagen triple helix produce kniest dysplasia. 10406661|a|Kniest dysplasia is a moderately severe type II collagenopathy, characterized by short trunk and limbs, kyphoscoliosis, midface hypoplasia, severe myopia, and hearing loss. Mutations in the gene that encodes type II collagen (COL2A1), the predominant protein of cartilage, have been identified in a number of individuals with Kniest dysplasia. All but two of these previously described mutations cause in-frame deletions in type II collagen, either by small deletions in the gene or splice site alterations. Furthermore, all but one of these mutations is located between exons 12 and 24 in the COL2A1 gene. We used heteroduplex analysis to identify sequence anomalies in five individuals with Kniest dysplasia. Sequencing of the index patients genomic DNA identified four new dominant mutations in COL2A1 that result in Kniest dysplasia a 21-bp deletion in exon 16, an 18-bp deletion in exon 19, and 4-bp deletions in the splice donor sites of introns 14 and 20. A previously described 28-bp deletion at the COL2A1 exon 12-intron 12 junction, deleting the splice donor site, was identified in the fifth case. The latter three mutations are predicted to result in exon skipping in the mRNA encoded from the mutant allele. These data suggest that Kniest dysplasia results from shorter type II collagen monomers, and support the hypothesis that alteration of a specific COL2A1 domain, which may span from exons 12 to 24, leads to the Kniest dysplasia phenotype.. 10406661 61 77 kniest dysplasia SpecificDisease C537207 10406661 79 95 Kniest dysplasia SpecificDisease C537207 10406661 119 141 type II collagenopathy DiseaseClass C535964 10406661 183 197 kyphoscoliosis DiseaseClass OMIM:610170 10406661 199 217 midface hypoplasia SpecificDisease OMIM:300194 10406661 226 232 myopia DiseaseClass D009216 10406661 238 250 hearing loss DiseaseClass D034381 10406661 405 421 Kniest dysplasia SpecificDisease C537207 10406661 772 788 Kniest dysplasia SpecificDisease C537207 10406661 899 915 Kniest dysplasia SpecificDisease C537207 10406661 1325 1341 Kniest dysplasia SpecificDisease C537207 10406661 1511 1527 Kniest dysplasia Modifier C537207 8071957|t|Adenomatous polyposis coli and a cytogenetic deletion of chromosome 5 resulting from a maternal intrachromosomal insertion. 8071957|a|We present the clinical and laboratory findings in an institutionalised adult patient originally referred for autism. A high risk of colorectal cancer was predicted when an interstitial deletion of the long arm of chromosome 5, del (5) (q15q22. 3), was detected in her lymphocytes and deletion of the MCC and APC genes confirmed by molecular analysis. Adenomatous polyposis coli and carcinoma of the rectum were subsequently diagnosed in the patient. She was profoundly mentally retarded, autistic, and had minor dysmorphic features consistent with those of previous patients with similar deletions. The deletion arose as a result of recombination within the small insertion loop formed at meiosis by the direct insertion (dir ins (5) (q22. 3q14. 2q15)) found in the patients mother. This family further confirms the cytogenetic mapping of both MCC and APC genes to 5q22 and comparison with other recent cases suggests that both genes and their closely linked markers lie within the 5q22. 1 subband 8071957 0 26 Adenomatous polyposis coli SpecificDisease D011125 8071957 234 240 autism SpecificDisease D001321 8071957 257 274 colorectal cancer SpecificDisease D015179 8071957 433 436 APC Modifier D011125 8071957 476 502 Adenomatous polyposis coli SpecificDisease D011125 8071957 507 530 carcinoma of the rectum SpecificDisease D012004 8071957 594 611 mentally retarded Modifier D008607 8071957 613 621 autistic Modifier D001321 8071957 637 656 dysmorphic features SpecificDisease D000013 8071957 977 980 APC Modifier D011125 1338904|t|Somatic mutations of the APC gene in colorectal tumors: mutation cluster region in the APC gene. 1338904|a|We examined somatic mutations of the adenomatous polyposis coli (APC) gene in 63 colorectal tumors (16 adenomas and 47 carcinomas) developed in familial adenomatous polyposis (FAP) and non-FAP patients. In addition to loss of heterozygosity (LOH) at the APC locus in 30 tumors, 43 other somatic mutations were detected. Twenty-one of them were point mutations; 16 nonsense and two missense mutations, and three occurred in introns at the splicing site. Twenty-two tumors had frameshift mutations due to deletion or insertion; nineteen of them were deletions of one to 31 bp and three were a 1-bp insertion. One tumor had a 1-bp deletion in an intron near the splicing site. Hence, 41 (95%) of 43 mutations resulted in truncation of the APC protein. Over 60% of the somatic mutations in the APC gene were clustered within a small region of exon 15, designated as MCR (mutation cluster region), which accounted for less than 10% of the coding region. Combining these data and the results of LOH, more than 80% of tumors (14 adenomas and 39 carcinomas) had at least one mutation in the APC gene, of which more than 60% (9 adenomas and 23 carcinomas) had two mutations. These results strongly suggest that somatic mutations of the APC gene are associated with development of a great majority of colorectal tumors.. 1338904 25 28 APC Modifier D011125 1338904 37 54 colorectal tumors DiseaseClass D015179 1338904 87 90 APC Modifier D011125 1338904 134 160 adenomatous polyposis coli Modifier D011125 1338904 162 165 APC Modifier D011125 1338904 178 195 colorectal tumors DiseaseClass D015179 1338904 200 208 adenomas DiseaseClass D000236 1338904 216 226 carcinomas DiseaseClass D009369 1338904 241 271 familial adenomatous polyposis Modifier D011125 1338904 273 276 FAP Modifier D011125 1338904 351 354 APC Modifier D011125 1338904 367 373 tumors DiseaseClass D009369 1338904 561 567 tumors DiseaseClass D009369 1338904 708 713 tumor DiseaseClass D009369 1338904 833 836 APC Modifier D011125 1338904 887 890 APC Modifier D011125 1338904 1108 1114 tumors DiseaseClass D009369 1338904 1119 1127 adenomas DiseaseClass D000236 1338904 1135 1145 carcinomas DiseaseClass D009369 1338904 1180 1183 APC Modifier D011125 1338904 1216 1224 adenomas DiseaseClass D000236 1338904 1232 1242 carcinomas DiseaseClass D009369 1338904 1324 1327 APC Modifier D011125 1338904 1388 1405 colorectal tumors DiseaseClass D015179 3362213|t|Identification of an altered splice site in Ashkenazi Tay-Sachs disease. 3362213|a|Tay-Sachs disease is an autosomal recessive genetic disorder resulting from mutation of the HEXA gene encoding the alpha-subunit of the lysosomal enzyme, beta-N-acetylhexosaminidase A (ref. 1). A relatively high frequency of carriers (1/27) of a lethal, infantile form of the disease is found in the Ashkenazi Jewish population, but it is not yet evident whether this has resulted from a founder effect and random genetic drift or from a selective advantage of heterozygotes. We have identified a single-base mutation in a cloned fragment of the HEXA gene from an Ashkenazi Jewish patient. This change, the substitution of a C for G in the first nucleotide of intron 12 is expected to result in defective splicing of the messenger RNA. A test for the mutant allele based on amplification of DNA by the polymerase chain rection and cleavage of a DdeI restriction site generated by the mutation revealed that this case and two other cases of the Ashkenazi, infantile form of Tay-Sachs disease are heterozygous for two different mutations. The occurrence of multiple mutant alleles warrants further examination of the selective advantage hypothesis.. 3362213 54 71 Tay-Sachs disease SpecificDisease D013661 3362213 73 90 Tay-Sachs disease SpecificDisease D013661 3362213 97 133 autosomal recessive genetic disorder DiseaseClass D030342 3362213 1046 1063 Tay-Sachs disease SpecificDisease D013661 10484772|t|Coats' disease of the retina (unilateral retinal telangiectasis) caused by somatic mutation in the NDP gene: a role for norrin in retinal angiogenesis. 10484772|a|Coats disease is characterized by abnormal retinal vascular development (so-called retinal telangiectasis) which results in massive intraretinal and subretinal lipid accumulation (exudative retinal detachment). The classical form of Coats disease is almost invariably isolated, unilateral and seen in males. A female with a unilateral variant of Coats disease gave birth to a son affected by Norrie disease. Both carried a missense mutation within the NDP gene on chromosome Xp11. 2 2. Subsequently analysis of the retinas of nine enucleated eyes from males with Coats disease demonstrated in one a somatic mutation in the NDP gene which was not present within non-retinal tissue. We suggest that Coats telangiectasis is secondary to somatic mutation in the NDP gene which results in a deficiency of norrin (the protein product of the NDP gene) within the developing retina. This supports recent observations that the protein is critical for normal retinal vasculogenesis. 10484772 0 14 Coats' disease SpecificDisease D058456 10484772 30 63 unilateral retinal telangiectasis SpecificDisease D058456 10484772 152 165 Coats disease SpecificDisease D058456 10484772 186 223 abnormal retinal vascular development DiseaseClass D058456 10484772 235 257 retinal telangiectasis SpecificDisease D058456 10484772 284 330 intraretinal and subretinal lipid accumulation CompositeMention D006949 10484772 332 360 exudative retinal detachment DiseaseClass D012163 10484772 385 398 Coats disease SpecificDisease D058456 10484772 498 511 Coats disease SpecificDisease D058456 10484772 544 558 Norrie disease SpecificDisease C537849 10484772 715 728 Coats disease SpecificDisease D058456 10484772 849 869 Coats telangiectasis SpecificDisease D058456 10484772 938 958 deficiency of norrin DiseaseClass C537849 1357962|t|Trisomy 15 with loss of the paternal 15 as a cause of Prader-Willi syndrome due to maternal disomy. 1357962|a|Uniparental disomy has recently been recognized to cause human disorders, including Prader-Willi syndrome (PWS). We describe a particularly instructive case which raises important issues concerning the mechanisms producing uniparental disomy and whose evaluation provides evidence that trisomy may precede uniparental disomy in a fetus. Chorionic villus sampling performed for advanced maternal age revealed trisomy 15 in all direct and cultured cells, though the fetus appeared normal. Chromosome analysis of amniocytes obtained at 15 wk was normal in over 100 cells studied. The child was hypotonic at birth, and high-resolution banding failed to reveal the deletion of 15q11-13, a deletion which is found in 50% -70% of patients with PWS. Over time, typical features of PWS developed. Molecular genetic analysis using probes for chromosome 15 revealed maternal disomy. Maternal nondisjunction with fertilization of a disomic egg by a normal sperm, followed by loss of the paternal 15, is a likely cause of confined placental mosaicism and uniparental disomy in this case of PWS, and advanced maternal age may be a predisposing factor.. 1357962 0 10 Trisomy 15 SpecificDisease C538037 1357962 54 75 Prader-Willi syndrome SpecificDisease D011218 1357962 83 98 maternal disomy SpecificDisease D024182 1357962 100 118 Uniparental disomy SpecificDisease D024182 1357962 184 205 Prader-Willi syndrome SpecificDisease D011218 1357962 207 210 PWS SpecificDisease D011218 1357962 323 341 uniparental disomy SpecificDisease D024182 1357962 406 424 uniparental disomy SpecificDisease D024182 1357962 508 518 trisomy 15 SpecificDisease C538037 1357962 691 700 hypotonic SpecificDisease D009123 1357962 837 840 PWS SpecificDisease D011218 1357962 873 876 PWS SpecificDisease D011218 1357962 1142 1160 uniparental disomy SpecificDisease D024182 1357962 1177 1180 PWS SpecificDisease D011218 492335|t|Genetic defect in secretion of complement C5 in mice. 492335|a|A genetic deficiency of the fifth (C5) component of complement1-3, a serum glycoprotein of molecular weight (MW) 220, 000 (ref. 4), has been found in 39% of inbred strains of mice3. Sera of deficient mice lack detectable C5 activity and protein2, 3. In addition deficient mice produce antibody to mouse C5 when injected with sera from C5 sufficient (normal) strains. Levy et al. 5 showed that somatic cell hybrids between C5 deficient (B10. D2/old line) macrophages and either C5 sufficient (B10. D2/new line) mouse kidney or chicken erythroblasts secreted haemolytically active mouse C5 in vitro. Several possible molecular mechanisms to account for the findings were considered, but insufficient direct data were available to choose among them. We recently reported that mouse (CD. 1 strain) peritoneal cells in culture synthesise and secrete a single chain precursor, pro-C5 (MW approximately 210, 000), of the two-chain (alpha chain, 125, 000 and beta chain 83, 000 MW) C5 protein6. Radiolabelled precursor C5 was contained within the cells and was secreted into the tissue culture media. Using similar methods, we now find that C5 deficiency in each of five different mouse strains (AKR, SWR, DBA/2J8 A/HeJ and B10. D2/old line) is due to a failure in secretion of C5 protein and not to a failure in biosynthesis of pro-C5 492335 8 44 defect in secretion of complement C5 SpecificDisease OMIM:609536 492335 64 119 deficiency of the fifth (C5) component of complement1-3 SpecificDisease OMIM:609536 492335 1187 1200 C5 deficiency SpecificDisease OMIM:609536 8528199|t|WASP gene mutations in Wiskott-Aldrich syndrome and X-linked thrombocytopenia. 8528199|a|The WASP gene has been recently cloned from Xp11. 23 and shown to be mutated in three patients with the Wiskott-Aldrich syndrome (WAS). We have developed a screening protocol for identifying WASP gene alterations in genomic DNA and have identified a spectrum of novel mutations in 12 additional unrelated families. These missense, nonsense and frameshift mutations involve eight of the 12 exons of the gene. Two mutations creating premature termination codons were associated with lack of detectable mRNA on Northern blots. Four amino acid substitutions, Leu27Phe, Thr48Ile, Val75Met and Arg477Lys, were found in patients with congenital thrombocytopenia and no clinically evident immune defect indicating that the WASP gene is the site for mutations in X-linked thrombocytopenia as well as in WAS. A T-cell line from a WAS patient contained two independent DNA alterations, a constitutional frameshift mutation, also present in peripheral blood leukocytes from the patient, and compensatory splice site mutation unique to the cell line. The distribution of eight missense mutations provides valuable information on amino acids which are essential for normal protein function, and suggests that sites in the first two exons are hot-spots for mutation. 8528199 23 47 Wiskott-Aldrich syndrome SpecificDisease D014923 8528199 52 77 X-linked thrombocytopenia SpecificDisease OMIM:313900 8528199 183 207 Wiskott-Aldrich syndrome SpecificDisease D014923 8528199 209 212 WAS SpecificDisease D014923 8528199 706 733 congenital thrombocytopenia SpecificDisease OMIM:313900 8528199 833 858 X-linked thrombocytopenia SpecificDisease OMIM:313900 8528199 873 876 WAS SpecificDisease D014923 8528199 899 902 WAS Modifier D014923 1307253|t|Detection of a nonsense mutation in the dystrophin gene by multiple SSCP. 1307253|a|A combination of multiplex PCR with the single strand conformation polymorphism (SSCP) technique was employed to screen for point mutations in the human dystrophin gene. Co-amplification of 11 exons from genomic DNA of Duchenne and Becker muscular dystrophy (DMD/BMD) patients with no deletion or duplication was performed and the samples subjected to multiple SSCP analysis. We report the case of a nonsense mutation in a Duchenne patient identified by this approach. The mutation introduces a termination codon within exon 8 of the dystrophin gene. It is predicted to cause a very premature translational termination accounting for the severe phenotype observed. The patient inherited this mutation from his mother. In addition the analysis revealed 5 polymorphisms useful for internal control.. 1307253 293 331 Duchenne and Becker muscular dystrophy Modifier D020388|C537666 1307253 333 336 DMD Modifier D020388 1307253 337 340 BMD Modifier C537666 1307253 497 505 Duchenne Modifier D020388 8346255|t|Molecular mechanisms of oncogenic mutations in tumors from patients with bilateral and unilateral retinoblastoma. 8346255|a|The RB1 gene from 12 human retinoblastoma tumors has been analyzed exon-by-exon with the single-strand conformation polymorphism technique. Mutations were found in all tumors, and one-third of the tumors had independent mutations in both alleles neither of which were found in the germ line, confirming their true sporadic nature. In the remaining two-thirds of the tumors only one mutation was found, consistent with the loss-of-heterozygosity theory of tumorigenesis. Point mutations, the majority of which were C-- > T transitions, were the most common abnormality and usually resulted in the conversion of an arginine codon to a stop codon. Small deletions were the second most common abnormality and most often created a downstream stop codon as the result of a reading frameshift. Deletions and point mutations also affected splice junctions. Direct repeats were present at the breakpoint junctions in the majority of deletions, supporting a slipped-mispairing mechanism. Point mutations generally produced DNA sequences which resulted in perfect homology with endogenous sequences which lay within 14 bp.. 8346255 47 53 tumors DiseaseClass D009369 8346255 73 112 bilateral and unilateral retinoblastoma CompositeMention D012175 8346255 141 162 retinoblastoma tumors SpecificDisease D012175 8346255 282 288 tumors DiseaseClass D009369 8346255 311 317 tumors DiseaseClass D009369 8346255 480 486 tumors DiseaseClass D009369 7951315|t|PAX6 gene dosage effect in a family with congenital cataracts, aniridia, anophthalmia and central nervous system defects. 7951315|a|The human eye malformation aniridia results from haploinsufficiency of PAX6, a paired box DNA-binding protein. To study this dosage effect, we characterized two PAX6 mutations in a family segregating aniridia and a milder syndrome consisting of congenital cataracts and late onset corneal dystrophy. The nonsense mutations, at codons 103 and 353, truncate PAX6 within the N-terminal paired and C-terminal PST domains, respectively. The wild-type PST domain activates transcription autonomously and the mutant form has partial activity. A compound heterozygote had severe craniofacial and central nervous system defects and no eyes. The pattern of malformations is similar to that in homozygous Sey mice and suggests a critical role for PAX6 in controlling the migration and differentiation of specific neuronal progenitor cells in the brain.. 7951315 41 61 congenital cataracts SpecificDisease D002386 7951315 63 71 aniridia SpecificDisease D015783 7951315 73 85 anophthalmia SpecificDisease D000853 7951315 90 120 central nervous system defects DiseaseClass D009421 7951315 149 157 aniridia SpecificDisease D015783 7951315 171 197 haploinsufficiency of PAX6 SpecificDisease OMIM:106210 7951315 322 330 aniridia SpecificDisease D015783 7951315 367 387 congenital cataracts SpecificDisease D002386 7951315 403 420 corneal dystrophy SpecificDisease D003317 7951315 693 740 craniofacial and central nervous system defects CompositeMention D019465|D009421 7951315 745 752 no eyes DiseaseClass D000853 409732|t|Hereditary C7 deficiency. Diagnosis and HLA studies in a French-Canadian family. 409732|a|The serum of a 44-yr-old woman of French-Canadian descent having a B-27 positive ankylosing spondylitis was deficient in the seventh component of complement (C7) as determined by hemolytic and immunochemical methods. No inhibitor against C7 was detected, and the levels of all other complement components were normal. No deficiency in the opsonic activity of the serum was found, and the results of basic coagulation studies of the plasma were normal. On investigation of the patients family, two sisters were found to have the same deficiency but were otherwise in good health. The seven other siblings were heterozygous for C7 deficiency, while the paternal aunt had a normal C7 level. In the third generation, six children of the three homozygous sisters and five children of heterozygotes were available for testing. Studies of the HLA antigens in all the 22 subjects and in three spouses indicated no close linkage between the CM deficienty and the HLA system. In addition, the simultaneous occurrence of two hereditary complement deficiencies (C2 and C7) was discovered in one family of this remarkable kindred.. 409732 0 24 Hereditary C7 deficiency SpecificDisease OMIM:610102 409732 162 184 ankylosing spondylitis SpecificDisease D013167 409732 189 237 deficient in the seventh component of complement SpecificDisease OMIM:610102 409732 707 720 C7 deficiency SpecificDisease OMIM:610102 409732 1013 1026 CM deficienty SpecificDisease OMIM:610102 409732 1106 1141 complement deficiencies (C2 and C7) CompositeMention OMIM:610102|OMIM:217000 10598815|t|Alstrom syndrome: further evidence for linkage to human chromosome 2p13. 10598815|a|Alstrom syndrome is a rare autosomal recessive disorder characterized by retinal degeneration, sensorineural hearing loss, early-onset obesity, and non-insulin-dependent diabetes mellitus. The gene for Alstrom syndrome (ALMS1) has been previously localized to human chromosome 2p13 by homozygosity mapping in two distinct isolated populations - French Acadian and North African. Pair-wise analyses resulted in maximum lod (logarithm of the odds ratio) scores of 3. 84 and 2. 9, respectively. To confirm these findings, a large linkage study was performed in twelve additional families segregating for Alstrom syndrome. A maximum two-point lod score of 7. 13 (theta = 0. 00) for marker D2S2110 and a maximum cumulative multipoint lod score of 9. 16 for marker D2S2110 were observed, further supporting linkage to chromosome 2p13. No evidence of genetic heterogeneity was observed in these families. Meiotic recombination events have localized the critical region containing ALMS1 to a 6. 1-cM interval flanked by markers D2S327 and D2S286. A fine resolution radiation hybrid map of 31 genes and markers has been constructed. 10598815 0 16 Alstrom syndrome SpecificDisease D056769 10598815 73 89 Alstrom syndrome SpecificDisease D056769 10598815 100 128 autosomal recessive disorder DiseaseClass D030342 10598815 146 166 retinal degeneration SpecificDisease D012162 10598815 168 194 sensorineural hearing loss SpecificDisease D006319 10598815 208 215 obesity SpecificDisease D009765 10598815 221 260 non-insulin-dependent diabetes mellitus SpecificDisease D003924 10598815 275 291 Alstrom syndrome SpecificDisease D056769 10598815 674 690 Alstrom syndrome SpecificDisease D056769 2575071|t|Haplotype and multipoint linkage analysis in Finnish choroideremia families. 2575071|a|Multipoint linkage analysis of choroideremia (TCD) and seven X chromosomal restriction fragment length polymorphisms (RFLPs) was carried out in 18 Finnish TCD families. The data place TCD distal to PGK and DXS72, very close to DXYS1 and DXYS5 (Zmax = 24 at theta = 0) and proximal to DXYS4 and DXYS12. This agrees with the data obtained from other linkage studies and from physical mapping. All the TCD males and carrier females studied have the same DXYS1 allele in coupling with TCD. In Northeastern Finland, 66/69 chromosomes carrying TCD had the same haplotype at loci DXS72, DXYS1, DXYS4, and DXYS12. The same haplotype is seen in only 15/99 chromosomes not carrying TCD. Moreover, in 71/104 non-TCD chromosomes, the haplotype at six marker loci is different from those seen in any of the 76 TCD chromosomes. This supports the previously described hypothesis that the large Northern Finnish choroideremia pedigrees, comprising a total of over 80 living patients representing more than a fifth of all TCD patients described worldwide, carry the same mutation. These linkage and haplotype data provide improved opportunities for prenatal diagnosis based on RFLP studies.. 2575071 53 66 choroideremia Modifier D015794 2575071 108 121 choroideremia SpecificDisease D015794 2575071 123 126 TCD SpecificDisease D015794 2575071 232 235 TCD Modifier D015794 2575071 261 264 TCD SpecificDisease D015794 2575071 476 479 TCD Modifier D015794 2575071 558 561 TCD SpecificDisease D015794 2575071 615 618 TCD SpecificDisease D015794 2575071 749 752 TCD SpecificDisease D015794 2575071 874 877 TCD Modifier D015794 2575071 973 986 choroideremia Modifier D015794 2575071 1082 1085 TCD Modifier D015794 2253938|t|Genetic heterogeneity at the glucose-6-phosphate dehydrogenase locus in southern Italy: a study on a population from the Matera district. 2253938|a|Glucose-6-phosphate dehydrogenase (G6PD) has been analyzed by gel electrophoresis and by quantitative assay in an unselected sample of 1524 schoolboys from the province of Matera (Lucania) in southern Italy. We have identified 43 subjects with a G6PD variant. Of these, 31 had severe G6PD deficiency, nine had mild to moderate deficiency, and three had a non-deficient electrophoretic variant. The overall rate of G6PD deficiency was 2. 6%. The frequency of G6PD deficiency, ranging from 7. 2% on the Ionian Coast to zero on the eastern side of the Lucanian Apennines, appears to be inversely related to the distance of each town examined from the Ionian Coast, suggesting that this geographic distribution may reflect, at least in part, gene flow from Greek settlers. Biochemical characterization has shown that most of the G6PD deficiency in this population is accounted for by G6PD Mediterranean. In addition, we have found several examples of two other known polymorphic variants (G6PD Cagliari and G6PD A-); three new polymorphic variants, G6PD Metaponto (class III), G6PD Montalbano (class III), and G6PD Pisticci (class IV); and two sporadic variants, G6PD Tursi (class III) and G6PD Ferrandina (class II). These data provide further evidence for the marked genetic heterogeneity of G6PD deficiency within a relatively narrow geographic area and they prove the presence in the Italian peninsula of a gene (GdA-) regarded as characteristically African. 2253938 422 437 G6PD deficiency SpecificDisease D005955 2253938 552 567 G6PD deficiency SpecificDisease D005955 2253938 596 611 G6PD deficiency SpecificDisease D005955 2253938 963 978 G6PD deficiency SpecificDisease D005955 2253938 1428 1443 G6PD deficiency SpecificDisease D005955 10411929|t|Defective CTLA-4 cycling pathway in Chediak-Higashi syndrome: a possible mechanism for deregulation of T lymphocyte activation. 10411929|a|Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4, also known as CD152) has been shown to play a major role in the regulation of T cell activation. Its membrane expression is highly regulated by endocytosis and trafficking through the secretory lysosome pathway. Chediak-Higashi syndrome (CHS) is an inherited disorder caused by mutations in the lysosomal trafficking regulator gene, LYST. It results in defective membrane targeting of the proteins present in secretory lysosomes, and it is associated with a variety of features, including a lymphoproliferative syndrome with hemophagocytosis. The murine equivalent of CHS, beige mice, present similar characteristics but do not develop the lymphoproliferative syndrome. We show herein that CTLA-4 is present in enlarged, abnormal vesicles in CHS T cells and is not properly expressed at the cell surface after T cell activation, whereas its surface expression is not impaired. It is therefore proposed that the defective surface expression of CTLA-4 by CHS T cells is involved in the generation of lymphoproliferative disease. This observation may provide insight into the role of CTLA-4 in humans.. 10411929 36 60 Chediak-Higashi syndrome SpecificDisease D002609 10411929 393 417 Chediak-Higashi syndrome SpecificDisease D002609 10411929 419 422 CHS SpecificDisease D002609 10411929 430 448 inherited disorder DiseaseClass D030342 10411929 672 700 lymphoproliferative syndrome SpecificDisease D008232 10411929 706 722 hemophagocytosis DiseaseClass D051359 10411929 749 752 CHS SpecificDisease D002609 10411929 821 849 lymphoproliferative syndrome SpecificDisease D008232 10411929 923 926 CHS Modifier D002609 10411929 1134 1137 CHS Modifier D002609 10411929 1179 1206 lymphoproliferative disease SpecificDisease D008232 10398436|t|Beta-catenin accumulation and mutation of the CTNNB1 gene in hepatoblastoma. 10398436|a|Hepatoblastoma is a rare malignant tumor of the liver that occurs in children at an average age of 2 to 3 years. Epidemiologic studies have shown an increased frequency of this tumor type in families affected by adenomatous polyposis coli. In addition to the epidemiologic data, molecular genetic studies suggest that inactivation of the APC tumor suppressor may be involved in hepatoblastoma tumorigenesis. A major function of APC is the downregulation of beta-catenin, a transcription-activating protein with oncogenic potential. In an ongoing immunohistochemical study of beta-catenin expression in sporadic cases of tumor types that are associated with adenomatous polyposis coli, we observed increased beta-catenin levels in the cytoplasm and in the nuclei of three investigated hepatoblastomas. Sequencing of exon 3 of the beta-catenin gene (CTNNB1) revealed an activating mutation in one of the tumor samples. Our data indicate for the first time that beta-catenin accumulation may play a role in the development of hepatoblastoma and that activating mutations of the beta-catenin gene may substitute biallelic APC inactivation in this tumor type. Genes Chromosomes Cancer 25 399-402, 1999.. 10398436 61 75 hepatoblastoma SpecificDisease D018197 10398436 77 91 Hepatoblastoma SpecificDisease D018197 10398436 102 130 malignant tumor of the liver DiseaseClass D008113+D018198 10398436 254 259 tumor Modifier D009369 10398436 289 315 adenomatous polyposis coli SpecificDisease D011125 10398436 415 424 APC tumor Modifier D011125 10398436 455 469 hepatoblastoma Modifier D018197 10398436 697 702 tumor Modifier D009369 10398436 734 760 adenomatous polyposis coli SpecificDisease D011125 10398436 861 876 hepatoblastomas SpecificDisease D018197 10398436 979 984 tumor Modifier D009369 10398436 1100 1114 hepatoblastoma SpecificDisease D018197 10398436 1220 1225 tumor Modifier D009369 1833974|t|Sequence of DNA flanking the exons of the HEXA gene, and identification of mutations in Tay-Sachs disease. 1833974|a|The rapid identification of mutations causing Tay-Sachs disease requires the capacity to readily screen the regions of the HEXA gene most likely to be affected by mutation. We have sequenced the portions of the introns flanking each of the 14 HEXA exons in order to specify oligonucleotide primers for the PCR-dependent amplification of each exon and splice-junction sequence. The amplified products were analyzed, by electrophoresis in nondenaturing polyacrylamide gels, for the presence of either heteroduplexes, derived from the annealing of normal and mutant DNA strands, or single-strand conformational polymorphisms (SSCP), derived from the renaturation of single-stranded DNA. Five novel mutations from Tay-Sachs disease patients were detected a 5-bp deletion of TCTCC in IVS-9; a 2-bp deletion of TG in exon 5; G78 to A, giving a stop codon in exon 1; G533 to T in exon 5, producing the third amino acid substitution detected at this site; and G to C at position 1 of IVS-2, expected to produce abnormal splicing. In addition, two mutations, (G1496 to A in exon 13 and a 4-bp insertion in exon 11) that have previously been reported were identified.. 1833974 88 105 Tay-Sachs disease SpecificDisease D013661 1833974 153 170 Tay-Sachs disease SpecificDisease D013661 1833974 817 834 Tay-Sachs disease Modifier D013661 10742101|t|In vivo modulation of Hmgic reduces obesity. 10742101|a|The HMGI family of proteins consists of three members, HMGIC, HMGI and HMGI (Y), that function as architectural factors and are essential components of the enhancesome. HMGIC is predominantly expressed in proliferating, undifferentiated mesenchymal cells and is not detected in adult tissues. It is disrupted and misexpressed in a number of mesenchymal tumour cell types, including fat-cell tumours (lipomas). In addition Hmgic-/- mice have a deficiency in fat tissue. To study its role in adipogenesis and obesity, we examined Hmgic expression in the adipose tissue of adult, obese mice. Mice with a partial or complete deficiency of Hmgic resisted diet-induced obesity. Disruption of Hmgic caused a reduction in the obesity induced by leptin deficiency (Lepob/Lepob) in a gene-dose-dependent manner. Our studies implicate a role for HMGIC in fat-cell proliferation, indicating that it may be an adipose-specific target for the treatment of obesity.. 10742101 36 43 obesity SpecificDisease D009765 10742101 386 404 mesenchymal tumour Modifier D008637 10742101 427 443 fat-cell tumours DiseaseClass D018205 10742101 445 452 lipomas DiseaseClass D008067 10742101 552 559 obesity SpecificDisease D009765 10742101 622 627 obese Modifier D009765 10742101 646 685 partial or complete deficiency of Hmgic CompositeMention OMIM:600698 10742101 708 715 obesity SpecificDisease D009765 10742101 763 770 obesity SpecificDisease D009765 10742101 782 799 leptin deficiency SpecificDisease OMIM:164160 10742101 987 994 obesity SpecificDisease D009765 10210128|t|Prenatal diagnosis by FISH in a family with Pelizaeus-Merzbacher disease caused by duplication of PLP gene. 10210128|a|A diagnosis of Pelizaeus-Merzbacher disease (MIM 312080) was made in a young boy. No mutation in the coding region of the proteolipid protein (PLP) gene had been found. The boys maternal aunt came for prenatal diagnosis when 16 + weeks pregnant and carrying a male fetus. Samples were tested for duplication of the PLP gene, by interphase FISH, in lymphocyte preparations from the proband, his aunt and an amniotic fluid cell preparation from the fetus. The proband was found to carry the duplication, thus confirming the diagnosis of Pelizaeus Merzbacher disease, but neither the aunt nor the fetus carried a duplication.. 10210128 44 72 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 10210128 123 151 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 10210128 643 671 Pelizaeus Merzbacher disease SpecificDisease OMIM:312080 1056013|t|Somatic rearrangement of chromosome 14 in human lymphocytes. 1056013|a|Ataxia-telangiectasia is a rare genetic disorder associated with immune deficiency, chromosome instability, and a predisposition to lymphoid malignancy. We have detected chromosomally anomalous clones of lymphocytes in eight patients with this disorder. Chromosome banding disclosed that the clones are consistently marked by structural rearrangement of the long arm (q) of chromosome 14. A translocation involving 14q was found in clones obtained from seven of the eight patients whereas a ring 14 chromosome was found in a clone obtained from the other. These findings as well as data obtained by others for patients with ataxia-telangiectasia suggest that structural rearrangement of 14q is the initial chromosomal change in lymphocyte clones of patients with this disorder. Chromosomes of lymphocytes from one of the patients were studied before and after the onset of chronic lymphocytic leukemia. Before leukemia was diagnosed, the patient had a lymphocyte clone with a 14q translocation. This clone appears to have given rise to the leukemic cells. We hypothesize that structural rearrangement of 14q is directly related to abnormal growth of lymphocytes and that it may be a step toward the development of lymphoid malignancies. Increasing evidence, provided by others, for the nonrandom involvement of 14q in African-type Burkitts lymphoma and other lymphoid neoplasms further strengthens this hypothesis.. 1056013 61 82 Ataxia-telangiectasia SpecificDisease D001260 1056013 93 109 genetic disorder DiseaseClass D030342 1056013 126 143 immune deficiency DiseaseClass D007153 1056013 145 167 chromosome instability DiseaseClass D043171 1056013 193 212 lymphoid malignancy SpecificDisease D008223 1056013 685 706 ataxia-telangiectasia SpecificDisease D001260 1056013 934 962 chronic lymphocytic leukemia SpecificDisease D015451 1056013 971 979 leukemia DiseaseClass D007938 1056013 1101 1109 leukemic Modifier D007938 1056013 1275 1296 lymphoid malignancies SpecificDisease D008223 1056013 1392 1409 Burkitts lymphoma SpecificDisease D002051 1056013 1420 1438 lymphoid neoplasms DiseaseClass D008223 1301200|t|Two missense mutations causing mild hyperphenylalaninemia associated with DNA haplotype 12. 1301200|a|The genetic defects responsible for most phenylketonuria (PKU) and hyperphenylalaninemia (HPA) cases are located in the phenylalanine hydroxylase (PAH) gene. Approximately 50-60 mutations have been reported in Caucasians and are reflected in a wide range of clinical severities. Most mutations are linked to specific haplotypes, as defined by eight polymorphic restriction sites in the PAH gene. We hypothesized that there is at least one mild mutation linked to haplotype 12 in the Swedish PKU/HPA population, since 7 of 8 patients carrying haplotype 12 had mild HPA. Sequence analysis revealed a C-to-G transversion at the second base of codon 322, resulting in a substitution of glycine for alanine, in four mutant haplotype 12 genes, and a G-to-A transition at the second base of codon 408, resulting in a substitution of glutamine for arginine, in another three mutant haplotype 12 genes. These mutations segregated with mutant haplotype 12 alleles in nuclear families but were not present on normal or other mutant alleles. Both mutations were tested in a eukaryotic expression system in which enzyme activities of different mutant PAH enzymes reflect the relative severities of the mutations, although these in vitro activities cannot be translated directly into in vivo hepatic activities. The A322G mutant PAH had about 75% and the R408Q mutant PAH about 55% of the wild-type PAH enzyme activity. These in vitro activities are the highest reported for mutant PAH enzymes produced in the same expression system. (ABSTRACT TRUNCATED AT 250 WORDS). 1301200 36 57 hyperphenylalaninemia DiseaseClass D010661 1301200 96 111 genetic defects DiseaseClass D030342 1301200 133 148 phenylketonuria SpecificDisease D010661 1301200 150 153 PKU SpecificDisease D010661 1301200 159 180 hyperphenylalaninemia DiseaseClass D010661 1301200 182 185 HPA DiseaseClass D010661 1301200 583 586 PKU Modifier D010661 1301200 587 590 HPA Modifier D010661 1301200 656 659 HPA DiseaseClass D010661 7959759|t|Genomic organization of the adrenoleukodystrophy gene. 7959759|a|Adrenoleukodystrophy (ALD), the most frequent peroxisomal disorder, is a severe neurodegenerative disease associated with an impairment of very long chain fatty acids beta-oxidation. We have recently identified by positional cloning the gene responsible for ALD, located in Xq28. It encodes a new member of the " ABC " superfamily of membrane-associated transporters that shows, in particular, significant homology to the 70-kDa peroxisomal membrane protein (PMP70). We report here a detailed characterization of the ALD gene structure. It extends over 21 kb and consists of 10 exons. To facilitate the detection of mutations in ALD patients, we have determined the intronic sequences flanking the exons as well as the sequence of the 3 untranslated region and of the immediate 5 promoter region. Sequences present in distal exons cross-hybridize strongly to additional sequences in the human genome. The ALD gene has been positioned on a pulsed-field map between DXS15 and the L1CAM gene, about 650 kb upstream from the color pigment genes. The frequent occurrence of color vision anomalies observed in patients with adrenomyeloneuropathy (the adult onset form of ALD) thus does not represent a contiguous gene syndrome but a secondary manifestation of ALD.. 7959759 28 48 adrenoleukodystrophy Modifier D000326 7959759 55 75 Adrenoleukodystrophy SpecificDisease D000326 7959759 77 80 ALD SpecificDisease D000326 7959759 101 121 peroxisomal disorder DiseaseClass D018901 7959759 135 160 neurodegenerative disease DiseaseClass D019636 7959759 180 236 impairment of very long chain fatty acids beta-oxidation DiseaseClass OMIM:300100 7959759 313 316 ALD SpecificDisease D000326 7959759 572 575 ALD Modifier D000326 7959759 684 687 ALD Modifier D000326 7959759 960 963 ALD Modifier D000326 7959759 1173 1194 adrenomyeloneuropathy SpecificDisease D000326 7959759 1220 1223 ALD SpecificDisease D000326 7959759 1251 1275 contiguous gene syndrome DiseaseClass D025063 7959759 1309 1312 ALD SpecificDisease D000326 10220405|t|BRCA1 interacts with components of the histone deacetylase complex. 10220405|a|Germ-line mutations in the BRCA1 tumor-suppressor gene are associated with an increased susceptibility to breast and ovarian cancer. BRCA1 contains a carboxyl-terminal domain (BRCT) that is shared with several other proteins involved in maintaining genome integrity. In an effort to understand the function of BRCA1, we sought to isolate proteins that interact with the BRCT domain. Purified BRCT polypeptide was used as a probe to screen a human placenta cDNA expression library by Far Western analysis. Here we report that BRCA1 interacts in vivo and in vitro with the Rb-binding proteins, RbAp46 and RbAp48, as well as with Rb. Moreover, the BRCT domain associates with the histone deacetylases HDAC1 and HDAC2. These results demonstrate that BRCA1 interacts with components of the histone deacetylase complex, and therefore may explain the involvement of BRCA1 in multiple processes such as transcription, DNA repair, and recombination.. 10220405 174 199 breast and ovarian cancer CompositeMention D061325 1279971|t|Deletion of the KIT and PDGFRA genes in a patient with piebaldism. 1279971|a|We have previously shown that human piebaldism results from mutations of the KIT gene, which encodes the receptor for the mast/stem cell growth factor and is located in chromosome segment 4q12. Using DNA of a patient with piebaldism, mental retardation, and multiple congenital anomalies associated with a 46, XY, del (4) (q12q21. 1) karyotype, we carried out quantitative Southern blot hybridization analyses of the KIT gene and the adjacent PDGFRA (platelet-derived growth factor receptor alpha subunit) genes. The patient was hemizygous for both the KIT and PDGFRA genes, indicating that both of these genes are included within the deleted region. Therefore, deletion of the KIT and PDGFRA genes may account for the piebald phenotype in this patient. 1279971 55 65 piebaldism SpecificDisease D016116 1279971 103 113 piebaldism SpecificDisease D016116 1279971 289 299 piebaldism SpecificDisease D016116 1279971 301 319 mental retardation DiseaseClass D008607 1279971 325 354 multiple congenital anomalies DiseaseClass D000013 1279971 786 793 piebald Modifier D016116 3198117|t|Glucose-6-phosphate dehydrogenase variants and their frequency in Guangdong, China. 3198117|a|Erythrocyte glucose-6-phosphate dehydrogenase (G6PD) was characterized in blood samples obtained from 97 randomly selected males with enzyme deficiency from various regions of Guangdong Province, China. Nine new variants (Gd Kaiping, Gd Boluo, Gd Huiyang, Gd Gaomin, Gd Qing-Baijiang, Gd Gaozhou, Gd Huazhou, Gd Nanhai, and Gd Guangzhou) were identified. Of the 31 variants found in this province, Gd Kaiping, Gd Taiwan-Hakka, Gd Haad Yai, Gd Haad Yai-like and Gd Huiyang occurred most frequently. The frequency of each variant was calculated. The results demonstrated that the genetic heterogeneity of G6PD deficiency was high in this area.. 3198117 218 235 enzyme deficiency DiseaseClass D008661 3198117 687 702 G6PD deficiency SpecificDisease D005955 10369860|t|A common molecular basis for rearrangement disorders on chromosome 22q11. 10369860|a|The chromosome 22q11 region is susceptible to rearrangements that are associated with congenital anomaly disorders and malignant tumors. Three congenital anomaly disorders, cat-eye syndrome, der () syndrome and velo-cardio-facial syndrome/DiGeorge syndrome (VCFS/DGS) are associated with tetrasomy, trisomy or monosomy, respectively, for part of chromosome 22q11. VCFS/DGS is the most common syndrome associated with 22q11 rearrangements. In order to determine whether there are particular regions on 22q11 that are prone to rearrangements, the deletion end-points in a large number of VCFS/DGS patients were defined by haplotype analysis. Most VCFS/DGS patients have a similar 3 Mb deletion, some have a nested distal deletion breakpoint resulting in a 1. 5 Mb deletion and a few rare patients have unique deletions or translocations. The high prevalence of the disorder in the population and the fact that most cases occur sporadically suggest that sequences at or near the breakpoints confer susceptibility to chromosome rearrangements. To investigate this hypothesis, we developed hamster-human somatic hybrid cell lines from VCFS/DGS patients with all three classes of deletions and we now show that the breakpoints occur within similar low copy repeats, termed LCR22s. To support this idea further, we identified a family that carries an interstitial duplication of the same 3 Mb region that is deleted in VCFS/DGS patients. We present models to explain how the LCR22s can mediate different homologous recombination events, thereby generating a number of rearrangements that are associated with congenital anomaly disorders. We identified five additional copies of the LCR22 on 22q11 that may mediate other rearrangements leading to disease. 10369860 29 52 rearrangement disorders DiseaseClass D002869 10369860 160 188 congenital anomaly disorders DiseaseClass D000013 10369860 193 209 malignant tumors DiseaseClass D018198 10369860 217 245 congenital anomaly disorders DiseaseClass D000013 10369860 247 263 cat-eye syndrome SpecificDisease C535918|OMIM:115470 10369860 265 280 der () syndrome SpecificDisease C535733 10369860 285 312 velo-cardio-facial syndrome SpecificDisease D004062 10369860 313 330 DiGeorge syndrome SpecificDisease D004062 10369860 332 336 VCFS SpecificDisease D004062 10369860 337 340 DGS SpecificDisease D004062 10369860 438 442 VCFS SpecificDisease D004062 10369860 443 446 DGS SpecificDisease D004062 10369860 660 664 VCFS Modifier D004062 10369860 665 668 DGS Modifier D004062 10369860 719 723 VCFS Modifier D004062 10369860 724 727 DGS Modifier D004062 10369860 1204 1208 VCFS Modifier D004062 10369860 1209 1212 DGS Modifier D004062 10369860 1486 1490 VCFS Modifier D004062 10369860 1491 1494 DGS Modifier D004062 10369860 1675 1703 congenital anomaly disorders DiseaseClass D000013 1424237|t|Typical and partial cat eye syndrome: identification of the marker chromosome by FISH. 1424237|a|Three children are reported with typical cat eye syndrome (CES) and three more children with partial CES because of absence of coloboma, in which the supernumerary marker chromosome was studied by FISH. Using a genomic library, and also a centromeric and particularly a cosmid probe of 22q11, partial tetrasomy was shown in all cases.. 1424237 20 36 cat eye syndrome SpecificDisease C535918|OMIM:115470 1424237 128 144 cat eye syndrome SpecificDisease C535918|OMIM:115470 1424237 146 149 CES SpecificDisease C535918|OMIM:115470 1424237 188 191 CES SpecificDisease C535918|OMIM:115470 1424237 388 397 tetrasomy DiseaseClass D058670 1313112|t|An intrachromosomal insertion causing 5q22 deletion and familial adenomatous polyposis coli in two generations. 1313112|a|We report familial adenomatous polyposis coli (FAPC) with epidermoid cysts, osteomata, and areas of congenital hypertrophy of the retinal pigment epithelium (CHRPEs) in a male patient and his maternal aunt, both of whom suffered a mild to moderate degree of mental handicap. Both had an interstitial deletion of the long arm of chromosome 5 (del (5) (q22q23. 2)). Two other normal family members had the underlying direct insertion of chromosome 5 (dir ins (5) (q31. 3q22q23 3q22q23. 2)). Molecular genetic and fluorescent hybridisation studies have shown that loci D5S37 and D5S98 are outside the deletion whereas loci detected by probes EF5. 44 and YN5. 48 are lost. As expected, the molecular analyses indicate loss of one allele at the MCC and APC loci. The APC gene is located within band 5q22. Familial direct insertions should be considered as a cause of recurrent microdeletion syndromes. 1313112 56 91 familial adenomatous polyposis coli SpecificDisease D011125 1313112 122 157 familial adenomatous polyposis coli SpecificDisease D011125 1313112 159 163 FAPC SpecificDisease D011125 1313112 170 186 epidermoid cysts SpecificDisease D004814 1313112 188 197 osteomata SpecificDisease D010016 1313112 212 268 congenital hypertrophy of the retinal pigment epithelium SpecificDisease D012164 1313112 270 276 CHRPEs SpecificDisease D012164 1313112 370 385 mental handicap DiseaseClass D008607 1313112 860 863 APC Modifier D011125 1313112 874 877 APC Modifier D011125 1303170|t|Cloning of the Huntington disease region in yeast artificial chromosomes. 1303170|a|The gene responsible for Huntington disease has been localized to a 2. 5 million base pair (Mb) region between the loci D4S10 and D4S168 on the short arm of chromosome 4. As part of a strategy to clone the HD gene on the basis of its chromosomal location, we isolated genomic DNA from the HD region as a set of overlapping yeast artificial chromosome (YAC) clones. Twenty-eight YAC clones were identified by screening human YAC libraries with twelve PCR-based sequence-tagged sites (STSs) from the region. We assembled the YAC clones into overlapping sets by hybridizing them to a large number of DNA probes from the HD region, including the STSs. In addition, we isolated the ends of the human DNA inserts of most of the YAC clones to assist in the construction of the contig. Although almost half of the YACs appear to contain chimeric inserts and several contain internal deletions or other rearrangements, we were able to obtain over 2. 2 Mb of the HD region in YACs, including one continuous segment of 2. 0 Mb covering the region that most likely contains the HD gene. Ten of the twenty eight YAC clones comprise a minimal set spanning the 2. 2 Mb. These clones provide reagents for the complete characterization of this region of the genome and for the eventual isolation of the HD gene. 1303170 15 33 Huntington disease Modifier D006816 1303170 99 117 Huntington disease SpecificDisease D006816 1303170 280 282 HD Modifier D006816 1303170 363 365 HD Modifier D006816 1303170 691 693 HD Modifier D006816 1303170 1027 1029 HD Modifier D006816 1303170 1140 1142 HD Modifier D006816 1303170 1360 1362 HD Modifier D006816 8113388|t|Three novel mutations in five unrelated subjects with hereditary protein S deficiency type I. 8113388|a|A panel of eight unrelated subjects with inherited type I protein S deficiency was screened for mutations in the PROS1 gene. In five subjects an abnormality was found but mutations were not detected in the remaining three subjects. Two subjects shared a G-- > A transition at position + 5 of the donor splice site consensus sequence of intron 10. Also in two subjects an A-- > T transversion was detected in the stopcodon of the PROS1 gene; this transversion predicts a protein S molecule that is extended by 14 amino acids. The fifth subject was found to possess two sequence abnormalities. One allele carried a G-- > A transition near the donor splice junction of intron 2, but this abnormality is probably neutral, since it was inherited from the parent with normal protein S antigen levels. In the other allele a single T insertion in codon -25 was found. Analysis of platelet RNA showed that only the mRNA with the A-- > T mutation in the stopcodon is present in amounts comparable to wildtype RNA. mRNA from the alleles with the other two mutations was either undetectable or present in greatly reduced amounts. The latter indicates that a mRNA based approach is not feasible for the genetic analysis of protein S deficiency type I.. 8113388 65 92 protein S deficiency type I SpecificDisease D018455 8113388 145 172 type I protein S deficiency SpecificDisease D018455 8113388 1304 1331 protein S deficiency type I SpecificDisease D018455 3029599|t|A potential animal model for Lesch-Nyhan syndrome through introduction of HPRT mutations into mice. 3029599|a|The human Lesch-Nyhan syndrome is a rare neurological and behavioural disorder, affecting only males, which is caused by an inherited deficiency in the level of activity of the purine salvage enzyme hypoxanthine-guanosine phosphoribosyl transferase (HPRT). How the resulting alterations in purine metabolism lead to the severe symptoms characteristic of Lesch-Nyhan patients is still not understood. No mutations at the Hprt locus leading to loss of activity have been described in laboratory animals. To derive an animal model for the Lesch-Nyhan syndrome, we have used cultured mouse embryonic stem cells, mutagenized by retroviral insertion and selected for loss of HPRT activity, to construct chimaeric mice. Two clonal lines carrying different mutant Hprt alleles have given rise to germ cells in chimaeras, allowing the derivation of strains of mutant mice having the same biochemical defect as Lesch-Nyhan patients. Male mice carrying the mutant alleles are viable and analysis of their cells shows a total lack of HPRT activity.. 3029599 29 49 Lesch-Nyhan syndrome SpecificDisease D007926 3029599 110 130 Lesch-Nyhan syndrome SpecificDisease D007926 3029599 141 178 neurological and behavioural disorder DiseaseClass D009422|D001523 3029599 224 244 inherited deficiency DiseaseClass D030342 3029599 454 465 Lesch-Nyhan Modifier D007926 3029599 636 656 Lesch-Nyhan syndrome SpecificDisease D007926 3029599 1001 1012 Lesch-Nyhan Modifier D007926 2316519|t|Duplicational mutation at the Duchenne muscular dystrophy locus: its frequency, distribution, origin, and phenotypegenotype correlation. 2316519|a|Partial gene deletion is the major cause of mutation leading to Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). Partial gene duplication has also been recognized in a few cases. We have conducted a survey for duplication in 72 unrelated nondeletion patients, analyzed by Southern blot hybridization with clones representing the entire DMD cDNA. With careful quantitative analysis of hybridization band intensity, 10 cases were found to carry a duplication of part of the gene, a frequency of 14% for nondeletion cases (10/72), or 6% for all cases (10/181). The extent of these duplications has been characterized according to the published exon-containing HindIII fragment map, and in six of the 10 duplications a novel restriction fragment that spanned the duplication junction was detected. The resulting translational reading frame of mRNA has been predicted for nine duplications. A shift of the reading frame was predicted in four of the six DMD cases and in one of the two intermediate cases, while the reading frame remained uninterrupted in both BMD cases. RFLP and quantitative Southern blot analyses revealed a grandpaternal origin of duplication in four families and grandmaternal origin in one family. In all five families, the duplication was found to originate from a single X chromosome. Unequal sister-chromatid exchange is proposed to be the mechanism for the formation of these duplications.. 2316519 30 57 Duchenne muscular dystrophy Modifier D020388 2316519 201 228 Duchenne muscular dystrophy SpecificDisease D020388 2316519 230 233 DMD SpecificDisease D020388 2316519 239 264 Becker muscular dystrophy SpecificDisease C537666 2316519 266 269 BMD SpecificDisease C537666 2316519 495 498 DMD Modifier D020388 2316519 1107 1110 DMD Modifier D020388 2316519 1214 1217 BMD Modifier C537666 6087154|t|Molecular evidence for new mutation at the hprt locus in Lesch-Nyhan patients. 6087154|a|Hypoxanthine-guanine phosphoribosyltransferase (HPRT; EC2. 4. 2. 8), which functions in the metabolic salvage of purines, is encoded by an X-linked gene in man. Partial HPRT deficiencies are associated with gouty arthritis, while absence of activity results in Lesch-Nyhan syndrome (L-N). L-N patients fail to reproduce and the heterozygous state appears to confer no selective advantage. Thus, Haldanes principle predicts that new mutations at the hprt locus must occur frequently in order for L-N syndrome to be maintained in the population. This constant introduction of new mutations would be expected to result in a heterogeneous collection of genetic lesions, some of which may be novel. As we report here, the mutations in the hprt gene of seven L-N patients, selected from an initial survey of 28 patients, have been characterized and all were found to be distinctly different, as predicted. The origin of one unusual mutation has been identified by analysis of DNA from four generations of family members. Further molecular analysis of the origin of new mutations at the hprt locus should aid in resolving the issue of an apparent difference in the frequency of hprt mutations in males and females 6087154 57 68 Lesch-Nyhan Modifier D007926 6087154 248 265 HPRT deficiencies DiseaseClass OMIM:300323 6087154 286 301 gouty arthritis SpecificDisease D015210 6087154 340 360 Lesch-Nyhan syndrome SpecificDisease D007926 6087154 362 365 L-N SpecificDisease D007926 6087154 368 371 L-N Modifier D007926 6087154 574 586 L-N syndrome SpecificDisease D007926 6087154 728 743 genetic lesions DiseaseClass D020022 6087154 832 835 L-N Modifier D007926 3600793|t|The mapping of a cDNA from the human X-linked Duchenne muscular dystrophy gene to the mouse X chromosome. 3600793|a|The recent discovery of sequences at the site of the Duchenne muscular dystrophy (DMD) gene in humans has opened up the possibility of a detailed molecular analysis of the genes in humans and in related mammalian species. Until relatively recently, there was no obvious mouse model of this genetic disease for the development of therapeutic strategies. The identification of a mouse X-linked mutant showing muscular dystrophy, mdx, has provided a candidate mouse genetic homologue to the DMD locus; the relatively mild pathological features of mdx suggest it may have more in common with mutations of the Becker muscular dystrophy type at the same human locus, however. But the close genetic linkage of mdx to G6PD and Hprt on the mouse X chromosome, coupled with its comparatively mild pathology, have suggested that the mdx mutation may instead correspond to Emery Dreifuss muscular dystrophy which itself is closely linked to DNA markers at Xq28-qter in the region of G6PD on the human X chromosome. Using an interspecific mouse domesticus/spretus cross, segregating for a variety of markers on the mouse X chromosome, we have positioned on the mouse X chromosome sequences homologous to a DMD cDNA clone. These sequences map provocatively close to the mdx mutation and unexpectedly distant from sparse fur, spf, the mouse homologue of OTC (ornithine transcarbamylase) which is closely linked to DMD on the human X chromosome.. 3600793 37 73 X-linked Duchenne muscular dystrophy Modifier D020388 3600793 159 186 Duchenne muscular dystrophy Modifier D020388 3600793 188 191 DMD Modifier D020388 3600793 396 411 genetic disease DiseaseClass D030342 3600793 513 531 muscular dystrophy SpecificDisease D009136 3600793 594 597 DMD Modifier D020388 3600793 711 736 Becker muscular dystrophy Modifier C537666 3600793 967 1000 Emery Dreifuss muscular dystrophy SpecificDisease D020389 3600793 1299 1302 DMD Modifier D020388 3600793 1505 1508 DMD SpecificDisease D020388 10094559|t|Identification of the mutation in the alkaptonuria mouse model. 10094559|a|Alkaptonuria (aku), an inborn error of metabolism caused by the loss of homogentisate 1, 2-dioxygenase (HGD), has been described in a mouse model created by ethylnitrosourea mutagenesis but the mutation in these mice has not previously been identified. We used RT-PCR to amplify the Hgd cDNA from Hgd (aku)/Hgd (aku) mice. Two products shorter than the wild-type product were amplified. Restriction mapping and DNA sequencing were then used to identify the Hgd (aku) mouse mutation, found to be a single base change in a splice donor consensus sequence, causing exon skipping and frame-shifted products. This base change allowed us to create a non-radioactive genotyping assay for this allele. 10094559 38 50 alkaptonuria Modifier D000474 10094559 64 76 Alkaptonuria SpecificDisease D000474 10094559 78 81 aku SpecificDisease D000474 10094559 87 113 inborn error of metabolism DiseaseClass D008661 1347968|t|Common sequence motifs at the rearrangement sites of a constitutional X/autosome translocation and associated deletion. 1347968|a|Reciprocal chromosome translocations are common de novo rearrangements that occur randomly throughout the human genome. To learn about causative mechanisms, we have cloned and sequenced the breakpoints of a cytologically balanced constitutional reciprocal translocation, t (X; 4) (p21. 2; q31. 22), present in a girl with Duchenne muscular dystrophy (DMD). Physical mapping of the derivative chromosomes, after their separation in somatic cell hybrids, reveals that the translocation disrupts the DMD gene in Xp21 within the 18-kb intron 16. Restriction mapping and sequencing of clones that span both translocation breakpoints as well as the corresponding normal regions indicate the loss of approximately 5 kb in the formation of the derivative X chromosome, with 4-6 bp deleted from chromosome 4. RFLP and Southern analyses indicate that the de novo translocation is a paternal origin and that the fathers X chromosome contains the DNA that is deleted in the derivative X. Most likely, deletion and translation arose simultaneously from a complex rearrangement event that involves three chromosomal breakpoints. Short regions of sequence homology were present at the three sites. A 5-bp sequence, GGAAT, found exactly at the translocation breakpoints on both normal chromosomes X and 4, has been preserved only on the der (4) chromosome. It is likely that the X-derived sequence GGAATCA has been lost in the formation of the der (X) chromosome, as it matches an inverted GAATCA sequence present on the opposite strand exactly at the other end of the deleted 5-kb fragment. 1347968 442 469 Duchenne muscular dystrophy SpecificDisease D020388 1347968 471 474 DMD SpecificDisease D020388 1347968 617 620 DMD Modifier D020388 10874302|t|The human factor IX gene as germline mutagen test: samples from Mainland China have the putatively endogenous pattern of mutation. 10874302|a|Germline mutations are the major source of genetic variation that allows a species to evolve over time but at the cost of Mendelian disease and genetic predisposition to multifactorial diseases. Previous analyses have revealed that the pattern of germline mutations in the factor IX gene (F9) is similar among a variety of ethnically and geographically diverse populations and compatible with the ancient pattern that has shaped the mammalian genome. Here, we compare the pattern of germline mutation in a population of hemophilia B patients from Mainland China (n = 66) to that in U. S. Caucasians, Blacks, and Mexican Hispanics and stratify by disease severity and ethnicity. The similar pattern of germline mutation in all ethnic groups studied to date provides additional data compatible with the inference that endogenous processes predominate in germline mutations. 10874302 253 270 Mendelian disease DiseaseClass D030342 10874302 301 324 multifactorial diseases DiseaseClass D004194 10874302 651 663 hemophilia B Modifier D002836 3591825|t|Phenotype heterogeneity among hemizygotes in a family biochemically screened for adrenoleukodystrophy. 3591825|a|We report on two clinically, neurologically normal relatives of a boy affected by adrenoleukodystrophy (ALD); they were found repeatedly to have the biochemical defect of an ALD hemizygote. The assay consisted in the determination of very-long-chain fatty acids in lyophilized and reconstituted plasma. While no evidence of neurologic disease (leukodystrophy or myeloneuropathy) was present in these hemizygotes, adrenocortical insufficiency provoking compensatory high ACTH release was found in both. These findings should be taken into consideration when counseling families in which cases with clinically expressed ALD are represented in several generations.. 3591825 81 101 adrenoleukodystrophy SpecificDisease D000326 3591825 185 205 adrenoleukodystrophy SpecificDisease D000326 3591825 207 210 ALD SpecificDisease D000326 3591825 277 280 ALD Modifier D000326 3591825 427 445 neurologic disease DiseaseClass D009422 3591825 447 461 leukodystrophy SpecificDisease D007966 3591825 465 480 myeloneuropathy SpecificDisease D009422 3591825 516 544 adrenocortical insufficiency DiseaseClass D000224 3591825 721 724 ALD SpecificDisease D000326 10721669|t|Novel mutations of the ATP7B gene in Japanese patients with Wilson disease. 10721669|a|Wilson disease (WD) is an autosomal recessive disorder characterized by copper accumulation in the liver, brain, kidneys, and corneas, and culminating in copper toxication in these organs. In this study, we analyzed mutations of the responsible gene, ATP7B, in four Japanese patients with WD. By direct sequencing, we identified five mutations, of which two were novel, and 16 polymorphisms, of which 6 were novel. The mutations 2871delC and 2513delA shift the reading frame so that truncated abnormal protein is expected. In contrast to these mutations found in patients with hepatic-type of early onset, the mutations A874V, R778L, and 3892delGTC were either missense mutations or in frame 1-amino acid deletion, and occurred in the patients with hepato-neurologic type of late onset. The mutations 2871delC and R778L have been previously reported in a relatively large number of Japanese patients. In particular, R778L is known to be more prevalent in Asian countries than in other countries of the world. Our data are compatible with the hypothesis that the mutations tend to occur in a population-specific manner. Therefore, the accumulation of the types of mutations in Japanese patients with WD will facilitate the fast and effective genetic diagnosis of WD in Japanese patients.. 10721669 60 74 Wilson disease SpecificDisease D006527 10721669 76 90 Wilson disease SpecificDisease D006527 10721669 92 94 WD SpecificDisease D006527 10721669 102 130 autosomal recessive disorder DiseaseClass D030342 10721669 365 367 WD SpecificDisease D006527 10721669 1275 1277 WD SpecificDisease D006527 10721669 1338 1340 WD SpecificDisease D006527 7759106|t|Structural analysis of the 5' region of mouse and human Huntington disease genes reveals conservation of putative promoter region and di- and trinucleotide polymorphisms. 7759106|a|We have previously cloned and characterized the murine homologue of the Huntington disease (HD) gene and shown that it maps to mouse chromosome 5 within a region of conserved synteny with human chromosome 4p16. 3 3. Here we present a detailed comparison of the sequence of the putative promoter and the organization of the 5 genomic region of the murine (Hdh) and human HD genes encompassing the first five exons. We show that in this region these two genes share identical exon boundaries, but have different-size introns. Two dinucleotide (CT) and one trinucleotide intronic polymorphism in Hdh and an intronic CA polymorphism in the HD gene were identified. Comparison of 940-bp sequence 5 to the putative translation start site reveals a highly conserved region (78. 8% nucleotide identity) between Hdh and the HD gene from nucleotide -56 to -206 (of Hdh). Neither Hdh nor the HD gene have typical TATA or CCAAT elements, but both show one putative AP2 binding site and numerous potential Sp1 binding sites. The high sequence identity between Hdh and the HD gene for approximately 200 bp 5 to the putative translation start site indicates that these sequences may play a role in regulating expression of the Huntington disease gene 7759106 56 74 Huntington disease Modifier D006816 7759106 243 261 Huntington disease Modifier D006816 7759106 263 265 HD Modifier D006816 7759106 541 543 HD Modifier D006816 7759106 807 809 HD Modifier D006816 7759106 986 988 HD Modifier D006816 7759106 1052 1054 HD Modifier D006816 7759106 1230 1232 HD Modifier D006816 7759106 1383 1401 Huntington disease Modifier D006816 6103091|t|Complement deficiency and nephritis. A report of a family. 6103091|a|A family is described in which three children had homozygous deficiency of C3 and in which both parents and two other children were heterozygous for the C3 null gene. One child with heterozygous C3 deficiency was found to have membranoproliferative glomerulonephritis; proteinuria and/or microscopical haematuria was present in all three homozygous C3-deficient children. All children with homozygous or heterozygous C3 deficiency were, to a varying degree, susceptible to infection. The only child of the family with normal complement had no increased risk of infection and no renal disease. This family study provides further support for the proposal that C3 deficiency predisposes to nephritis.. 6103091 0 21 Complement deficiency SpecificDisease D007153 6103091 26 35 nephritis SpecificDisease D009393 6103091 120 136 deficiency of C3 SpecificDisease OMIM:613779 6103091 254 267 C3 deficiency SpecificDisease OMIM:613779 6103091 308 326 glomerulonephritis SpecificDisease D005921 6103091 328 339 proteinuria SpecificDisease D011507 6103091 361 371 haematuria SpecificDisease D006417 6103091 408 420 C3-deficient Modifier OMIM:613779 6103091 476 489 C3 deficiency SpecificDisease OMIM:613779 6103091 637 650 renal disease DiseaseClass D007674 6103091 717 730 C3 deficiency SpecificDisease OMIM:613779 6103091 746 755 nephritis SpecificDisease D009393 1222588|t|Cytogenetic investigations in families with ataxia-telangiectasia. 1222588|a|Chromosomal studies were performed on peripheral blood lymphocytes and cultured skin fibroblasts from five Israeli-Moroccan families with ataxia-telangiectasia. A total of 24 individuals, including seven propositi, was investigated. Among the probands, significantly elevated rates of chromosome damage were observed in both blood and skin. Skin fibroblasts of affected individuals showed several orders of magnitude more chromosome breakage than lymphocytes. Increased rates of chromosome damage were also observed in the fibroblasts of some phenotypically normal family members (obligate heterozygotes and sibs) when compared to normal controls. An apparent abnormal clone of cells, possessing a large acrocentric marker chromosome (14q +), was observed in varying proportions among cells of all the propositi (2-5% of lymphocytes; 1-9% of fibroblasts).. 1222588 44 65 ataxia-telangiectasia SpecificDisease D001260 1222588 205 226 ataxia-telangiectasia SpecificDisease D001260 7991123|t|Predominance of the adrenomyeloneuropathy phenotype of X-linked adrenoleukodystrophy in The Netherlands: a survey of 30 kindreds. 7991123|a|X-linked adrenoleukodystrophy (X-ALD) is an inherited disorder of peroxisomal beta-oxidation associated with accumulation of saturated very long-chain fatty acids, which results in central and peripheral demyelination and in impaired function of adrenal cortex and testes. The phenotypic expression is highly variable, childhood cerebral ALD (CCALD) and adrenomyeloneuropathy (AMN) being the main variants. We explored the 30 Dutch kindreds well known to the Dutch X-ALD/AMN Study Group and phenotyped 77 male patients 35 (46%) had AMN and 24 (31%) CCALD or adolescent cerebral ALD (AdolCALD). These percentages differ significantly from previous reports, in which 25 to 28% of the patients developed AMN and 53 to 57% CCALD or AdolCALD. Our findings indicate that--at least in the Netherlands--AMN may be the most frequent phenotype of X-ALD.. 7991123 20 41 adrenomyeloneuropathy Modifier D000326 7991123 55 84 X-linked adrenoleukodystrophy SpecificDisease D000326 7991123 130 159 X-linked adrenoleukodystrophy SpecificDisease D000326 7991123 161 166 X-ALD SpecificDisease D000326 7991123 174 192 inherited disorder DiseaseClass D030342 7991123 334 347 demyelination DiseaseClass D003711 7991123 355 401 impaired function of adrenal cortex and testes CompositeMention D000303 7991123 449 471 childhood cerebral ALD SpecificDisease D000326 7991123 473 478 CCALD SpecificDisease D000326 7991123 484 505 adrenomyeloneuropathy SpecificDisease D000326 7991123 507 510 AMN SpecificDisease D000326 7991123 595 600 X-ALD Modifier D000326 7991123 601 604 AMN Modifier D000326 7991123 663 666 AMN SpecificDisease D000326 7991123 680 685 CCALD SpecificDisease D000326 7991123 689 712 adolescent cerebral ALD SpecificDisease D000326 7991123 714 722 AdolCALD SpecificDisease D000326 7991123 832 835 AMN SpecificDisease D000326 7991123 850 855 CCALD SpecificDisease D000326 7991123 859 867 AdolCALD SpecificDisease D000326 7991123 926 929 AMN SpecificDisease D000326 7991123 968 973 X-ALD SpecificDisease D000326 7550230|t|Three novel aniridia mutations in the human PAX6 gene. 7550230|a|Aniridia (iris hypoplasia) is an autosomal dominant congenital disorder of the eye. Mutations in the human aniridia (PAX6) gene have now been identified in many patients from various ethnic groups. In the study reported here we describe PAX6 mutations in one sporadic and five familial cases with aniridia. Of the four different mutations identified, one was identical to a previously reported mutation (C-- > T transition at codon 240), and three were novel two in the glycine-rich region and one in the proline/serine/threonine-rich (PST) region. One PAX6 mutation found in the PST region was associated with cataracts in an aniridia family. Another splice mutation in the PST domain occurred in an aniridia patient with anosmia (inability to smell). The six new aniridia cases reported here have mutations predicted to generate incomplete PAX6 proteins. These results support the theory that human aniridia is caused by haploinsufficiency of PAX6.. 7550230 12 20 aniridia Modifier D015783 7550230 55 63 Aniridia SpecificDisease D015783 7550230 65 80 iris hypoplasia SpecificDisease D007499 7550230 88 137 autosomal dominant congenital disorder of the eye DiseaseClass D005124 7550230 162 170 aniridia Modifier D015783 7550230 352 360 aniridia SpecificDisease D015783 7550230 667 676 cataracts SpecificDisease D002386 7550230 683 691 aniridia Modifier D015783 7550230 757 765 aniridia Modifier D015783 7550230 779 786 anosmia SpecificDisease D000857 7550230 821 829 aniridia Modifier D015783 7550230 957 965 aniridia SpecificDisease D015783 7550230 979 1005 haploinsufficiency of PAX6 SpecificDisease OMIM:106210 3354603|t|Hepatoblastoma, pigmented ocular fundus lesions and jaw lesions in Gardner syndrome. 3354603|a|Hepatoblastoma is a rare neoplasm of infants and children only recently documented in association with hereditary adenomatous polyposis of the colon [Kingston et al., 1983]. We report four children with hepatoblastoma from four unrelated families with Gardner syndrome (GS). One child, now 19 years old, survived after a resection of a hepatoblastoma in infancy and recently was found to have GS. He has an associated odontoma and pigmented ocular fundus lesions, both of which have been shown to be clinical markers of GS. Many individuals in these four GS families, both affected and at risk, have osteomatous jaw lesions and pigmented ocular fundus lesions. A search for colonic polyps should be made in families of infants and children with hepatoblastoma. If the child survives, he or she should be monitored for the later appearance of colonic polyps. The finding of jaw lesions and/or pigmented ocular fundus lesions in relatives at risk are indications of the possible presence of the GS gene. 3354603 0 14 Hepatoblastoma SpecificDisease D018197 3354603 16 47 pigmented ocular fundus lesions DiseaseClass D015821 3354603 52 63 jaw lesions DiseaseClass D007571 3354603 67 83 Gardner syndrome SpecificDisease D005736 3354603 85 99 Hepatoblastoma SpecificDisease D018197 3354603 110 118 neoplasm DiseaseClass D009369 3354603 188 233 hereditary adenomatous polyposis of the colon SpecificDisease D011125 3354603 288 302 hepatoblastoma SpecificDisease D018197 3354603 337 353 Gardner syndrome SpecificDisease D005736 3354603 355 357 GS SpecificDisease D005736 3354603 421 435 hepatoblastoma SpecificDisease D018197 3354603 478 480 GS SpecificDisease D005736 3354603 503 511 odontoma SpecificDisease D009810 3354603 516 547 pigmented ocular fundus lesions DiseaseClass D015821 3354603 605 607 GS SpecificDisease D005736 3354603 640 642 GS Modifier D005736 3354603 685 708 osteomatous jaw lesions DiseaseClass D007571 3354603 713 744 pigmented ocular fundus lesions DiseaseClass D015821 3354603 759 773 colonic polyps DiseaseClass D003111 3354603 830 844 hepatoblastoma SpecificDisease D018197 3354603 927 941 colonic polyps DiseaseClass D003111 3354603 958 969 jaw lesions DiseaseClass D007571 3354603 977 1008 pigmented ocular fundus lesions DiseaseClass D015821 3354603 1078 1080 GS Modifier D005736 7981671|t|Mutation spectrum in the CHM gene of Danish and Swedish choroideremia patients. 7981671|a|The recent isolation of the complete open reading frame of the choroideremia (CHM) gene and the characterization of the exon-intron boundaries has paved the way to mutation detection in patients with classical choroideremia. We have performed mutation screening in patients from 15 Danish and Swedish families by using Southern blot hybridization and the polymerase chain reaction single-strand conformation polymorphism (PCR-SSCP) technique. Causative mutations in the CHM gene were detected in at least 12 families, indicating that a substantial part of the mutations can be identified by this approach. In four of these families deletions of different sizes were found. Thus, in one patient, the deletion resulted in the absence of only one exon, while in another the deletion comprised the entire CHM gene. Mapping of the deletion endpoints in these four patients and in another 11 male patients with sizeable deletions enabled us to construct a very detailed map of intervals 2 and 3 of Xq21. In the remaining 11 Danish and Swedish families at least 8 causative mutations were found by PCR-SSCP analysis and direct sequencing. Interestingly, all CHM gene mutations detected thus far in choroideremia patients give rise to the introduction of a premature stop codon.. 7981671 25 28 CHM Modifier D015794 7981671 56 69 choroideremia Modifier D015794 7981671 143 156 choroideremia Modifier D015794 7981671 158 161 CHM Modifier D015794 7981671 290 303 choroideremia SpecificDisease D015794 7981671 550 553 CHM Modifier D015794 7981671 881 884 CHM Modifier D015794 7981671 1231 1234 CHM Modifier D015794 7981671 1271 1284 choroideremia Modifier D015794 1562739|t|Diverse point mutations result in glucose-6-phosphate dehydrogenase (G6PD) polymorphism in Taiwan. 1562739|a|Glucose-6-PHOSPHATE dehydrogenase (G6PD; EC 1.1. 1. 49) deficiency is the most common human enzymopathy, affecting more than 200 million people worldwide. Although greater than 400 variants have been described based on clinical and biochemical criteria, little is known about the molecular basis of these G6PD deficiencies. Recently, the gene that encodes human G6PD has been cloned and sequenced, which enables us to examine directly the heterogeneity of G6PD at the DNA level. During the past 10 years, we examined the G6PD activity in 21, 271 newborn Chinese infants (11, 400 males and 9, 871 females) and identified 314 (2. 8%) males and 246 (2. 5%) females having low G6PD activity. The G6PD gene from 10 randomly selected affected individuals and their relatives was polymerase chain reaction (PCR) amplified, subcloned, and sequenced. Our results indicate that at least four types of mutation are responsible for the G6PD polymorphism in Taiwan. The first type of mutation (487 G----A) was found in an affected Chinese with a G to A change at nucleotide 487, which results in a (163) Gly to Ser substitution. The second type of mutation (493 A----G) is a novel mutation that has not been reported in any other ethnic group and was identified in two affected Chinese. This mutation causes an A to G change at nucleotide position 493, producing an (165) Asn to Asp substitution. Interestingly, the 487 G----A and 493 A----G mutations create Alu I and Ava II recognition sites, respectively, which enabled us to rapidly detect these two mutations by PCR/restriction enzyme (RE) digestion method. The third mutation (1376 G----T) was found in four affected Chinese. This mutation causes a G to T change at nucleotide position 1376 that results in an (459) Arg to Leu substitution. The 1376 G----T mutation seems to be the dominant allele that causes G6PD deficiency in Taiwan. Finally, two affected Chinese were identified as having the fourth mutation (1388 G----A). This mutation causes a G to A change at nucleotide 1388 that produces an (463) Arg to His substitution. Our studies provide the direct proof of the genetic heterogeneity of G6PD deficiency in the Chinese populations of Taiwan and the PCR/RE digestion method is suitable for simultaneous detection of the 487 G----A and 493 A----G mutations. 1562739 99 165 Glucose-6-PHOSPHATE dehydrogenase (G6PD; EC 1.1. 1. 49) deficiency SpecificDisease D005955 1562739 191 202 enzymopathy DiseaseClass D008661 1562739 404 421 G6PD deficiencies SpecificDisease D005955 1562739 1952 1967 G6PD deficiency SpecificDisease D005955 1562739 2243 2258 G6PD deficiency SpecificDisease D005955 10875918|t|Fas preassociation required for apoptosis signaling and dominant inhibition by pathogenic mutations. 10875918|a|Heterozygous mutations encoding abnormal forms of the death receptor Fas dominantly interfere with Fas-induced lymphocyte apoptosis in human autoimmune lymphoproliferative syndrome. This effect, rather than depending on ligand-induced receptor oligomerization, was found to stem from ligand- independent interaction of wild-type and mutant Fas receptors through a specific region in the extracellular domain. Preassociated Fas complexes were found in living cells by means of fluorescence resonance energy transfer between variants of green fluorescent protein. These results show that formation of preassociated receptor complexes is necessary for Fas signaling and dominant interference in human disease.. 10875918 242 281 autoimmune lymphoproliferative syndrome SpecificDisease D056735 8434621|t|Familial Mediterranean fever in the colchicine era: the fate of one family. 8434621|a|In order to demonstrate the effect of prophylactic colchicine treatment on the natural history of familial Mediterranean fever (FMF), a family is presented with 6 out of 9 siblings affected by FMF. Each patient represents a different stage of the amyloidotic kidney disease of FMF and the effect of continuous colchicine treatment on its course. Considered together, the members of this family present an almost complete clinical, genetic, and behavioral picture of the disease.. 8434621 0 28 Familial Mediterranean fever SpecificDisease D010505 8434621 174 202 familial Mediterranean fever SpecificDisease D010505 8434621 204 207 FMF SpecificDisease D010505 8434621 269 272 FMF SpecificDisease D010505 8434621 323 349 amyloidotic kidney disease SpecificDisease D007674 8434621 353 356 FMF SpecificDisease D010505 1684088|t|Two new arylsulfatase A (ARSA) mutations in a juvenile metachromatic leukodystrophy (MLD) patient. 1684088|a|Fragments of the arylsulfatase A (ARSA) gene from a patient with juvenile-onset metachromatic leukodystrophy (MLD) were amplified by PCR and ligated into MP13 cloning vectors. Clones hybridizing with cDNA for human ARSA were selected, examined for appropriate size inserts, and used to prepare single-stranded phage DNA. Examination of the entire coding and most of the intronic sequence revealed two putative disease-related mutations. One, a point mutation in exon 3, resulted in the substitution of isoleucine by serine. Introduction of this alteration into the normal ARSA cDNA sequence resulted in a substantial decrease in ARSA activity on transient expression in cultured baby hamster kidney cells. About 5% of the control expression was observed, suggesting a small residual activity in the mutated ARSA. The second mutation, a G-to-A transition, occurred in the other allele and resulted in an altered splice-recognition sequence between exon 7 and the following intron. The mutation also resulted in the loss of a restriction site. Apparently normal levels of mRNA were generated from this allele, but no ARSA activity or immuno-cross-reactive material could be detected. A collection of DNA samples from known or suspected MLD patients, members of their families, and normal controls was screened for these mutations. Four additional individuals carrying each of the mutations were found among the nearly 100 MLD patients in the sample. Gene segregation in the original patients family was consistent with available clinical and biochemical data. No individuals homozygous for either of these two mutations were identified. However, combinations with other MLD mutations suggest that the point mutation in exon 3 does result in some residual enzyme activity and is associated with late-onset forms of the disease. The splice-site mutation following exon 7 produces late-infantile MLD when combined with other enzyme-null mutations, implying that it is completely silent enzymatically.. 1684088 55 83 metachromatic leukodystrophy Modifier D007966 1684088 85 88 MLD Modifier D007966 1684088 179 207 metachromatic leukodystrophy SpecificDisease D007966 1684088 209 212 MLD SpecificDisease D007966 1684088 1333 1336 MLD Modifier D007966 1684088 1519 1522 MLD Modifier D007966 1684088 1767 1770 MLD Modifier D007966 1684088 1990 1993 MLD SpecificDisease D007966 10441329|t|Null mutation of the murine ATP7B (Wilson disease) gene results in intracellular copper accumulation and late-onset hepatic nodular transformation. 10441329|a|The Atp7b protein is a copper-transporting ATPase expressed predominantly in the liver and to a lesser extent in most other tissues. Mutations in the ATP7B gene lead to Wilson disease, a copper toxicity disorder characterized by dramatic build-up of intracellular hepatic copper with subsequent hepatic and neuro-logical abnormalities. Using homologous recombination to disrupt the normal translation of ATP7B, we have generated a strain of mice that are homozygous mutants (null) for the Wilson disease gene. The ATP7B null mice display a gradual accumulation of hepatic copper that increases to a level 60-fold greater than normal by 5 months of age. An increase in copper concentration was also observed in the kidney, brain, placenta and lactating mammary glands of homo-zygous mutants, although milk from the mutant glands was copper deficient. Morphological abnormalities resembling cirrhosis developed in the majority of the livers from homozygous mutants older than 7 months of age. Progeny of the homozygous mutant females demonstrated neurological abnormalities and growth retardation characteristic of copper deficiency. Copper concentration in the livers of the newborn homozygous null mutants was decreased dramatically. In summary, inactivation of the murine ATP7B gene produces a form of cirrhotic liver disease that resembles Wilson disease in humans and the toxic milk phenotype in the mouse.. 10441329 35 49 Wilson disease SpecificDisease D006527 10441329 67 100 intracellular copper accumulation DiseaseClass C535468 10441329 105 146 late-onset hepatic nodular transformation DiseaseClass D020518 10441329 317 331 Wilson disease SpecificDisease D006527 10441329 335 359 copper toxicity disorder DiseaseClass C535468 10441329 443 482 hepatic and neuro-logical abnormalities CompositeMention D008107|D009422 10441329 637 651 Wilson disease Modifier D006527 10441329 980 996 copper deficient DiseaseClass C535468 10441329 998 1025 Morphological abnormalities DiseaseClass D000013 10441329 1037 1046 cirrhosis SpecificDisease D008103 10441329 1193 1219 neurological abnormalities DiseaseClass D009461 10441329 1224 1242 growth retardation DiseaseClass D006130 10441329 1261 1278 copper deficiency SpecificDisease C535468 10441329 1451 1474 cirrhotic liver disease DiseaseClass D008103 10441329 1490 1504 Wilson disease SpecificDisease D006527 218453|t|Adrenoleukodystrophy and adrenomyeloneuropathy associated with partial adrenal insufficiency in three generations of a kindred. 218453|a|Four cases of adrenoleukodystrophy (ALD) and one case of adrenomyeloneuropathy (AMN) have developed in a kindred over three generations demonstrating that AMN is a clinical variant of ALD. Pituitary-adrenal function studies were performed in 10 family members, including two affected males and four females identified as carriers of ALD/AMN. No pituitary-adrenal abnormality was found in the carriers. However, basal morning plasma adrenocorticotropic hormone (ACTH) levels were markedly elevated in the two males with ALD and AMN, despite the fact that they had no clinical signs of adrenal insufficiency and that morning plasma cortisol levels and their response to maximal exogenous ACTH stimulation appeared to be normal. In addition, the integrated 24-hour response to the administration were also subnormal in these two cases. Thus, people with ALD and AMN may have subclinical partial adrenocrotical insufficiency. No other endocrinologic dysfunction was identified.. 218453 0 20 Adrenoleukodystrophy SpecificDisease D000326 218453 25 46 adrenomyeloneuropathy SpecificDisease D000326 218453 71 92 adrenal insufficiency DiseaseClass D000309 218453 142 162 adrenoleukodystrophy SpecificDisease D000326 218453 164 167 ALD SpecificDisease D000326 218453 185 206 adrenomyeloneuropathy SpecificDisease D000326 218453 208 211 AMN SpecificDisease D000326 218453 283 286 AMN SpecificDisease D000326 218453 312 315 ALD SpecificDisease D000326 218453 461 464 ALD SpecificDisease D000326 218453 465 468 AMN SpecificDisease D000326 218453 473 502 pituitary-adrenal abnormality DiseaseClass D010900+D000307 218453 647 650 ALD SpecificDisease D000326 218453 655 658 AMN SpecificDisease D000326 218453 712 733 adrenal insufficiency DiseaseClass D000309 218453 979 982 ALD SpecificDisease D000326 218453 987 990 AMN SpecificDisease D000326 218453 1020 1048 adrenocrotical insufficiency DiseaseClass D000224 218453 1059 1085 endocrinologic dysfunction DiseaseClass D004700 8566952|t|Identification of mutations in the ALD-gene of 20 families with adrenoleukodystrophy/adrenomyeloneuropathy. 8566952|a|Adrenoleukodystrophy (ALD), an X-linked inherited metabolic disorder, is the most frequent inborn peroxisomal disease. It leads to demyelination in the central and peripheral nervous system. Defective beta-oxidation of saturated very long chain fatty acids (VLCFAs; C22 0-C26 0) in peroxisomes has been shown to lead to an accumulation of VLCFAs in leukoid areas of the central nervous system, peripheral nerves, adrenal gland, and blood. The ALD gene has been recently identified and encodes a 745-amino-acid protein. We screened patients with adrenoleukodystrophy/adrenomyeloneuropathy (ALD/AMN) from 20 kindreds for mutations in the ALD gene. Eleven missense and two nonsense mutations, five deletions, and one insertion were detected by direct sequencing of eight reverse transcribed fragments of the ALD-gene mRNA. Four mutations could be shown to be de novo. All mutations could be confirmed in carriers by sequencing genomic DNA. No correlation between the type of mutation and the severity of the phenotype could be observed. The mutations were not detected in the ALD gene of 30 healthy persons.. 8566952 64 84 adrenoleukodystrophy SpecificDisease D000326 8566952 85 106 adrenomyeloneuropathy SpecificDisease D000326 8566952 108 128 Adrenoleukodystrophy SpecificDisease D000326 8566952 130 133 ALD SpecificDisease D000326 8566952 139 176 X-linked inherited metabolic disorder DiseaseClass D008661 8566952 199 225 inborn peroxisomal disease DiseaseClass D018901 8566952 239 297 demyelination in the central and peripheral nervous system DiseaseClass D003711 8566952 553 556 ALD Modifier D000326 8566952 655 675 adrenoleukodystrophy SpecificDisease D000326 8566952 676 697 adrenomyeloneuropathy SpecificDisease D000326 8566952 699 702 ALD SpecificDisease D000326 8566952 703 706 AMN SpecificDisease D000326 8566952 746 749 ALD Modifier D000326 8566952 915 918 ALD Modifier D000326 8566952 1183 1186 ALD Modifier D000326 8128954|t|Gonosomal mosaicism in myotonic dystrophy patients: involvement of mitotic events in (CTG)n repeat variation and selection against extreme expansion in sperm. 8128954|a|Myotonic dystrophy (DM) is caused by abnormal expansion of a polymorphic (CTG) n repeat, located in the DM protein kinase gene. We determined the (CTG) n repeat lengths in a broad range of tissue DNAs from patients with mild, classical, or congenital manifestation of DM. Differences in the repeat length were seen in somatic tissues from single DM individuals and twins. Repeats appeared to expand to a similar extent in tissues originating from the same embryonal origin. In most male patients carrying intermediate- or small-sized expansions in blood, the repeat lengths covered a markedly wider range in sperm. In contrast, male patients with large allele expansions in blood (> 700 CTGs) had similar or smaller repeats in sperm, when detectable. Sperm alleles with > 1, 000 CTGs were not seen. We conclude that DM patients can be considered gonosomal mosaics, i. e e., combined somatic and germ-line tissue mosaics. Most remarkably, we observed multiple cases where the length distributions of intermediate- or small-sized alleles in fathers sperm were significantly different from that in their offsprings blood. Our combined findings indicate that intergenerational length changes in the unstable CTG repeat are most likely to occur during early embryonic mitotic divisions in both somatic and germ-line tissue formation. Both the initial CTG length, the overall number of cell divisions involved in tissue formation, and perhaps a specific selection process in spermatogenesis may influence the dynamics of this process. A model explaining mitotic instability and sex-dependent segregation phenomena in DM manifestation is discussed 8128954 23 41 myotonic dystrophy Modifier D009223 8128954 159 177 Myotonic dystrophy SpecificDisease D009223 8128954 179 181 DM SpecificDisease D009223 8128954 263 265 DM Modifier D009223 8128954 427 429 DM Modifier D009223 8128954 505 507 DM Modifier D009223 8128954 975 977 DM Modifier D009223 8128954 1770 1772 DM Modifier D009223 10712209|t|Combined analysis of hereditary prostate cancer linkage to 1q24-25: results from 772 hereditary prostate cancer families from the International Consortium for Prostate Cancer Genetics. 10712209|a|A previous linkage study provided evidence for a prostate cancer-susceptibility locus at 1q24-25. Subsequent reports in additional collections of families have yielded conflicting results. In addition, evidence for locus heterogeneity has been provided by the identification of other putative hereditary prostate cancer loci on Xq27-28, 1q42-43, and 1p36. The present study describes a combined analysis for six markers in the 1q24-25 region in 772 families affected by hereditary prostate cancer and ascertained by the members of the International Consortium for Prostate Cancer Genetics (ICPCG) from North America, Australia, Finland, Norway, Sweden, and the United Kingdom. Overall, there was some evidence for linkage, with a peak parametric multipoint LOD score assuming heterogeneity (HLOD) of 1. 40 (P =. 01) at D1S212. The estimated proportion of families (alpha) linked to the locus was. 06 (1-LOD support interval. 01-. 12). This evidence was not observed by a nonparametric approach, presumably because of the extensive heterogeneity. Further parametric analysis revealed a significant effect of the presence of male-to-male disease transmission within the families. In the subset of 491 such families, the peak HLOD was 2. In the subset of 491 such families, the peak HLOD was 2. 56 (P =. 0006) and alpha =. 11 (1-LOD support interval. 04-. 19), compared with HLODs of 0 in the remaining 281 families. Within the families with male-to-male disease transmission, alpha increased with the early mean age at diagnosis (< 65 years, alpha =. 19, with 1-LOD support interval. 06-. 34) and the number of affected family members (five or more family members, alpha =. 15, with 1-LOD support interval. 04-. 28). The highest value of alpha was observed for the 48 families that met all three criteria (peak HLOD = 2. 25, P =. 001, alpha =. 29, with 1-LOD support interval. 08-. 53). These results support the finding of a prostate cancer-susceptibility gene linked to 1q24-25, albeit in a defined subset of prostate cancer families. Although HPC1 accounts for only a small proportion of all families affected by hereditary prostate cancer, it appears to play a more prominent role in the subset of families with several members affected at an early age and with male-to-male disease transmission. 10712209 21 47 hereditary prostate cancer Modifier C537243 10712209 85 111 hereditary prostate cancer Modifier C537243 10712209 159 174 Prostate Cancer Modifier D011471 10712209 234 249 prostate cancer Modifier D011471 10712209 478 504 hereditary prostate cancer Modifier C537243 10712209 655 681 hereditary prostate cancer SpecificDisease C537243 10712209 749 764 Prostate Cancer Modifier D011471 10712209 2109 2124 prostate cancer Modifier D011471 10712209 2194 2209 prostate cancer Modifier D011471 10712209 2299 2325 hereditary prostate cancer SpecificDisease C537243 8466512|t|A novel disease with deficiency of mitochondrial very-long-chain acyl-CoA dehydrogenase. 8466512|a|Palmitoyl-CoA dehydrogenase activity in skin fibroblasts from seven patients with unidentified defects of fatty acid oxidation was measured in the presence and absence of antibodies against medium-chain, long-chain, and very-long-chain acyl-CoA dehydrogenases (VLCAD). Two of the patients, 4-5 month old boys, were found to have a novel disease, VLCAD deficiency, as judged from the results of very low palmitoyl-CoA dehydrogenase activity and the lack of immunoreactivity toward antibody raised to purified VLCAD.. 8466512 21 87 deficiency of mitochondrial very-long-chain acyl-CoA dehydrogenase SpecificDisease C536353 8466512 435 451 VLCAD deficiency SpecificDisease C536353 1327525|t|Inherited WT1 mutation in Denys-Drash syndrome. 1327525|a|Patients with the Denys-Drash syndrome (Wilms tumor, genital anomalies, and nephropathy) have been demonstrated to carry de novo constitutional mutations in WT1, the Wilms tumor gene at chromosome 11p13. We report three new cases, two carrying a previously described WT1 exon 9 mutation and one with a novel WT1 exon 8 mutation. However, unlike patients in previous reports, one of our three patients inherited the affected allele from his phenotypically unaffected father. This observation indicates that the WT1 exon 9 mutation affecting 394Arg demonstrated in over one-half of the patients with the Denys-Drash syndrome may exhibit incomplete penetrance. Consequently, familial studies in patients affected by this syndrome are recommended.. 1327525 26 46 Denys-Drash syndrome SpecificDisease D030321 1327525 66 86 Denys-Drash syndrome SpecificDisease D030321 1327525 88 99 Wilms tumor SpecificDisease D009396 1327525 101 118 genital anomalies DiseaseClass D014564 1327525 124 135 nephropathy SpecificDisease D007674 1327525 214 225 Wilms tumor Modifier D009396 1327525 650 670 Denys-Drash syndrome SpecificDisease D030321 2008213|t|Glucose/galactose malabsorption caused by a defect in the Na+/glucose cotransporter. 2008213|a|Glucose/galactose malabsorption (GGM) is an autosomal recessive disease manifesting within the first weeks of life and characterized by a selective failure to absorb dietary glucose and galactose from the intestine. The consequent severe diarrhoea and dehydration are usually fatal unless these sugars are eliminated from the diet. Intestinal biopsies of GGM patients have revealed a specific defect in Na (+) -dependent absorption of glucose in the brush border. Normal glucose absorption is mediated by the Na +/glucose cotransporter in the brush border membrane of the intestinal epithelium. Cellular influx is driven by the transmembrane Na + electrochemical potential gradient; thereafter the sugar moves to the blood across the basolateral membrane via the facilitated glucose carrier. We have previously cloned and sequenced a Na +/glucose cotransporter from normal human ileum and shown that this gene, SGLT1, resides on the distal q arm of chromosome 22. We have now amplified SGLT1 complementary DNA and genomic DNA from members of a family affected with GGM by the polymerase chain reaction. Sequence analysis of the amplified products has revealed a single missense mutation in SGLT1 which cosegregates with the GGM phenotype and results in a complete loss of Na (+) -dependent glucose transport in Xenopus oocytes injected with this complementary RNA.. 2008213 0 31 Glucose/galactose malabsorption SpecificDisease OMIM:606824 2008213 85 116 Glucose/galactose malabsorption SpecificDisease OMIM:606824 2008213 118 121 GGM SpecificDisease OMIM:606824 2008213 129 156 autosomal recessive disease DiseaseClass D030342 2008213 323 332 diarrhoea SpecificDisease D003967 2008213 337 348 dehydration SpecificDisease D003681 2008213 440 443 GGM Modifier OMIM:606824 2008213 1150 1153 GGM SpecificDisease OMIM:606824 2008213 1309 1312 GGM Modifier OMIM:606824 10639175|t|Atm and Bax cooperate in ionizing radiation-induced apoptosis in the central nervous system. 10639175|a|Ataxia-telangiectasia is a hereditary multisystemic disease resulting from mutations of ataxia telangiectasia, mutated (ATM) and is characterized by neurodegeneration, cancer, immune defects, and hypersensitivity to ionizing radiation. The molecular details of ATM function in the nervous system are unclear, although the neurological lesion in ataxia-telangiectasia becomes apparent early in life, suggesting a developmental origin. The central nervous system (CNS) of Atm-null mice shows a pronounced defect in apoptosis induced by genotoxic stress, suggesting ATM functions to eliminate neurons with excessive genomic damage. Here, we report that the death effector Bax is required for a large proportion of Atm-dependent apoptosis in the developing CNS after ionizing radiation (IR). Although many of the same regions of the CNS in both Bax-/- and Atm-/- mice were radioresistant, mice nullizygous for both Bax and Atm showed additional reduction in IR-induced apoptosis in the CNS. Therefore, although the major IR-induced apoptotic pathway in the CNS requires Atm and Bax, a p53-dependent collateral pathway exists that has both Atm- and Bax-independent branches. Further, Atm- and Bax-dependent apoptosis in the CNS also required caspase-3 activation. These data implicate Bax and caspase-3 as death effectors in neurodegenerative pathways.. 10639175 93 114 Ataxia-telangiectasia SpecificDisease D001260 10639175 120 152 hereditary multisystemic disease DiseaseClass D030342 10639175 181 202 ataxia telangiectasia SpecificDisease D001260 10639175 242 259 neurodegeneration DiseaseClass D019636 10639175 261 267 cancer DiseaseClass D009369 10639175 269 283 immune defects DiseaseClass D007154 10639175 289 327 hypersensitivity to ionizing radiation DiseaseClass D004194 10639175 415 434 neurological lesion DiseaseClass D019636 10639175 438 459 ataxia-telangiectasia SpecificDisease D001260 1301190|t|Novel Tay-Sachs disease mutations from China. 1301190|a|We describe three HEXA mutations associated with infantile Tay-Sachs disease (TSD) in three unrelated nonconsanguineous Chinese families. Novel mutations were found in two of these families. The third is a previously reported mutation (G-- > A transition at nt 1444) (Nakano et al., 1988). Direct sequencing of PCR products identified a novel insertion of an A after nt 547 in family 1. This change generates an early termination codon 6 bp downstream from the insertion site. Allele-specific oligonucleotide hybridization confirmed homozygosity in the proband. Single strand conformational polymorphism analysis and direct sequencing of amplified exon 13 revealed a T-- > C transition at nt 1453 with the corresponding amino acid substitution W485R in the second family. This mutation creates an Fnu4HI restriction site. The proband is homozygous for this allele. When the site-specific mutagenized alpha cDNA carrying the T-- > C transition at nt 1453 was expressed in COS 1 cells hexosaminidase S activity was not detectable above background. A G-- > A transition at nt 1444 (exon 13) corresponding to the E482K substitution was found in the third family. This mutation occurs at a CpG dinucleotide. It has been reported in an Italian TSD proband and causes defective intracellular transport of the alpha-subunit from the rough endoplasmic reticulum to the Golgi apparatus. 1301190 6 23 Tay-Sachs disease Modifier D013661 1301190 105 122 Tay-Sachs disease SpecificDisease D013661 1301190 124 127 TSD SpecificDisease D013661 1301190 1284 1287 TSD Modifier D013661 3348216|t|Glucose 6-phosphate dehydrogenase deficiency and incidence of hematologic malignancy. 3348216|a|We have evaluated the hypothesis of a negative association between glucose 6-phosphate dehydrogenase (G6PD) deficiency and cancer in a cohort of 481 Sardinian males with hematological malignancies. The frequency of G6PD deficiency in the patients was not different from the incidence in a group of 16, 219 controls. The same conclusion resulted from the comparison of the frequency of expression of the GdB gene in 23 heterozygous women having a clonal hematologic disease and a control group of 37 healthy heterozygotes. Therefore at present there is no evidence that G6PD deficiency has a protective effect against development of hematologic neoplasms.. 3348216 0 44 Glucose 6-phosphate dehydrogenase deficiency SpecificDisease D005955 3348216 62 84 hematologic malignancy DiseaseClass D019337 3348216 153 204 glucose 6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 3348216 209 215 cancer DiseaseClass D009369 3348216 256 282 hematological malignancies DiseaseClass D019337 3348216 301 316 G6PD deficiency SpecificDisease D005955 3348216 539 558 hematologic disease DiseaseClass D006402 3348216 655 670 G6PD deficiency SpecificDisease D005955 3348216 718 739 hematologic neoplasms DiseaseClass D019337 7951316|t|A physical map and candidate genes in the BRCA1 region on chromosome 17q12-21. 7951316|a|We have constructed a physical map of a 4 cM region on chromosome 17q12-21 that contains the hereditary breast and ovarian cancer gene BRCA1. The map comprises a contig of 137 overlapping yeast artificial chromosomes and P1 clones, onto which we have placed 112 PCR markers. We have localized more than 20 genes on this map, ten of which had not been mapped to the region previously, and have isolated 30 cDNA clones representing partial sequences of as yet unidentified genes. Two genes that lie within a narrow region defined by meiotic breakpoints in BRCA1 patients have been sequenced in breast cancer patients without revealing any deleterious mutations. These new reagents should facilitate the identification of BRCA1.. 7951316 172 208 hereditary breast and ovarian cancer Modifier D061325 7951316 671 684 breast cancer Modifier D001943 10404839|t|Sulfate transport is not impaired in pendred syndrome thyrocytes. 10404839|a|Pendred syndrome is the most common form of syndromic deafness, characterized by dyshormonogenic goiter associated with sensory-neural deafness. The gene responsible for the disease (PDS) has been cloned, but its function is as yet unknown and the connection between thyroid goiter and sensory-neural deafness remains an enigma. PDS codes for a novel protein, pendrin, which is closely related to a number of sufate transporters. Mechanisms by which abnormal sulfate transport could deleteriously affect iodide organification have been proposed. We tested sulfate transport in thyrocytes obtained from Pendred syndrome patients and found that it was not defective. This suggests that pendrin in fact may not be a sulfate transporter, and emphasizes the importance of functional studies on this novel protein.. 10404839 37 53 pendred syndrome Modifier C536648 10404839 66 82 Pendred syndrome SpecificDisease C536648 10404839 110 128 syndromic deafness DiseaseClass D003638 10404839 147 169 dyshormonogenic goiter SpecificDisease D006042 10404839 186 209 sensory-neural deafness DiseaseClass D006319 10404839 249 252 PDS SpecificDisease C536648 10404839 333 347 thyroid goiter DiseaseClass D006042 10404839 352 375 sensory-neural deafness DiseaseClass D006319 10404839 668 684 Pendred syndrome Modifier C536648 10430841|t|Spinal xanthomatosis: a variant of cerebrotendinous xanthomatosis. 10430841|a|We describe seven Dutch patients from six families with a slowly progressive, mainly spinal cord syndrome that remained for many years the sole expression of cerebrotendinous xanthomatosis (CTX). MRI demonstrated white matter abnormalities in the lateral and dorsal columns of the spinal cord. Post-mortem examination of one of the patients showed extensive myelin loss in these columns. An array of genotypes was found in these patients. We conclude that spinal xanthomatosis is a clinical and radiological separate entity of CTX that should be included in the differential diagnosis of chronic myelopathy.. 10430841 0 20 Spinal xanthomatosis SpecificDisease D014973 10430841 35 65 cerebrotendinous xanthomatosis DiseaseClass D019294 10430841 152 172 spinal cord syndrome SpecificDisease D013118 10430841 225 255 cerebrotendinous xanthomatosis SpecificDisease D019294 10430841 257 260 CTX SpecificDisease D019294 10430841 280 306 white matter abnormalities DiseaseClass D002493 10430841 523 543 spinal xanthomatosis SpecificDisease D014973 10430841 594 597 CTX SpecificDisease D019294 10430841 655 673 chronic myelopathy DiseaseClass D002908+D013118 10827109|t|The exon 13 duplication in the BRCA1 gene is a founder mutation present in geographically diverse populations. The BRCA1 Exon 13 Duplication Screening Group. 10827109|a|Recently, a 6-kb duplication of exon 13, which creates a frameshift in the coding sequence of the BRCA1 gene, has been described in three unrelated U. S S. families of European ancestry and in one Portuguese family. Here, our goal was to estimate the frequency and geographic diversity of carriers of this duplication. To do this, a collaborative screening study was set up that involved 39 institutions from 19 countries and included 3, 580 unrelated individuals with a family history of the disease and 934 early-onset breast and/or ovarian cancer cases. A total of 11 additional families carrying this mutation were identified in Australia (1), Belgium (1), Canada (1), Great Britain (6), and the United States (2). Haplotyping showed that they are likely to derive from a common ancestor, possibly of northern British origin. Our results demonstrate that it is strongly advisable, for laboratories carrying out screening either in English-speaking countries or in countries with historical links with Britain, to include within their BRCA1 screening protocols the polymerase chain reaction-based assay described in this report. 10827109 679 707 breast and/or ovarian cancer Modifier D061325 7493024|t|Germline mutations of the BRCA1 gene in breast and ovarian cancer families provide evidence for a genotype-phenotype correlation. 7493024|a|Mutations in the BRCA1 gene, discovered in 1994, are associated with an 80-90% lifetime risk of breast cancer. We have analysed 60 families with a history of breast and/or ovarian cancer for germline mutations in BRCA1. Twenty-two different mutations were detected in 32 families (53%), of which 14 are previously unreported. We observed a significant correlation between the location of the mutation in the gene and the ratio of breast to ovarian cancer incidence within each family. Our data suggest a transition in risk such that mutations in the 3 third of the gene are associated with a lower proportion of ovarian cancer. Haplotype analysis supports previous data which suggest some BRCA1 mutation carriers have common ancestors; however, we have found at least two examples where recurrent mutations appear to have arisen independently.. 7493024 40 65 breast and ovarian cancer Modifier D061325 7493024 226 239 breast cancer SpecificDisease D001943 7493024 288 316 breast and/or ovarian cancer CompositeMention D061325 7493024 560 584 breast to ovarian cancer Modifier D001943|D010051 7493024 742 756 ovarian cancer SpecificDisease D010051 10767347|t|Inactivation of the Friedreich ataxia mouse gene leads to early embryonic lethality without iron accumulation. 10767347|a|Friedreich ataxia (FRDA), the most common autosomal recessive ataxia, is caused in almost all cases by homozygous intronic expansions resulting in the loss of frataxin, a mitochondrial protein conserved through evolution, and involved in mitochondrial iron homeostasis. Yeast knockout models, and histological and biochemical data from patient heart biopsies or autopsies indicate that the frataxin defect causes a specific iron-sulfur protein deficiency and mitochondrial iron accumulation leading to the pathological changes. Affected human tissues are rarely available to further examine this hypothesis. To study the mechanism of the disease, we generated a mouse model by deletion of exon 4 leading to inactivation of the Frda gene product. We show that homozygous deletions cause embryonic lethality a few days after implantation, demonstrating an important role for frataxin during early development. These results suggest that the milder phenotype in humans is due to residual frataxin expression associated with the expansion mutations. Surprisingly, in the frataxin knockout mouse, no iron accumulation was observed during embryonic resorption, suggesting that cell death could be due to a mechanism independent of iron accumulation.. 10767347 20 37 Friedreich ataxia Modifier D005621 10767347 64 83 embryonic lethality SpecificDisease D020964 10767347 111 128 Friedreich ataxia SpecificDisease D005621 10767347 130 134 FRDA SpecificDisease D005621 10767347 153 179 autosomal recessive ataxia DiseaseClass D013132 10767347 535 565 iron-sulfur protein deficiency SpecificDisease OMIM:255125 10767347 838 842 Frda Modifier D005621 10767347 897 916 embryonic lethality SpecificDisease D020964 1301189|t|A glycine250--> aspartate substitution in the alpha-subunit of hexosaminidase A causes juvenile-onset Tay-Sachs disease in a Lebanese-Canadian family. 1301189|a|The mutation causing juvenile Tay-Sachs disease (TSD) in two sibs of Lebanese-Maronite origin is described. An mRNA-containing extract of cultured fibroblasts obtained from one of the probands was used as a template to amplify the coding sequence of the hexosaminidase A (Hex A) alpha-subunit. Sequencing of amplified cDNA fragments revealed a single alteration, guanine to adenine at nt 749 creating a G250D mutation. The mutation introduces a new recognition site for the restriction enzyme Eco RV, permitting identification of heterozygotes for this allele following PCR amplification and Eco RV digestion of exon 7 sequences from genomic DNA templates. In order to test the effect of this substitution, an in vitro mutagenized cDNA construct was introduced into a mammalian expression vector and transfected into monkey Cos-1 cells separately or along with a beta-cDNA expression vector. When the mutant alpha-cDNA was the only gene introduced into COS cells no enzymatic activity above endogenous COS cell activity was detected. Cotransfection of normal alpha-cDNA and beta-cDNA followed by immunoprecipitation of human Hex A resulted in 20-fold increase in the ratio between positive and negative (mock transfection) control values. This allowed the detection of some residual activity (12% of the positive control) when the mutant alpha-cDNA replaced its wild-type counterpart. The predicted protein environment in which the mutation occurs is compared to that of the adult-onset Tay-Sachs disease mutation caused by a Gly269-- > Ser substitution in exon 7. (ABSTRACT TRUNCATED AT 250 WORDS). 1301189 102 119 Tay-Sachs disease SpecificDisease D013661 1301189 181 198 Tay-Sachs disease SpecificDisease D013661 1301189 200 203 TSD SpecificDisease D013661 1301189 1638 1655 Tay-Sachs disease Modifier D013661 8252631|t|Restriction of ocular fundus lesions to a specific subgroup of APC mutations in adenomatous polyposis coli patients. 8252631|a|In humans, alteration of the tumor suppressor gene, APC, causes adenomatous polyposis coli, a condition causing predisposition to colorectal cancer. The syndrome inconsistently associates characteristic patches of congenital hypertrophy of the retinal pigment epithelium (CHRPE). Ocular examination revealed that patients expressing CHRPE tend to cluster within specific families. The exact APC mutation was identified in 42 unrelated patients. In all cases these mutations were predicted to lead to the synthesis of a truncated protein. The extent of CHRPE was found to be dependent on the position of the mutation along the coding sequence. CHRPE lesions are almost always absent if the mutation occurs before exon 9, but are systematically present if it occurs after this exon. Thus, the range of phenotypic expression observed among affected patients may result in part from different allelic manifestations of APC mutations.. 8252631 63 66 APC Modifier D011125 8252631 80 106 adenomatous polyposis coli Modifier D011125 8252631 146 151 tumor Modifier D009369 8252631 169 172 APC Modifier D011125 8252631 181 207 adenomatous polyposis coli SpecificDisease D011125 8252631 247 264 colorectal cancer SpecificDisease D015179 8252631 331 387 congenital hypertrophy of the retinal pigment epithelium SpecificDisease D012164 8252631 389 394 CHRPE SpecificDisease D012164 8252631 450 455 CHRPE SpecificDisease D012164 8252631 508 511 APC Modifier D011125 8252631 669 674 CHRPE SpecificDisease D012164 8252631 760 765 CHRPE Modifier D012164 8252631 1032 1035 APC Modifier D011125 7759076|t|Linkage analysis of 26 Canadian breast and breast-ovarian cancer families. 7759076|a|We have examined 26 Canadian families with hereditary breast or ovarian cancer for linkage to markers flanking the BRCA1 gene on chromosome 17q12-q21. Of the 15 families that contain cases of ovarian cancer, 94% were estimated to be linked to BRCA1. In contrast, there was no overall evidence of linkage in the group of 10 families with breast cancer without ovarian cancer. A genetic recombinant in a breast-ovarian cancer family indicates a placement of BRCA1 telomeric to D17S776, and helps to define the region of assignment of the cancer susceptibility gene. Other cancers of interest that appeared in the BRCA1-linked families included primary peritoneal cancer, cancer of the fallopian tube, and malignant melanoma.. 7759076 32 64 breast and breast-ovarian cancer Modifier D001943|D061325 7759076 118 153 hereditary breast or ovarian cancer CompositeMention D061325 7759076 267 281 ovarian cancer SpecificDisease D010051 7759076 412 425 breast cancer SpecificDisease D001943 7759076 434 448 ovarian cancer SpecificDisease D010051 7759076 477 498 breast-ovarian cancer Modifier D061325 7759076 611 617 cancer Modifier D009369 7759076 645 652 cancers DiseaseClass D009369 7759076 717 742 primary peritoneal cancer SpecificDisease D010534 7759076 744 772 cancer of the fallopian tube SpecificDisease D005185 7759076 778 796 malignant melanoma SpecificDisease D008545 1248000|t|Malignant neoplasms in the families of patients with ataxia-telangiectasia. 1248000|a|Ataxia-telangiectasia (A-T) is an autosomal recessive syndrome associated with a greatly increased incidence of malignant neoplasms in homozygous affected individuals. Heterozygotes for the gene for A-T are thought to comprise about 1% of the general population and, therefore, it is important to know whether this gene also predisposes the heterozygous carrier to cancers. Heterozygous carriers of this gene are common among the close relatives of patients with A-T, although individual carriers cannot be identified by any clinical criterion or laboratory test. For this reason, we compared the incidence of death from malignant neoplasms in 2 families of patients with A-T to that expected in a random sample of the general population. There were 59 deaths from malignant neoplasms in relatives dying before age 75, compared to 42. 6 expected (p less than 0. 02). For A-T heterozygotes younger than age 45, the risk of dying from a malignant neoplasm was estimated to be greater than 5 times the risk for the general population. A-T heterozygotes may comprise more than 5% of all persons dying from a cancer before age 45. The incidence of ovarian, gastric, and biliary system carcinomas and of leukemia and lymphoma was increased in these A-T families. Other neoplasms that may be associated with this gene in heterozygotes include pancreatic, basal cell, colonic, breast, and cervical carcinomas. 1248000 0 19 Malignant neoplasms DiseaseClass D009369 1248000 53 74 ataxia-telangiectasia SpecificDisease D001260 1248000 76 97 Ataxia-telangiectasia SpecificDisease D001260 1248000 99 102 A-T SpecificDisease D001260 1248000 110 138 autosomal recessive syndrome DiseaseClass D030342 1248000 188 207 malignant neoplasms DiseaseClass D009369 1248000 275 278 A-T SpecificDisease D001260 1248000 441 448 cancers DiseaseClass D009369 1248000 539 542 A-T SpecificDisease D001260 1248000 697 716 malignant neoplasms DiseaseClass D009369 1248000 748 751 A-T SpecificDisease D001260 1248000 841 860 malignant neoplasms DiseaseClass D009369 1248000 947 950 A-T Modifier D001260 1248000 1011 1029 malignant neoplasm DiseaseClass D009369 1248000 1108 1111 A-T Modifier D001260 1248000 1180 1186 cancer DiseaseClass D009369 1248000 1219 1266 ovarian, gastric, and biliary system carcinomas CompositeMention D010051|D013274|D001661 1248000 1274 1282 leukemia DiseaseClass D007938 1248000 1287 1295 lymphoma DiseaseClass D008223 1248000 1319 1322 A-T Modifier D001260 1248000 1339 1348 neoplasms DiseaseClass D009369 1248000 1412 1476 pancreatic, basal cell, colonic, breast, and cervical carcinomas CompositeMention D010190|D002280|D015179|D001943|D002583 2309142|t|Molecular genetics of PKU in eastern Europe: a nonsense mutation associated with haplotype 4 of the phenylalanine hydroxylase gene. 2309142|a|Phenylketonuria (PKU) is a genetic disorder secondary to a deficiency of hepatic phenylalanine hydroxylase (PAH). Several mutations in the PAH gene have recently been reported, and linkage disequilibrium was observed between RFLP haplotypes and specific mutations. A new molecular lesion has been identified in exon 7 of the PAH gene in a Hungarian PKU patient by direct sequencing of PCR-amplified DNA. The C-to-T transition causes the substitution of Arg243 to a termination codon, and the mutant allele is associated with haplotype 4 of the PAH gene. The mutation is present in two of nine mutant haplotype 4 alleles among Eastern Europeans and is not present among Western Europeans and Asians. The rarity of this mutant allele and its restricted geographic distribution suggest that the mutational event occurred recently on a normal haplotype 4 background in Eastern Europe.. 2309142 22 25 PKU SpecificDisease D010661 2309142 132 147 Phenylketonuria SpecificDisease D010661 2309142 149 152 PKU SpecificDisease D010661 2309142 159 175 genetic disorder DiseaseClass D030342 2309142 191 238 deficiency of hepatic phenylalanine hydroxylase SpecificDisease OMIM:261600 2309142 403 419 molecular lesion DiseaseClass D030342 2309142 481 484 PKU Modifier D010661 8575748|t|Isolation of the mouse homologue of BRCA1 and genetic mapping to mouse chromosome 11. 8575748|a|The BRCA1 gene is in large part responsible for hereditary human breast and ovarian cancer. Here we report the isolation of the murine Brca1 homologue cDNA clones. In addition, we identified genomic P1 clones that contain most, if not all, of the mouse Brca1 locus. DNA sequence analysis revealed that the mouse and human coding regions are 75% identical at the nucleotide level while the predicted amino acid identity is only 58%. A DNA sequence variant in the Brca1 locus was identified and used to map this gene on a (Mus m. musculus Czech II x C57BL/KsJ) F1 x C57BL/KsJ intersubspecific backcross to distal mouse chromosome 11. The mapping of this gene to a region highly syntenic with human chromosome 17, coupled with Southern and Northern analyses, confirms that we isolated the murine Brca1 homologue rather than a related RING finger gene. The isolation of the mouse Brca1 homologue will facilitate the creation of mouse models for germline BRCA1 defects.. 8575748 134 176 hereditary human breast and ovarian cancer CompositeMention D061325 8575748 1036 1049 BRCA1 defects SpecificDisease OMIM:604370 6783144|t|Human deficiency of the sixth component of complement in a patient with meningococcal meningitis and no haemostasis abnormality. 6783144|a|A case of human complete C6 deficiency is reported. The patient, a 31 year old white male, was seen on the occasion of an isolated episode of meningococcal meningitis. Serum complement hemolytic and bactericidal activities were lacking and could be restored to normal by addition of appropriate amounts of purified C6. No hemostatic abnormalities were observed.. 6783144 6 53 deficiency of the sixth component of complement SpecificDisease OMIM:612446 6783144 72 96 meningococcal meningitis SpecificDisease D008585 6783144 104 127 haemostasis abnormality DiseaseClass D020141 6783144 154 167 C6 deficiency SpecificDisease OMIM:612446 6783144 271 295 meningococcal meningitis SpecificDisease D008585 6783144 451 475 hemostatic abnormalities DiseaseClass D020141 1346773|t|The Wiskott-Aldrich syndrome: refinement of the localization on Xp and identification of another closely linked marker locus, OATL1. 1346773|a|The Wiskott-Aldrich syndrome (WAS) has previously been mapped to the proximal short arm of the X chromosome between the DXS14 and DXS7 loci. In this study, further segregation analysis has been performed using a newly identified WAS family as well as an additional marker probe, HOATL1. The results indicate close linkage between the WAS and OATL1 loci (Z = 6. 08 at theta = 0. 00) and localize the TIMP, OATL1, DXS255, and WAS loci distal to DXS146 and the OATL1 and WAS loci proximal to TIMP. These linkage data narrow the boundaries within which the WAS locus maps to the chromosomal region bracketed by TIMP and DXS146 and support the loci order Xpter-DXS7-TIMP- (OATL1, WAS, DXS255) -DXS146. 1346773 4 28 Wiskott-Aldrich syndrome SpecificDisease D014923 1346773 137 161 Wiskott-Aldrich syndrome SpecificDisease D014923 1346773 163 166 WAS SpecificDisease D014923 1346773 362 365 WAS Modifier D014923 1346773 467 470 WAS SpecificDisease D014923 1346773 557 560 WAS Modifier D014923 1346773 601 604 WAS Modifier D014923 1346773 686 689 WAS Modifier D014923 318684|t|Hereditary deficiency of the third component of complement in a child with fever, skin rash, and arthralgias: response to transfusion of whole blood. 318684|a|A previously well 34-month-old male presenting with fever, skin rash, and arthralgias was found to lack C3 by immunochemical (undetectable) and hemolytic (1% normal) assays. No infectious agent could be demonstrated. Protein levels of Clq. C4, C5, properdin, and C3b-INA and hemolytic activities of complement components C1 to C9 except C3 were normal or elevated; total hemolytic complement activity was 13% of normal and was reconstituted by purified C3. Properdin factor B was 702 (normal 175 to 275) mug/ml, and was not cleaver upon addition of zymosan or cobra venom factor. The serum had normal immune adherence activity, but was deficient in ability to opsonize Candida albicans for uptake and Escherichia coli for killing by neurophils, generate neutrophil chemotactic factors and inhibit the growth of E. coli; these activities were restored by purified C3. A transfusion of 320 ml 1-hour-old normal whole blood on the fifty-second day resulted in transitory elevation of the C3 level to 25 mg/dl with a fall-off (approximately 2 1/2% per hour) to undetectable levels by 69 hours; it was followed by disappearance of the skin rash and arthralgias and return to normal of the previously elevated temperature and CRP levels. C3 levels in family members (seven of 24 half-normal), lack of anti-C3 activity, normal C3b-INA levels and a normal rate of catabolism of transfused C3 indicated that the deficiency was inherited with autosomal codominance and involved decreased synthesis of C3. Thus, this child is a unique individual with inherited C3 deficiency presenting with absence of repeated infections, whose symptoms of fever, skin rash, and arthralgia were abated by whole blood transfusion.. 318684 0 58 Hereditary deficiency of the third component of complement SpecificDisease OMIM:613779 318684 75 80 fever DiseaseClass D005334 318684 82 91 skin rash DiseaseClass D005076 318684 97 108 arthralgias DiseaseClass D018771 318684 202 207 fever DiseaseClass D005334 318684 209 218 skin rash DiseaseClass D005076 318684 224 235 arthralgias DiseaseClass D018771 318684 1280 1289 skin rash DiseaseClass D005076 318684 1294 1305 arthralgias DiseaseClass D018771 318684 1618 1643 decreased synthesis of C3 DiseaseClass OMIM:613779 318684 1690 1713 inherited C3 deficiency SpecificDisease OMIM:613779 318684 1780 1785 fever DiseaseClass D005334 318684 1787 1796 skin rash DiseaseClass D005076 318684 1802 1812 arthralgia DiseaseClass D018771 2544995|t|Cloning of breakpoints of a chromosome translocation identifies the AN2 locus. 2544995|a|Chromosome translocations involving 11p13 have been associated with familial aniridia in two kindreds highlighting the chromosomal localization of the AN2 locus. This locus is also part of the WAGR complex (Wilms tumor, aniridia, genitourinary abnormalities, and mental retardation). In one kindred, the translocation is associated with a deletion, and probes for this region were used to identify and clone the breakpoints of the translocation in the second kindred. Comparison of phage restriction maps exclude the presence of any sizable deletion in this case. Sequences at the chromosome 11 breakpoint are conserved in multiple species, suggesting that the translocation falls within the AN2 gene.. 2544995 147 164 familial aniridia SpecificDisease D015783 2544995 272 276 WAGR Modifier D017624 2544995 286 297 Wilms tumor SpecificDisease D009396 2544995 299 307 aniridia SpecificDisease D015783 2544995 309 336 genitourinary abnormalities DiseaseClass D014564 2544995 342 360 mental retardation DiseaseClass D008607 10923035|t|Study of the voltage-gated sodium channel beta 1 subunit gene (SCN1B) in the benign familial infantile convulsions syndrome (BFIC). 10923035|a|Benign familial infantile convulsions (BFIC) is a rare autosomal dominant epilepsy syndrome. This syndrome has been recently described in Italian and French pedigrees. Patients present with partial, then generalized seizures, with onset at age three months. The seizures usually spontaneously cease after one year without treatment, leaving no neurological abnormalities. We have mapped BFIC to chromosome 19q in five Italian pedigrees. The sodium channel beta1 subunit gene (SCN1B) maps to this candidate region and has been shown to be involved in one Australian pedigree with generalized epilepsy and febrile seizures " plus " (GEFS +). In this family, a missense mutation in SCN1B cosegregates with the GEFS + phenotype. BFIC and GEFS + have clinical features in common, therefore SCN1B is a candidate gene for BFIC. We studied SCN1B exons 1, 2, 3, 4, and 5, using four SSCP methods in 10 Caucasian BFIC probands of Western Europe. We found no exon variants. One variant was identified in intron 5 (IVS5-10C > G), which did not segregate with BFIC and was observed in 9. 2% controls. A second variant in intron 5 was identified (IVS5 + 30G > A). It was rare, as not observed in controls, but not segregating with the BFIC phenotype. 10923035 77 123 benign familial infantile convulsions syndrome SpecificDisease D020936 10923035 125 129 BFIC SpecificDisease D020936 10923035 132 169 Benign familial infantile convulsions SpecificDisease D020936 10923035 171 175 BFIC SpecificDisease D020936 10923035 187 223 autosomal dominant epilepsy syndrome DiseaseClass D030342+D004827 10923035 476 502 neurological abnormalities DiseaseClass D009461 10923035 519 523 BFIC SpecificDisease D020936 10923035 711 761 generalized epilepsy and febrile seizures plus SpecificDisease D004829+D003294 10923035 857 861 BFIC SpecificDisease D020936 10923035 947 951 BFIC SpecificDisease D020936 10923035 1035 1039 BFIC Modifier D020936 10923035 1179 1183 BFIC SpecificDisease D020936 10923035 1353 1357 BFIC Modifier D020936 7696601|t|Brain disease and molecular analysis in myotonic dystrophy. 7696601|a|Abnormal amplification of a CTG repeat on chromosome 19 is the molecular basis of myotonic dystrophy (DM). Expansion of the repeat has been correlated with severity of several clinical features of the disease. We performed extensive cognitive testing, cerebral magnetic resonance imaging (MRI) and a molecular analysis in 28 cases of DM to determine the relationship between the molecular defect and brain disease. Performance in two or more cognitive tests was pathological in 10 cases. Fourteen patients had subcortical white matter lesions on MRI, 14 had cerebral atrophy. Amplification of the CTG repeat showed a strong correlation with cognitive test deficits when exceeding a length of over 1000 trinucleotides. MRI lesions were associated with impaired psychometric performance, but MRI and molecular findings were only weakly related. Disease duration influenced the appearance and amount of white matter lesions on MRI. Quantification of CTG repeat size may allow an early estimate on the probability of brain involvement in DM; cognitive dysfunction is associated with white matter lesions and cerebral atrophy later on in the course.. 7696601 0 13 Brain disease DiseaseClass D001927 7696601 40 58 myotonic dystrophy SpecificDisease D009223 7696601 142 160 myotonic dystrophy SpecificDisease D009223 7696601 162 164 DM SpecificDisease D009223 7696601 394 396 DM SpecificDisease D009223 7696601 460 473 brain disease DiseaseClass D001927 7696601 582 602 white matter lesions DiseaseClass D056784 7696601 618 634 cerebral atrophy SpecificDisease D001284 7696601 960 980 white matter lesions DiseaseClass D056784 7696601 1094 1096 DM SpecificDisease D009223 7696601 1098 1119 cognitive dysfunction DiseaseClass D003072 7696601 1139 1159 white matter lesions DiseaseClass D056784 7696601 1164 1180 cerebral atrophy SpecificDisease D001284 10528853|t|A highly accurate, low cost test for BRCA1 mutations. 10528853|a|The hereditary breast and ovarian cancer syndrome is associated with a high frequency of BRCA1 mutations. However, the widespread use of BRCA1 testing has been limited to date by three principal concerns the fear of loss of health and life insurance, the uncertain clinical value of a positive test result, and the current lack of an inexpensive and sensitive screening test for BRCA1 mutations. We have developed an inexpensive system for gene mutational scanning, based on a combination of extensive multiplex PCR amplification and two dimensional electrophoresis. The efficiency of this system, as a screening test for BRCA1 mutations, was evaluated in a panel of 60 samples from high risk women, 14 of which contained a previously identified mutation in BRCA1. All 14 mutations were identified, as well as an additional five that had previously escaped detection. In addition to the 19 mutations, a total of 15 different polymorphic variants were scored, most of which were recurring. All were confirmed by nucleotide sequencing. The cost of screening per sample was calculated to be approximately US $ 70 for the manual technique used in this study, and may be reduced to approximately US $ 10 with the introduction of commercially available PCR robotics and fluorescent imaging. Implementation of this method of mutation screening in the research and clinical setting should permit rapid accrual of quantitative data on genotype-phenotype associations for the evaluation of diagnostic testing.. 10528853 58 103 hereditary breast and ovarian cancer syndrome CompositeMention D061325 10083733|t|Germline mutations of the APC gene in Korean familial adenomatous polyposis patients. 10083733|a|We extensively analyzed genomic DNA and messenger RNA (mRNA) from 62 unrelated Korean patients with familial adenomatous polyposis (FAP) for identification of germline adenomatous polyposis coli (APC) gene mutations. We adopted both single-strand conformation polymorphism (SSCP) analysis and a method of analysis involving the reverse transcription-polymerase chain reaction (RT-PCR) followed by a protein truncation test (PTT). DNA sequencing confirmed all alterations represented by aberrant bands. Germline mutations were identified in 38 patients (61%). Nineteen of the detected mutations were presumed to be novel, thus emphasizing the heterogeneity of the mutational spectrum in Korean FAP patients. In the initial 48 patients, SSCP analysis was followed by PTT for those patients for whom no detectable mutations were found by SSCP. Using this combined approach, we identified germline APC gene mutations in 29 of the 48 FAP patients (60%), including 6 patients in whom SSCP analysis failed to distinguish the mutant allele. In the 14 later patients, we identified truncating mutations in 9 patients (64%) using PTT only. Our results confirm that the mutation detection rate with PTT was superior to that with SSCP, and suggest that PTT would be a more practical screening method to detect germline mutations of the APC gene in FAP patients. 10083733 26 29 APC Modifier D011125 10083733 45 75 familial adenomatous polyposis Modifier D011125 10083733 186 216 familial adenomatous polyposis SpecificDisease D011125 10083733 218 221 FAP SpecificDisease D011125 10083733 254 280 adenomatous polyposis coli Modifier D011125 10083733 282 285 APC Modifier D011125 10083733 779 782 FAP Modifier D011125 10083733 980 983 APC Modifier D011125 10083733 1015 1018 FAP Modifier D011125 10083733 1410 1413 APC Modifier D011125 10083733 1422 1425 FAP Modifier D011125 10842298|t|Homozygosity mapping in a family with microcephaly, mental retardation, and short stature to a Cohen syndrome region on 8q21.3-8q22.1: redefining a clinical entity. 10842298|a|A syndrome of microcephaly, progressive postnatal growth deficiency, and mental retardation was observed in two brothers and their cousin from a multiply consanguineous kindred of Lebanese descent. Hypotonia, chorioretinal dystrophy, and myopia were also identified. The severity of the condition varied among the closely related patients. Because of absence of a distinctive facial appearance, the degree of mental retardation, and short stature, the initially considered clinical diagnosis of Cohen syndrome was withdrawn and a novel genetic entity was assumed. Homozygosity mapping in this family assigned the gene to a 26. 8-cM region on the chromosome band 8q21. 3 -22. 1, between the microsatellites at D8S270 and D8S514. The maximum two-point LOD score was found for marker at D8S267 (Zmax = 3.. 237 at Omax = 0. 00). Intriguingly enough, the identified gene region overlaps the refined gene region for Cohen syndrome (COH1) [Kolehmainen et al., 1997 Euro J Hum Genet 5 206-213]. This fact encourages the hypothesis that the described kindred segregates for a variant of Cohen syndrome and suggests a redefinition of its phenotype 10842298 38 50 microcephaly DiseaseClass D008831 10842298 52 70 mental retardation DiseaseClass D008607 10842298 76 89 short stature DiseaseClass D006130 10842298 95 109 Cohen syndrome Modifier C536438 10842298 179 191 microcephaly DiseaseClass D008831 10842298 205 232 postnatal growth deficiency DiseaseClass D006130 10842298 238 256 mental retardation DiseaseClass D008607 10842298 363 372 Hypotonia DiseaseClass D009123 10842298 374 397 chorioretinal dystrophy DiseaseClass D015862+D058499 10842298 403 409 myopia DiseaseClass D009216 10842298 574 592 mental retardation DiseaseClass D008607 10842298 598 611 short stature DiseaseClass D006130 10842298 660 674 Cohen syndrome SpecificDisease C536438 10842298 1075 1089 Cohen syndrome SpecificDisease C536438 10842298 1245 1259 Cohen syndrome SpecificDisease C536438 2835825|t|Spontaneous reversion of novel Lesch-Nyhan mutation by HPRT gene rearrangement. 2835825|a|Molecular analysis of an unusual patient with the Lesch-Nyhan syndrome has suggested that the mutation is due to a partial HPRT gene duplication. We now report the cloning and sequencing of the mutant HPRT cDNA which shows the precise duplication of exons 2 and 3. This mutation is the result of an internal duplication of 16-20 kilobases of the gene. The structure of the mutant gene suggests that the duplication was not generated by a single unequal crossing-over event between two normal HPRT alleles. Growth of Epstein-Barr virus-transformed lymphoblasts from this patient in selective medium has permitted isolation of spontaneous HPRT + revertants of this mutation. The reversion event involves a second major HPRT gene rearrangement where most or all of the duplicated portion of the mutant gene is deleted. The original mutation therefore has the potential for spontaneous somatic reversion. This may explain the relatively mild symptoms of the Lesch-Nyhan syndrome exhibited by this patient.. 2835825 31 42 Lesch-Nyhan Modifier D007926 2835825 130 150 Lesch-Nyhan syndrome SpecificDisease D007926 2835825 596 608 Epstein-Barr Modifier D020031 2835825 1034 1054 Lesch-Nyhan syndrome SpecificDisease D007926 7422429|t|Nephropathy in the Wiskott-Aldrich syndrome. 7422429|a|Nephropathy was detected in five of 32 patients with the Wiskott-Aldrich syndrome who were participating in a study of transfer factor (TF) therapy. In two patients, nephropathy was present before TF and did not appear changed by TF therapy. One of these patients subsequently developed progressive renal failure requiring dialysis beginning 5 1/2 years after TF therapy. In two patients, decreased renal function appeared very soon after the administration of TF. One patient showed gradually decreasing renal function beginning after two years of TF therapy. An additional patient was identified who died with renal failure without having received TF. The results suggest that renal failure occurs in the Wiskott-Aldrich syndrome more frequently than generally recognized and that administration of TF may precipitate or accelerate the renal disease in patients with this syndrome.. 7422429 0 11 Nephropathy SpecificDisease D007674 7422429 19 43 Wiskott-Aldrich syndrome SpecificDisease D014923 7422429 45 56 Nephropathy SpecificDisease D007674 7422429 102 126 Wiskott-Aldrich syndrome SpecificDisease D014923 7422429 211 222 nephropathy SpecificDisease D007674 7422429 344 357 renal failure SpecificDisease D051437 7422429 657 670 renal failure SpecificDisease D051437 7422429 724 737 renal failure SpecificDisease D051437 7422429 752 776 Wiskott-Aldrich syndrome SpecificDisease D014923 7422429 883 896 renal disease SpecificDisease D007674 7795596|t|Aniridia-associated cytogenetic rearrangements suggest that a position effect may cause the mutant phenotype. 7795596|a|Current evidence suggests that aniridia (absence of iris) is caused by loss of function of one copy of the PAX6 gene, which maps to 11p13. We present the further characterisation of two aniridia pedigrees in which the disease segregates with chromosomal rearrangements which involve 11p13 but do not disrupt the PAX6 gene. We have isolated three human YAC clones which encompass the PAX6 locus and we have used these to show that in both cases the chromosomal breakpoint is at least 85 kb distal of the 3 end of PAX6. In addition, the open reading frame of PAX6 is apparently free of mutations. We propose that the PAX6 gene on the rearranged chromosome 11 is in an inappropriate chromatin environment for normal expression and therefore that a position effect is the underlying mechanism of disease in these families.. 7795596 0 8 Aniridia Modifier D015783 7795596 141 149 aniridia SpecificDisease D015783 7795596 151 166 absence of iris SpecificDisease D015783 7795596 296 304 aniridia Modifier D015783 7962532|t|Genetic cholesteryl ester transfer protein deficiency caused by two prevalent mutations as a major determinant of increased levels of high density lipoprotein cholesterol. 7962532|a|Genetic determinants of HDL cholesterol (HDL-C) levels in the general population are poorly understood. We previously described plasma cholesteryl ester transfer protein (CETP) deficiency due to an intron 14 G (+ 1) -to-A mutation (Int14 A) in several families with very high HDL-C levels in Japan. Subjects with HDL-C > or = 100 mg/dl (n = 130) were screened by PCR single strand conformational polymorphism analysis of the CETP gene. Two other mutations were identified by DNA sequencing or primer-mediated restriction map modification of PCR products a novel intron 14 splice donor site mutation caused by a T insertion at position + 3 from the exon14/intron14 boundary (Int14 T) and a missense mutation (Asp442 to Gly) within exon 15 (D442G). The Int14 T mutation was only found in one family. However, the D442G and Int14 A mutations were highly prevalent in subjects with HDL-C > or = 60 mg/dl, with combined allele frequencies of 9%, 12%, 21% and 43% for HDL-C 60-79, 80-99, 100-119, and > or = 120 mg/dl, respectively. Furthermore, prevalences of the D442G and Int14 A mutations were extremely high in a general sample of Japanese men (n = 236), with heterozygote frequencies of 7% and 2%, respectively. These two mutations accounted for about 10% of the total variance of HDL-C in this population. The phenotype in a genetic compound heterozygote (Int14 T and Int14 A) was similar to that of Int14 A homozygotes (no detectable CETP and markedly increased HDL-C), indicating that the Int14 T produces a null allele. In four D442G homozygotes, mean HDL-C levels (86 +/- 26 mg/dl) were lower than in Int14 A homozygotes (158 +/- 35 mg/dl), reflecting residual CETP activity in plasma. In 47 D442G heterozygotes, mean HDL-C levels were 91 +/- 23 mg/dl, similar to the level in D442G homozygotes, and significantly greater than mean HDL-C levels in Int14 A heterozygotes (69 +/- 15 mg/dl). Thus, the D442G mutation acts differently to the null mutations with weaker effects on HDL in the homozygous state and stronger effects in the heterozygotes, suggesting dominant expression of a partially defective allele. CETP deficiency, reflecting two prevalent mutations (D442G and Int14 A), is the first example of a genetic deficiency state which is sufficiently common to explain a significant fraction of the variation in HDL-C in the general population.. 7962532 8 53 cholesteryl ester transfer protein deficiency SpecificDisease OMIM:143470 7962532 307 359 cholesteryl ester transfer protein (CETP) deficiency SpecificDisease OMIM:143470 7962532 2289 2304 CETP deficiency SpecificDisease OMIM:143470 7962532 2388 2406 genetic deficiency Modifier D030342 3377761|t|X-linked glucose-6-phosphate dehydrogenase deficiency in Mus musculus. 3377761|a|A mouse with X-linked glucose-6-phosphate dehydrogenase (G6PD) deficiency has been recovered in offspring of 1-ethyl-1-nitrosourea-treated male mice. The activity alteration was detected in blood but can also be observed in other tissue extracts. Hemizygous, heterozygous, and homozygous mutants have, respectively, about 15, 60, and 15% G6PD remaining activity in the blood as compared to the wild type. Erythrocyte indices did not show differences between mutants and wild types. The mutation does not affect the electrophoretic migration, the isoelectric point, or the thermal stability. Kinetic properties, such as the Km for glucose-6-phosphate or for NADP and the relative utilization of substrate analogues, showed no differences between wild types and mutants with the exception of the relative utilization of deamino-NADP which was significantly lower in mutants. This is presently the only animal model for X-linked G6PD deficiency in humans.. 3377761 0 53 X-linked glucose-6-phosphate dehydrogenase deficiency SpecificDisease D005955 3377761 84 144 X-linked glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 3377761 988 1012 X-linked G6PD deficiency SpecificDisease D005955 10798358|t|Human glycine decarboxylase gene (GLDC) and its highly conserved processed pseudogene (psiGLDC): their structure and expression, and the identification of a large deletion in a family with nonketotic hyperglycinemia. 10798358|a|Mutations in the glycine decarboxylase gene (GLDC) cause nonketotic hyperglycinemia (NKH), an in-born error of metabolism characterized by severe neurological disturbance. We have determined the structure of GLDC and of its pseudogene (psiGLDC) and studied their expression for a molecular analysis of NKH. The GLDC gene spans at least 135 kb and consists of 25 exons. All donor and acceptor sites adhere to the canonical GT-AG rule, except for the donor site of intron 21, where a variant form GC is used instead of GT. The transcription initiation site has been assigned to a residue 163 bp upstream from the translation initiation triplet by primer extension analysis. The psiGLDC gene has no intron and shares 97. 5% homology with the coding region of functional GLDC, suggesting that psiGLDC is a processed pseudogene that arose from the GLDC transcript about 4-8 million years ago. RNA blotting analysis has revealed that GLDC is expressed in human liver, kidney, brain, and placenta. We have also examined a patient with NKH with no detectable GLDC mRNA in his lymphoblasts. Exons 1-3 of the functional GLDC gene from this patient are not amplified by polymerase chain reaction (PCR), whereas those from control subjects are. These results suggest a large homozygous deletion (at least 30 kb) in the patient. Furthermore, we have devised a semi-quantitative PCR to estimate the number of GLDC alleles by using psiGLDC as an internal control and have confirmed the homozygosity and heterozygosity of the deletion in the patient and his parents, respectively. Structural information of GLDC and psiGLDC should facilitate the molecular analysis of NKH. 10798358 189 215 nonketotic hyperglycinemia SpecificDisease D020158 10798358 274 300 nonketotic hyperglycinemia SpecificDisease D020158 10798358 302 305 NKH SpecificDisease D020158 10798358 311 338 in-born error of metabolism DiseaseClass D008661 10798358 363 387 neurological disturbance DiseaseClass D009461 10798358 519 522 NKH SpecificDisease D020158 10798358 1245 1248 NKH SpecificDisease D020158 10798358 1869 1872 NKH SpecificDisease D020158 8589721|t|Growth retardation and tumour inhibition by BRCA1. 8589721|a|Inherited mutations in BRCA1 predispose to breast and ovarian cancer, but the role of BRCA1 in sporadic breast and ovarian cancer has previously been elusive. Here, we show that retroviral transfer of the wild-type BRCA1 gene inhibits growth in vitro of all breast and ovarian cancer cell lines tested, but not colon or lung cancer cells or fibroblasts. Mutant BRCA1 has no effect on growth of breast cancer cells; ovarian cancer cell growth is not affected by BRCA1 mutations in the 5 portion of the gene, but is inhibited by 3 BRCA1 mutations. Development of MCF-7 tumours in nude mice is inhibited when MCF-7 cells are transfected with wild-type, but not mutant, BRCA1. Most importantly, among mice with established MCF-7 tumours, peritoneal treatment with a retroviral vector expressing wild-type BRCA1 significantly inhibits tumour growth and increased survival.. 8589721 0 18 Growth retardation DiseaseClass D006130 8589721 23 29 tumour Modifier D009369 8589721 94 119 breast and ovarian cancer CompositeMention D061325 8589721 155 180 breast and ovarian cancer CompositeMention D001943|D010051 8589721 309 334 breast and ovarian cancer Modifier D001943|D010051 8589721 362 382 colon or lung cancer Modifier D003110|D008175 8589721 445 458 breast cancer Modifier D001943 8589721 466 480 ovarian cancer Modifier D010051 8589721 612 625 MCF-7 tumours DiseaseClass D009369 8589721 770 783 MCF-7 tumours DiseaseClass D009369 8589721 881 887 tumour Modifier D009369 126380|t|Striking prevalence of ankylosing spondylitis in "healthy" w27 positive males and females. 126380|a|Ankylosing spondylitis is diagnosed once or twice in each 1000 males and one tenth as frequently in females, but the true prevalence is unknown. Indentification of genetic marker, HL-A W27, for susceptible persons has provided a tool facilitating epidemiologic studies and allowing identification of " control " populations without the marker. Evaluation by postal questionnaires, and pelvic radiography of 78 HL-A 27W-positive blood donors selected from a group of apparently healthy subjects revealed 14 who satisfied the criteria for definite ankylosing spondylitis. The prevalence was similar in both sexes. One hundred and twenty-six W27-negative controls matched for race, sex, and age failed to yield a single case. For a person of either sex with HL-A W27, there appears to be about a 20 per cent chance that ankylosing spondylitis will develop, suggesting a prevalence of 10 to 15 per thousand. Hitherto accepted figures may underestimate the frequency by a factor of 10 to 20.. 126380 23 45 ankylosing spondylitis SpecificDisease D013167 126380 91 113 Ankylosing spondylitis SpecificDisease D013167 126380 637 659 ankylosing spondylitis SpecificDisease D013167 126380 908 930 ankylosing spondylitis SpecificDisease D013167 7663517|t|Rapid detection of BRCA1 mutations by the protein truncation test. 7663517|a|More than 75% of the reported mutations in the hereditary breast and ovarian cancer gene, BRCA1, result in truncated proteins. We have used the protein truncation test (PTT) to screen for mutations in exon 11, which encodes 61% of BRCA1. In 45 patients from breast and/or ovarian cancer families we found six novel mutations two single nucleotide insertions, three small deletions (1-5 bp) and a nonsense mutation identified two unrelated families. Furthermore, we were able to amplify the remaining coding region by RT-PCR using lymphocyte RNA. Combined with PTT, we detected aberrantly spliced products affecting exons 5 and 6 in one of two BRCA1-linked families examined. The protein truncation test promises to become a valuable technique in detecting BRCA1 mutations.. 7663517 114 150 hereditary breast and ovarian cancer Modifier D061325 7663517 325 353 breast and/or ovarian cancer Modifier D001943|D010051 1376553|t|New variant in exon 3 of the proteolipid protein (PLP) gene in a family with Pelizaeus-Merzbacher disease. 1376553|a|A C--greater than G transversion has been found in exon 3 of the PLP gene of affected males and their mother in a single sibship with Pelizaeus-merzbacher disease (PMD). The transversion should not result in an amino acid change in the protein but it does result in the loss of a HaeIII restriction endonuclease cleavage site. It is concordant with the disease in this family. One-hundred-ten unrelated X chromosomes are negative for this mutation. No other sequence defect was found in the PLP exons of the affected males. The cause of disease in this family remains unknown, but the association between this rare mutation and PMD is intriguing. The mutation can serve as a marker for following segregation of the PLP gene.. 1376553 77 105 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 1376553 241 269 Pelizaeus-merzbacher disease SpecificDisease OMIM:312080 1376553 271 274 PMD SpecificDisease OMIM:312080 1376553 735 738 PMD SpecificDisease OMIM:312080 7390473|t|Variants of erythrocyte glucose-6-phosphate dehydrogenase (G6PD) in Bulgarian populations. 7390473|a|Ten variants of erythrocyte glucose-6-phosphate dehydrogenase were identified in 22 patients with G6PD deficiency from three districts of Bulgaria. Corinth-like and Fayoum-like variants were the most frequent; Mediterranean, Ohut II, Kilgore, Boston, Poznan, and Panay variants and two new variants, Petrich and Gotze Delchev, were each found in one or two carriers. No correlation was revealed between clinical and biochemical polymorphism.. 7390473 189 204 G6PD deficiency SpecificDisease D005955 10381492|t|The C282Y mutation causing hereditary hemochromatosis does not produce a null allele. 10381492|a|Targeted mutagenesis was used to produce two mutations in the murine hemochromatosis gene (Hfe) locus. The first mutation deletes a large portion of the coding sequence, generating a null allele. The second mutation introduces a missense mutation (C282Y) into the Hfe locus, but otherwise leaves the gene intact. This mutation is identical to the disease-causing mutation in patients with hereditary hemochromatosis. Mice carrying each of the two mutations were bred and analyzed. Homozygosity for either mutation results in postnatal iron loading. The effects of the null mutation are more severe than the effects of the C282Y mutation. Mice heterozygous for either mutation accumulate more iron than normal controls. Interestingly, although liver iron stores are greatly increased, splenic iron is decreased. We conclude that the C282Y mutation does not result in a null allele.. 10381492 27 53 hereditary hemochromatosis SpecificDisease D006432 10381492 155 170 hemochromatosis Modifier D006432 10381492 475 501 hereditary hemochromatosis SpecificDisease D006432 2209091|t|Regional localisation of the Friedreich ataxia locus to human chromosome 9q13----q21.1. 2209091|a|We have previously assigned the Friedreich ataxia locus (FRDA) to chromosome 9; the current maximal lod score between FRDA and MCT112 (D9S15) is greater than 50 at a recombination fraction of theta = 0. The physical assignment of the locus defined by MCT112, and hence FRDA, has not been determined, although linkage analysis of MCT112 with other chromosome 9 markers inferred a location close to the centromere. We have used in situ hybridisation with MCT112, a corresponding cosmid MJ1, and DR47 (D9S5), coupled with mapping studies on hybrid cell panels, to define more precisely the location of the disease locus. The in situ location of all three probes is 9q13----q21. 1, distal to the variable heterochromatin region. Physical assignment of FRDA will allow us to identify hybrid cell lines containing the mutated gene. 2209091 29 46 Friedreich ataxia Modifier D005621 2209091 120 137 Friedreich ataxia Modifier D005621 10470286|t|Mxi1 mutations in human neurofibrosarcomas. 10470286|a|Mxi1 is thought to negatively regulate Myc function and may therefore be a potential tumor suppressor gene. Little effort has yet been made to find alterations involving this gene in human solid tumors. We screened 31 human gastric cancers, 7 esophageal cancers, 85 bone and soft tissue tumors of various types, including 4 neurofibrosarcomas. We also examined 29 human tumor cell lines consisting of 12 esophageal cancers, 7 glioma/glioblastomas and 10 others for Mxi1 mutations in exons 1, 2, 4 (HLH domain), 5 and 6. Polymerase chain reaction-single-strand conformation polymorphism (PCR-SSCP) and subsequent sequencing revealed three distinct polymorphisms in the intron-exon boundary upstream from exon 6. We discovered a missense mutation, GCA to GTA (Ala 54 Val), in exon 2 in a neurofibrosarcoma patient (case 1), two missense mutations, AAA to CAA (Lys 118 Gln) and GAA to GGA (Glu 154 Gly) in exon 5 of another neurofibrosarcoma patient (case 2), and 3 amino acid substitutions, GTG to GCG (Val 179 Ala), GTT to GCT (Val 181 Ala) and TTC to CTC (Phe 186 Leu), in a third neurofibrosarcoma patient (case 3). In case 3, loss of heterozygosity was also demonstrated by informative (TTC) 3/(TTC) 2 polymorphism. Our data demonstrate that mutations occur in the Mxi1 gene in neurofibrosarcoma. Missense mutations in the functional domain of Mxi1 in these cases may be involved in the pathogenesis of neurofibrosarcoma.. 10470286 24 42 neurofibrosarcomas SpecificDisease D018319 10470286 129 134 tumor Modifier D009369 10470286 233 245 solid tumors DiseaseClass D009369 10470286 268 283 gastric cancers SpecificDisease D013274 10470286 287 305 esophageal cancers SpecificDisease D004938 10470286 310 337 bone and soft tissue tumors CompositeMention D001859|D012983 10470286 368 386 neurofibrosarcomas SpecificDisease D018319 10470286 414 419 tumor Modifier D009369 10470286 448 466 esophageal cancers SpecificDisease D004938 10470286 470 476 glioma SpecificDisease D005910 10470286 477 490 glioblastomas SpecificDisease D005909 10470286 830 847 neurofibrosarcoma Modifier D018319 10470286 965 982 neurofibrosarcoma Modifier D018319 10470286 1125 1142 neurofibrosarcoma Modifier D018319 10470286 1324 1341 neurofibrosarcoma SpecificDisease D018319 10470286 1449 1466 neurofibrosarcoma SpecificDisease D018319 7833921|t|Molecular basis of essential fructosuria: molecular cloning and mutational analysis of human ketohexokinase (fructokinase). 7833921|a|Essential fructosuria is one of the oldest known inborn errors of metabolism. It is a benign condition which is believed to result from deficiency of hepatic fructokinase (ketohexokinase, KHK, E. C. 2. 7. 1. 3). This enzyme catalyses the first step of metabolism of dietary fructose, conversion of fructose to fructose-1-phosphate. Despite the early recognition of this disorder, the primary structure of human KHK and the molecular basis of essential fructosuria have not been previously defined. In this report, the isolation and sequencing of full-length cDNA clones encoding human ketohexokinase are described. Alternative mRNA species and alternative KHK isozymes are produced by alternative polyadenylation and splicing of the KHK gene. The KHK proteins show a high level of sequence conservation relative to rat KHK. Direct evidence that mutation of the KHK structural gene is the cause of essential fructosuria was also obtained. In a well-characterized family, in which three of eight siblings have fructosuria, all affected individuals are compound heterozygotes for two mutations Gly40Arg and Ala43Thr. Both mutations result from G-- > A transitions, and each alters the same conserved region of the KHK protein. Neither mutation was seen in a sample of 52 unrelated control individuals. An additional conservative amino acid change (Val49IIe) was present on the KHK allele bearing Ala43Thr 7833921 19 40 essential fructosuria SpecificDisease C538068 7833921 124 145 Essential fructosuria SpecificDisease C538068 7833921 173 200 inborn errors of metabolism DiseaseClass D008661 7833921 260 294 deficiency of hepatic fructokinase SpecificDisease C538068 7833921 566 587 essential fructosuria SpecificDisease C538068 7833921 1021 1042 essential fructosuria SpecificDisease C538068 7833921 1132 1143 fructosuria DiseaseClass C538068 10449794|t|Loss of the ataxia-telangiectasia gene product causes oxidative damage in target organs. 10449794|a|Ataxia-telangiectasia (A-T) is characterized by a markedly increased sensitivity to ionizing radiation, increased incidence of cancer, and neurodegeneration, especially of the cerebellar Purkinje cells. Ionizing radiation oxidizes macromolecules and causes tissue damage through the generation of reactive oxygen species (ROS). We therefore hypothesized that A-T is due to oxidative damage resulting from loss of function of the A-T gene product. To assess this hypothesis, we employed an animal model of A-T, the mouse with a disrupted Atm gene. We show that organs which develop pathologic changes in the Atm-deficient mice are targets of oxidative damage, and that cerebellar Purkinje cells are particularly affected. These observations provide a mechanistic basis for the A-T phenotype and lay a rational foundation for therapeutic intervention.. 10449794 12 33 ataxia-telangiectasia Modifier D001260 10449794 89 110 Ataxia-telangiectasia SpecificDisease D001260 10449794 112 115 A-T SpecificDisease D001260 10449794 216 222 cancer DiseaseClass D009369 10449794 228 245 neurodegeneration DiseaseClass D019636 10449794 448 451 A-T SpecificDisease D001260 10449794 518 521 A-T Modifier D001260 10449794 594 597 A-T SpecificDisease D001260 10449794 865 868 A-T Modifier D001260 6604602|t|Family studies in Bechterew's syndrome (ankylosing spondylitis) III. Genetics. 6604602|a|The results of segregation analyses in 75 families where the proband had ankylosing spondylitis, are presented. Of the 278 adult, living first degree relatives, approximately 85% cooperated in the study. Clinical and radiographical examinations were performed and HLA typing was conducted. The results were in agreement with our hypothesis that ankylosing spondylitis is part of a syndrome where different genetic factors interact. Such known factors are HLA B27 associated disease susceptibility, susceptibility to psoriatic arthropathy and susceptibility to entero-arthropathy. Radiographical sacro-iliitis was restricted to HLA B27 positive relatives, and was more frequently found in relatives to probands with psoriasis than in relatives to probands without psoriasis. Environmental factors (intestinal bacteria) are known to trigger the disease at least in some persons, and we have postulated that all or most of them have the predisposition to develop disease. Thus, the syndrome has a multifactorial etiology. The phenotypic expressions of the different genetic predispositions involved, include sacro-iliitis, psoriasis, acute anterior uveitis, peripheral arthropathy and inflammatory bowel disease. We suggest the descriptive name HEREDITARY MULTIFOCAL RELAPSING INFLAMMATION (HEMRI) for this syndrome. Ankylosing spondylitis, psoriatic arthropathy and entero-arthropathy may be regarded as clinical sub-types of the syndrome.. 6604602 18 38 Bechterew's syndrome SpecificDisease D013167 6604602 40 62 ankylosing spondylitis SpecificDisease D013167 6604602 152 174 ankylosing spondylitis SpecificDisease D013167 6604602 424 446 ankylosing spondylitis SpecificDisease D013167 6604602 595 616 psoriatic arthropathy SpecificDisease D015535 6604602 639 657 entero-arthropathy SpecificDisease C538273+D001168 6604602 674 687 sacro-iliitis SpecificDisease D058566 6604602 794 803 psoriasis SpecificDisease D011565 6604602 842 851 psoriasis SpecificDisease D011565 6604602 1184 1197 sacro-iliitis SpecificDisease D058566 6604602 1199 1208 psoriasis SpecificDisease D011565 6604602 1210 1232 acute anterior uveitis SpecificDisease D014606 6604602 1245 1256 arthropathy SpecificDisease D007592 6604602 1261 1287 inflammatory bowel disease SpecificDisease D015212 6604602 1321 1365 HEREDITARY MULTIFOCAL RELAPSING INFLAMMATION SpecificDisease D007249+D030342 6604602 1367 1372 HEMRI SpecificDisease D007249+D030342 6604602 1393 1415 Ankylosing spondylitis SpecificDisease D013167 6604602 1417 1438 psoriatic arthropathy SpecificDisease D015535 6604602 1443 1461 entero-arthropathy SpecificDisease C538273+D007592 1315306|t|Chromosome mapping of the rod photoreceptor cGMP phosphodiesterase beta-subunit gene in mouse and human: tight linkage to the Huntington disease region (4p16.3). 1315306|a|The retinal degeneration mouse (gene symbol, rd) is an animal model for certain forms of human hereditary retinopathies. Recent findings of a nonsense mutation in the rd mouse PDE beta-subunit gene (Pdeb) prompted us to investigate the chromosome locations of the mouse and human genes. We have utilized backcross analysis in mice to verify and define more precisely the location of the Pdeb locus 6. 1 +/- 2. 3 cM distal of Mgsa on mouse chromosome 5. We have determined that the human gene (PDEB) maps to 4p16. 3, very close to the Huntington disease (HD) region. Analysis of the comparative map for mice and humans shows that the mouse homologue of the HD gene will reside on chromosome 5. Linkage of the mouse Pdeb locus with other homologues in the human 4p16. 3 region is maintained but gene order is not, suggesting at least three possible sites for the corresponding mouse HD gene. 1315306 126 144 Huntington disease Modifier D006816 1315306 166 186 retinal degeneration Modifier D012162 1315306 257 281 hereditary retinopathies DiseaseClass D015785 1315306 696 714 Huntington disease Modifier D006816 1315306 716 718 HD Modifier D006816 1315306 818 820 HD Modifier D006816 1315306 1043 1045 HD Modifier D006816 777027|t|Hereditary deficiency of the fifth component of complement in man. II. Biological properties of C5-deficient human serum. 777027|a|The first known human kindred with hereditary deficiency of the fifth component of complement (C5) was documented in the accompanying report. This study examines several biological properties of C5-deficient (C5D) human serum, particularly sera obtained from two C5D homozygotes. The proband, who has inactive systemic lupus erythematosus is completely lacking C5, while her healthy half-sister has 1-2% of normal levels. Both sera were severely impaired in their ability to generate chemotactic activity for normal human neutrophils upon incubation with aggregated human gamma-globulin or Escherichia coli endotoxin. This function was fully restored in the siblings serum, and substantially improved in the probands serum, by addition of highly purified human C5 to normal serum concentrations. Sera from eight family members who were apparently heterozygous for C5 deficiency gave normal chemotactic scores. The ability of C5D serum to opsonize Saccharomyces cerevisiae (bakers yeast) or Candida albicans for ingestion by normal neutrophils was completely normal. In addition, C5D serum was capable of promoting normal phagocytosis and intracellular killing of Staphylococcus aureus. The probands serum was incapable of mediating lysis of erythrocytes from a patient with paroxysmal nocturnal hemoglobinuria in both the sucrose hemolysia and acid hemolysis tests, and also lacked bactericidal activity against sensitized or unsensitized Salmonella typhi. The siblings serum, containing only 1-2% of normal C5, effectively lysed S. typhi, but only at eightfold lower serum dilutions as compared to normals. These findings underscore the critical role of C5 in the generation of chemotactic activity and in cytolytic reactions, as opposed to a nonobligatory or minimal role in opsonization, at least for the organisms under study.. 777027 0 58 Hereditary deficiency of the fifth component of complement SpecificDisease OMIM:609536 777027 96 108 C5-deficient Modifier OMIM:609536 777027 157 215 hereditary deficiency of the fifth component of complement SpecificDisease OMIM:609536 777027 317 329 C5-deficient Modifier OMIM:609536 777027 331 334 C5D Modifier OMIM:609536 777027 385 388 C5D Modifier OMIM:609536 777027 432 460 systemic lupus erythematosus SpecificDisease D008180 777027 986 999 C5 deficiency SpecificDisease OMIM:609536 777027 1047 1050 C5D Modifier OMIM:609536 777027 1201 1204 C5D Modifier OMIM:609536 777027 1396 1431 paroxysmal nocturnal hemoglobinuria SpecificDisease D006457 10807793|t|The gene for familial Mediterranean fever, MEFV, is expressed in early leukocyte development and is regulated in response to inflammatory mediators. 10807793|a|Familial Mediterranean fever (FMF) is a recessive disorder characterized by episodes of fever and neutrophil-mediated serosal inflammation. We recently identified the gene causing FMF, designated MEFV, and found it to be expressed in mature neutrophils, suggesting that it functions as an inflammatory regulator. To facilitate our understanding of the normal function of MEFV, we extended our previous studies. MEFV messenger RNA was detected by reverse transcriptase-polymerase chain reaction in bone marrow leukocytes, with differential expression observed among cells by in situ hybridization. CD34 hematopoietic stem-cell cultures induced toward the granulocytic lineage expressed MEFV at the myelocyte stage, concurrently with lineage commitment. The prepromyelocytic cell line HL60 expressed MEFV only at granulocytic and monocytic differentiation. MEFV was also expressed in the monocytic cell lines U937 and THP-1. Among peripheral blood leukocytes, MEFV expression was detected in neutrophils, eosinophils, and to varying degrees, monocytes. Consistent with the tissue specificity of expression, complete sequencing and analysis of upstream regulatory regions of MEFV revealed homology to myeloid-specific promoters and to more broadly expressed inflammatory promoter elements. In vitro stimulation of monocytes with the proinflammatory agents interferon (IFN) gamma, tumor necrosis factor, and lipopolysaccharide induced MEFV expression, whereas the antiinflammatory cytokines interleukin (IL) 4, IL-10, and transforming growth factor beta inhibited such expression. Induction by IFN-gamma occurred rapidly and was resistant to cycloheximide. IFN-alpha also induced MEFV expression. In granulocytes, MEFV was up-regulated by IFN-gamma and the combination of IFN-alpha and colchicine. These results refine understanding of MEFV by placing the gene in the myelomonocytic-specific proinflammatory pathway and identifying it as an IFN-gamma immediate early gene.. 10807793 13 41 familial Mediterranean fever SpecificDisease D010505 10807793 149 177 Familial Mediterranean fever SpecificDisease D010505 10807793 179 182 FMF SpecificDisease D010505 10807793 189 207 recessive disorder DiseaseClass D030342 10807793 237 242 fever DiseaseClass D005334 10807793 247 287 neutrophil-mediated serosal inflammation SpecificDisease D007249 10807793 329 332 FMF SpecificDisease D010505 10807793 1526 1531 tumor Modifier D009369 10556283|t|Spectrum of hSNF5/INI1 somatic mutations in human cancer and genotype-phenotype correlations. 10556283|a|The hSNF5/INI1 gene which encodes a member of the SWI/SNF chromatin ATP-dependent remodeling complex, is a new tumor suppressor gene localized on chromosome 22q11. 2 and recently shown to be mutated in malignant rhabdoid tumors. We have searched for hSNF5/INI1 mutations in 229 tumors of various origins using a screening method based on denaturing high-performance liquid chromatography. A total of 31 homozygous deletions and 36 point alterations were identified. Point mutations were scattered along the coding sequence and included 15 nonsense, 15 frameshift, three splice site, two missense and one editing mutations. Mutations were retrieved in most rhabdoid tumors, whatever their sites of occurrence, indicating the common pathogenetic origin of these tumors. Recurrent hSNF5/INI1 alterations were also observed in choroid plexus carcinomas and in a subset of central primitive neuroectodermal tumors (cPNETs) and medulloblastomas. In contrast, hSNF5/INI1 point mutations were not detected in breast cancers, Wilms tumors, gliomas, ependymomas, sarcomas and other tumor types, even though most analyzed cases harbored loss of heterozygosity at 22q11. 2 loci. These results suggest that rhabdoid tumors, choroid plexus carcinomas and a subset of medulloblastomas and cPNETs share common pathways of oncogenesis related to hSNF5/INI1 alteration and that hSNF5/INI1 mutations define a genetically homogeneous family of highly aggressive cancers mainly occurring in young children and frequently, but not always, exhibiting a rhabdoid phenotype 10556283 50 56 cancer DiseaseClass D009369 10556283 205 210 tumor Modifier D009369 10556283 296 321 malignant rhabdoid tumors DiseaseClass D018335 10556283 372 378 tumors DiseaseClass D009369 10556283 750 765 rhabdoid tumors DiseaseClass D018335 10556283 854 860 tumors DiseaseClass D009369 10556283 917 942 choroid plexus carcinomas SpecificDisease D020288 10556283 980 1002 neuroectodermal tumors DiseaseClass D017599 10556283 1016 1032 medulloblastomas SpecificDisease D008527 10556283 1095 1109 breast cancers SpecificDisease D001943 10556283 1111 1123 Wilms tumors SpecificDisease D009396 10556283 1125 1132 gliomas SpecificDisease D005910 10556283 1134 1145 ependymomas SpecificDisease D004806 10556283 1147 1155 sarcomas DiseaseClass D012509 10556283 1166 1171 tumor Modifier D009369 10556283 1288 1303 rhabdoid tumors DiseaseClass D018335 10556283 1305 1330 choroid plexus carcinomas SpecificDisease D020288 10556283 1347 1363 medulloblastomas SpecificDisease D008527 10556283 1536 1543 cancers DiseaseClass D009369 10556283 1624 1632 rhabdoid Modifier D018335 624546|t|Heterogeneity of glucose-6-phosphate dehydrogenase deficiency in Algeria. Study in Northern Algeria with description of five new variants. 624546|a|Glucose-6-phosphate dehydrogenase (G6PD) deficiency was found in 3. 2% of the male population living in the urban area of Algiers. The deficient subjects originated from multiple geographic regions of Northern Algeria, with prevalence of individuals of Berber-Kabyle origin. Red blood cell G6PD was partially purified and characterized in deficient males from 17 families, and six different variants were found. Among them, only one, the Gd (-) Kabyle variant, had been previously described. It was detected in nine families. The other five variants were new Gd (-) Laghouat (four cases), Gd (-) Blida (one case), Gd (-) Thenia (one case), Gd (-) Titteri (one case), and Gd (-) Alger (two brothers). Strikingly, the common Mediterranean variant was not found. G6PD deficiency is heterogeneous in northern Algeria where autochtonous variants seem to prevail. The Kabyle variant may be common in this country. 624546 17 61 glucose-6-phosphate dehydrogenase deficiency SpecificDisease D005955 624546 139 190 Glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 624546 900 915 G6PD deficiency SpecificDisease D005955 1302032|t|Fragile X syndrome without CCG amplification has an FMR1 deletion. 1302032|a|We describe a patient with typical clinical features of the fragile X syndrome, but without cytogenetic expression of the fragile X or an amplified CCG trinucleotide repeat fragment. The patient has a previously uncharacterized submicroscopic deletion encompassing the CCG repeat, the entire FMR1 gene and about 2. 5 megabases of flanking sequences. This finding confirms that the fragile X phenotype can exist, without amplification of the CCG repeat or cytogenetic expression of the fragile X, and that fragile X syndrome is a genetically homogeneous disorder involving FMR1. We also found random X-inactivation in the mother of the patient who was shown to be a carrier of this deletion. 1302032 0 18 Fragile X syndrome SpecificDisease D005600 1302032 127 145 fragile X syndrome SpecificDisease D005600 1302032 189 198 fragile X SpecificDisease D005600 1302032 448 457 fragile X Modifier D005600 1302032 552 561 fragile X SpecificDisease D005600 1302032 572 590 fragile X syndrome SpecificDisease D005600 10589394|t|Confirmation of linkage of Van der Woude syndrome to chromosome 1q32: evidence of association with STR alleles suggests possible unique origin of the disease mutation. 10589394|a|Van der Woude syndrome (VWS) is an autosomal dominant craniofacial disorder with high penetrance and variable expression. Its clinical features are variably expressed, but include cleft lip and/or cleft palate, lip pits and hypodontia. All VWS families studied to date map the disease gene to a < 2 cM region of chromosome 1q32, with no evidence of locus heterogeneity. The aim of this study is to refine the localization of the VWS gene and to further assess possible heterogeneity. We analyzed four multiplex VWS families. All available members were clinically assessed and genotyped for 19 short tandem repeat markers on chromosome 1 in the VWS candidate gene region. We performed two-point and multipoint limit of detection (LOD) score analyses using a high penetrance autosomal dominant model. All families showed positive LOD scores without any recombination in the candidate region. The largest two-point LOD score was 5. 87 87. Our assay method for short tandem repeat (STR) markers provided highly accurate size estimation of marker allele fragment sizes, and therefore enabled us to determine the specific alleles segregating with the VWS gene in each of our four families. We observed a striking pattern of STR allele sharing at several closely linked loci among our four Caucasian VWS families recruited at three different locations in the US. These results suggest the possibility of a unique origin for a mutation responsible for many or most cases of VWS. 10589394 27 49 Van der Woude syndrome SpecificDisease C536528 10589394 168 190 Van der Woude syndrome SpecificDisease C536528 10589394 192 195 VWS SpecificDisease C536528 10589394 203 243 autosomal dominant craniofacial disorder DiseaseClass D019465 10589394 348 357 cleft lip SpecificDisease D002971 10589394 365 377 cleft palate SpecificDisease D002972 10589394 379 387 lip pits SpecificDisease C536528 10589394 392 402 hypodontia SpecificDisease D000848 10589394 408 411 VWS Modifier C536528 10589394 597 600 VWS Modifier C536528 10589394 679 682 VWS Modifier C536528 10589394 812 815 VWS Modifier C536528 10589394 1313 1316 VWS Modifier C536528 10589394 1461 1464 VWS Modifier C536528 10589394 1634 1637 VWS SpecificDisease C536528 7550349|t|The carrier frequency of the BRCA1 185delAG mutation is approximately 1 percent in Ashkenazi Jewish individuals. 7550349|a|Since BRCA1, the first major gene responsible for inherited breast cancer, was cloned, more than 50 unique mutations have been detected in the germline of individuals with breast and ovarian cancer. In high-risk pedigrees, female carriers of BRCA1 mutations have an 80-90% lifetime risk of breast cancer, and a 40-50% risk of ovarian cancer. However, the mutation stats of individuals unselected for breast or ovarian cancer has not been determined, and it is not known whether mutations in such individuals confer the same risk of cancer as in individuals from the high-risk families studied so far. Following the finding of a 185delAG frameshift mutation in several Ashkenazi Jewish breast/ovarian families, we have determined the frequency of this mutation in 858 Ashkenazim seeking genetic testing for conditions unrelated to cancer, and in 815 reference individuals not selected for ethnic origin. We observed the 185delAG mutation in 0. 9% of Ashkenazim (95% confidence limit, 0. 4-1. 8%) and in none of the reference samples. Our results suggest that one in a hundred women of Ashkenazi descent may be at especially high risk of developing breast and/or ovarian cancer. 7550349 163 186 inherited breast cancer SpecificDisease D061325 7550349 285 310 breast and ovarian cancer CompositeMention D061325 7550349 403 416 breast cancer SpecificDisease D001943 7550349 439 453 ovarian cancer SpecificDisease D010051 7550349 513 537 breast or ovarian cancer CompositeMention D010051|D001943 7550349 645 651 cancer DiseaseClass D009369 7550349 943 949 cancer DiseaseClass D009369 7550349 1260 1288 breast and/or ovarian cancer CompositeMention D010051|D001943 8460149|t|Targeted modification of the apolipoprotein B gene results in hypobetalipoproteinemia and developmental abnormalities in mice. 8460149|a|Familial hypobetalipoproteinemia is an autosomal codominant disorder resulting in a dramatic reduction in plasma concentrations of apolipoprotein (apo) B, cholesterol, and beta-migrating lipoproteins. A benefit of hypobetalipoproteinemia is that mildly affected individuals may be protected from coronary vascular disease. We have used gene targeting to generate mice with a modified Apob allele. Mice containing this allele display all of the hallmarks of human hypobetalipoproteinemia they produce a truncated apoB protein, apoB70, and have markedly decreased plasma concentrations of apoB, beta-lipoproteins, and total cholesterol. In addition, the mice manifest several characteristics that are occasionally observed in human hypobetalipoproteinemia, including reduced plasma triglyceride concentrations, fasting chylomicronemia, and reduced high density lipoprotein cholesterol. An unexpected finding is that the modified Apob allele is strongly associated with exencephalus and hydrocephalus. These mice should help increase our understanding of hypobetalipoproteinemia, atherogenesis, and the etiology of exencephalus and hydrocephalus.. 8460149 62 85 hypobetalipoproteinemia SpecificDisease D006995 8460149 90 117 developmental abnormalities DiseaseClass D006130 8460149 127 159 Familial hypobetalipoproteinemia SpecificDisease D052476 8460149 166 195 autosomal codominant disorder DiseaseClass D030342 8460149 341 364 hypobetalipoproteinemia SpecificDisease D006995 8460149 423 448 coronary vascular disease DiseaseClass D003323 8460149 590 613 hypobetalipoproteinemia SpecificDisease D006995 8460149 858 881 hypobetalipoproteinemia SpecificDisease D006995 8460149 945 960 chylomicronemia SpecificDisease OMIM:238600 8460149 1095 1107 exencephalus SpecificDisease D009436 8460149 1112 1125 hydrocephalus SpecificDisease D006849 8460149 1180 1203 hypobetalipoproteinemia SpecificDisease D006995 8460149 1205 1218 atherogenesis DiseaseClass D050197 8460149 1240 1252 exencephalus SpecificDisease D009436 8460149 1257 1270 hydrocephalus SpecificDisease D006849 10398279|t|Common mutations in BRCA1 and BRCA2 do not contribute to early prostate cancer in Jewish men. 10398279|a|BACKGROUND Families with a high incidence of hereditary breast cancer, and subsequently shown to have terminating mutations in BRCA1 or BRCA2, appear to have a higher incidence of prostate cancer among male relatives. We aimed to determine whether the common germline mutations of BRCA1 or BRCA2 in Ashkenazi Jewish men predisposed them to prostate cancer. METHODS We examined genomic DNA from 83 (for BRCA1 185delAG) or 82 (for BRCA2 6174delT) Ashkenazi Jewish prostate cancer patients, most of whom were treated at a relatively young age, for the most common germline mutation in each gene seen in the Ashkenazi population. RESULTS Our study should have been able to detect a 4-5-fold increase in the risk of prostate cancer due to mutation of BRCA1 or BRCA2. However, only one (1. 15%; 95% confidence interval, 0-3. 6%) of the patients was heterozygous for the BRCA1 mutant allele, and only two were heterozygous for the BRCA2 mutation (2. 4%; 95% confidence interval, 0-6. 2%). CONCLUSIONS The incidence of each of the germline mutations in these prostate cancer patients closely matched their incidence (about 1%) in the general Ashkenazi Jewish population. This suggests that unlike cases of breast and ovarian cancers, mutations in BRCA1 or BRCA2 do not significantly predispose men to prostate cancer 10398279 63 78 prostate cancer SpecificDisease D011471 10398279 140 164 hereditary breast cancer SpecificDisease D001943 10398279 275 290 prostate cancer SpecificDisease D011471 10398279 435 450 prostate cancer SpecificDisease D011471 10398279 558 573 prostate cancer Modifier D011471 10398279 808 823 prostate cancer SpecificDisease D011471 10398279 1149 1164 prostate cancer Modifier D011471 10398279 1296 1322 breast and ovarian cancers CompositeMention D001943|D010051 10398279 1391 1406 prostate cancer SpecificDisease D011471 3565372|t|Two new variants of glucose-6-phosphate dehydrogenase associated with hereditary non-spherocytic hemolytic anemia: G6PD Wayne and G6PD Huron. 3565372|a|Two new deficient variants of glucose-6-phosphate dehydrogenase (G6PD) causing hereditary nonspherocytic hemolytic anemia (HNSHA) are described. Both of these are unique and they have been named G6PD Wayne and G6PD Huron. Patients with G6PD Wayne underwent splenectomy and no objective improvement was noted. The patients with G6PD Huron were under medical observation for a considerable period of time without the diagnosis of G6PD deficiency being entertained because the family was of Northern European origin. Since sporadic variants of G6PD causing HNSHA show no special racial predilection, the diagnosis of G6PD deficiency should always be considered in patients with this syndrome.. 3565372 70 113 hereditary non-spherocytic hemolytic anemia SpecificDisease D000746 3565372 221 263 hereditary nonspherocytic hemolytic anemia SpecificDisease D000746 3565372 265 270 HNSHA SpecificDisease D000746 3565372 570 585 G6PD deficiency SpecificDisease D005955 3565372 696 701 HNSHA SpecificDisease D000746 3565372 756 771 G6PD deficiency SpecificDisease D005955 6650504|t|The Tay-Sachs disease gene in North American Jewish populations: geographic variations and origin. 6650504|a|From data collected in a North American Tay-Sachs disease (TSD) heterozygote screening program, the TSD carrier frequency among 46, 304 Jewish individuals was found to be. 0324 (1 in 31 individuals). This frequency is consistent with earlier estimates based on TSD incidence data. TSD carrier frequencies were then examined by single country and single region of origin in 28, 029 Jews within this sample for whom such data were available for analysis. Jews with Polish and/or Russian ancestry constituted 88% of this sample and had a TSD carrier frequency of. 0327. No TSD carriers were observed among the 166 Jews of Near Eastern origins. Relative to Jews of Polish and Russian origins, there was at least a twofold increase in the TSD carrier frequency in Jews of Austrian, Hungarian, and Czechoslovakian origins (P less than. 005). These findings suggest that the TSD gene proliferated among the antecedents of modern Ashkenazi Jewry after the Second Diaspora (70 A. D.) and before their major migrations to regions of Poland and Russia (before 1100 A. D.). 6650504 4 21 Tay-Sachs disease Modifier D013661 6650504 139 156 Tay-Sachs disease Modifier D013661 6650504 158 161 TSD Modifier D013661 6650504 199 202 TSD Modifier D013661 6650504 360 363 TSD Modifier D013661 6650504 380 383 TSD Modifier D013661 6650504 634 637 TSD Modifier D013661 6650504 669 672 TSD Modifier D013661 6650504 833 836 TSD Modifier D013661 6650504 967 970 TSD Modifier D013661 8533757|t|Novel inherited mutations and variable expressivity of BRCA1 alleles, including the founder mutation 185delAG in Ashkenazi Jewish families. 8533757|a|Thirty-seven families with four or more cases of breast cancer or breast and ovarian cancer were analyzed for mutations in BRCA1. Twelve different germ-line mutations, four novel and eight previously observed, were detected in 16 families. Five families of Ashkenazi Jewish descent carried the 185delAG mutation and shared the same haplotype at eight polymorphic markers spanning approximately 850 kb at BRCA1. Expressivity of 185delAG in these families varied, from early-onset breast cancer without ovarian cancer. Mutation 4184delTCAA occurred independently in two families. In one family, penetrance was complete, with females developing early-onset breast cancer or ovarian cancer and the male carrier developing prostatic cancer, whereas, in the other family, penetrance was incomplete and only breast cancer occurred, diagnosed at ages 38-81 years. Two novel nonsense mutations led to the loss of mutant BRCA1 transcript in families with 10 and 6 cases of early-onset breast cancer and ovarian cancer. A 665-nt segment of the BRCA1 3-UTR and 1. 3 kb of genomic sequence including the putative promoter region were invariant by single-strand conformation analysis in 13 families without coding-sequence mutations. Overall in our series, BRCA1 mutations have been detected in 26 families 16 with positive BRCA1 lod scores, 7 with negative lod scores (reflecting multiple sporadic breast cancers), and 3 not tested for linkage. Three other families have positive lod scores for linkage to BRCA2, but 13 families without detected BRCA1 mutations have negative lod scores for both BRCA1 and BRCA2. 8533757 189 202 breast cancer SpecificDisease D001943 8533757 206 231 breast and ovarian cancer CompositeMention D001943+D010051 8533757 619 632 breast cancer SpecificDisease D001943 8533757 641 655 ovarian cancer SpecificDisease D010051 8533757 794 807 breast cancer SpecificDisease D001943 8533757 811 825 ovarian cancer SpecificDisease D010051 8533757 858 874 prostatic cancer SpecificDisease D011471 8533757 941 954 breast cancer SpecificDisease D001943 8533757 1115 1128 breast cancer SpecificDisease D001943 8533757 1133 1147 ovarian cancer SpecificDisease D010051 8533757 1526 1540 breast cancers SpecificDisease D001943 10767343|t|Transgenic mice carrying large human genomic sequences with expanded CTG repeat mimic closely the DM CTG repeat intergenerational and somatic instability. 10767343|a|Myotonic dystrophy (DM) is caused by a CTG repeat expansion in the 3UTR of the DM protein kinase (DMPK) gene. A very high level of instability is observed through successive generations and the size of the repeat is generally correlated with the severity of the disease and with age at onset. Furthermore, tissues from DM patients exhibit somatic mosaicism that increases with age. We generated transgenic mice carrying large human genomic sequences with 20, 55 or > 300 CTG, cloned from patients from the same affected DM family. Using large human flanking sequences and a large amplification, we demonstrate that the intergenerational CTG repeat instability is reproduced in mice, with a strong bias towards expansions and with the same sex- and size-dependent characteristics as in humans. Moreover, a high level of instability, increasing with age, can be observed in tissues and in sperm. Although we did not observe dramatic expansions (or big jumps over several hundred CTG repeats) as in congenital forms of DM, our model carrying > 300 CTG is the first to show instability so close to the human DM situation. Our three models carrying different sizes of CTG repeat provide insight on the different factors modulating the CTG repeat instability.. 10767343 98 100 DM Modifier D009223 10767343 155 173 Myotonic dystrophy SpecificDisease D009223 10767343 175 177 DM SpecificDisease D009223 10767343 234 236 DM Modifier D009223 10767343 474 476 DM Modifier D009223 10767343 675 677 DM Modifier D009223 10767343 1171 1173 DM SpecificDisease D009223 10767343 1259 1261 DM Modifier D009223 133535|t|Linkage of gene for C2 deficiency and the major histocompatibility complex MHC in man. Family study of a further case. 133535|a|Close linkage between HL-A and C2 deficiency was first reported by FU and co-workers in 1974. We present here a pedigree of a 31-year-old C2-deficient individual with clinical manifestations of Hodgkins disease. The following markers were tested C2 levels, factor B polymorphism, blood groups, and enzyme typing. In addition to close linkage between HL-A and C2 deficiency, both parents were heterozygous for Bf (HL-A linked, electrophoretic variation of B). The two HL-A haplotypes closely linked to C2 deficiency are different 2, W18 and W24, W18. They share, however, the SD2 antigen W18 and the LD type 7a.. 133535 20 33 C2 deficiency SpecificDisease OMIM:217000 133535 150 163 C2 deficiency SpecificDisease OMIM:217000 133535 257 269 C2-deficient Modifier OMIM:217000 133535 313 329 Hodgkins disease SpecificDisease D006689 133535 479 492 C2 deficiency SpecificDisease OMIM:217000 133535 621 634 C2 deficiency SpecificDisease OMIM:217000 10051007|t|Age of onset in Huntington disease : sex specific influence of apolipoprotein E genotype and normal CAG repeat length. 10051007|a|Age of onset (AO) of Huntington disease (HD) is known to be correlated with the length of an expanded CAG repeat in the HD gene. Apolipoprotein E (APOE) genotype, in turn, is known to influence AO in Alzheimer disease, rendering the APOE gene a likely candidate to affect AO in other neurological diseases too. We therefore determined APOE genotype and normal CAG repeat length in the HD gene for 138 HD patients who were previously analysed with respect to CAG repeat length. Genotyping for APOE was performed blind to clinical information. In addition to highlighting the effect of the normal repeat length upon AO in maternally inherited HD and in male patients, we show that the APOE epsilon2epsilon3 genotype is associated with significantly earlier AO in males than in females. Such a sex difference in AO was not apparent for any of the other APOE genotypes. Our findings suggest that subtle differences in the course of the neurodegeneration in HD may allow interacting genes to exert gender specific effects upon AO. 10051007 16 34 Huntington disease SpecificDisease D006816 10051007 140 158 Huntington disease SpecificDisease D006816 10051007 160 162 HD SpecificDisease D006816 10051007 239 241 HD Modifier D006816 10051007 319 336 Alzheimer disease SpecificDisease D000544 10051007 403 424 neurological diseases DiseaseClass D020271 10051007 504 506 HD Modifier D006816 10051007 520 522 HD Modifier D006816 10051007 760 762 HD SpecificDisease D006816 10051007 1051 1068 neurodegeneration DiseaseClass D019636 10051007 1073 1075 HD SpecificDisease D006816 1978564|t|Genetic linkage map of six polymorphic DNA markers around the gene for familial adenomatous polyposis on chromosome 5. 1978564|a|A genetic linkage map of six polymorphic DNA markers close to the gene (APC) for familial adenomatous polyposis (FAP) on chromosome 5q is reported. One hundred fifty-five typed members of nine FAP kindred provided more than 90 meioses for linkage analysis. A number of crucial recombination events have been identified which are informative at three or more loci, allowing confident ordering of parts of the map. There was no evidence of genetic heterogeneity, with all families showing linkage of at least one chromosome 5 marker to the gene. Recombination data and two-point linkage analysis support a locus order of centromere-pi 227-C11P11-ECB27-L5. 62-APC-EF5 62-APC-EF5. 44-YN5 44-YN5. 48-telomer e, although EF5. 44 could lie in the interval L5. 62-APC or ECB27-L5. 62. No recombinants were identified between APC and either EF5. 44 or YN5. 48, but published deletion mapping in colorectal carcinomas and linkage analysis in FAP suggest that YN5. 48 is 1-3 cM from APC. The present study suggests that YN5. 48 and L5. 62 delineate a small region of chromosome 5 within which the EF5. 44 locus lies very close to the APC gene. These data not only allow use of flanking markers for presymptomatic diagnosis of FAP but also provide a high-density map of the region for isolation of the APC gene itself and for further assessment of the role of chromosome 5 deletions in the biology of sporadic colorectal cancer. 1978564 71 101 familial adenomatous polyposis SpecificDisease D011125 1978564 200 230 familial adenomatous polyposis SpecificDisease D011125 1978564 232 235 FAP SpecificDisease D011125 1978564 312 315 FAP Modifier D011125 1978564 1005 1026 colorectal carcinomas SpecificDisease D015179 1978564 1051 1054 FAP SpecificDisease D011125 1978564 1242 1245 APC Modifier D011125 1978564 1334 1337 FAP SpecificDisease D011125 1978564 1409 1412 APC Modifier D011125 1978564 1517 1534 colorectal cancer SpecificDisease D015179 8370681|t|Detection of a novel arginine vasopressin defect by dideoxy fingerprinting. 8370681|a|Autosomal dominant neurohypophyseal diabetes insipidus is a familial form of diabetes insipidus. This disorder is associated with variable levels of arginine vasopressin (AVP) and diabetes insipidus of varying severity, which responds to exogenous AVP. To determine the molecular basis of autosomal dominant neurohypophyseal diabetes insipidus, the AVP genes of members of a large kindred were analyzed. A new method, called dideoxy fingerprinting, was used to detect an AVP mutation that was characterized by DNA sequencing. The novel defect found changes the last codon of the AVP signal peptide from alanine to threonine, which should perturb cleavage of mature AVP from its precursor protein and inhibit its secretion or action.. 8370681 76 130 Autosomal dominant neurohypophyseal diabetes insipidus SpecificDisease OMIM:125700 8370681 153 171 diabetes insipidus DiseaseClass D003919 8370681 256 274 diabetes insipidus DiseaseClass D003919 8370681 365 419 autosomal dominant neurohypophyseal diabetes insipidus SpecificDisease OMIM:125700 8259519|t|Association of the APC tumor suppressor protein with catenins. 8259519|a|Mutations of APC appear to initiate sporadic and inherited forms of human colorectal cancer. Although these mutations have been well characterized, little is known about the function of the APC gene product. Two cellular proteins that associate with APC were identified by nucleotide sequence analysis and peptide mapping as the E-cadherin-associated proteins alpha- and beta-catenin. A 27-residue fragment of APC containing a 15-amino acid repeat was sufficient for the interaction with the catenins. These results suggest an important link between tumor initiation and cell adhesion.. 8259519 19 28 APC tumor Modifier D011125 8259519 137 154 colorectal cancer SpecificDisease D015179 8259519 253 256 APC Modifier D011125 8259519 313 316 APC SpecificDisease D011125 8259519 613 618 tumor Modifier D009369 2703233|t|Myotonic dystrophy is closely linked to the gene for muscle-type creatine kinase (CKMM). 2703233|a|We have studied genetic linkage between the gene for creatine kinase muscle type (CKMM) and the gene for myotonic dystrophy (DM). In a panel of 65 myotonic dystrophy families from Canada and the Netherlands, a maximum lod score (Zmax) of 22. 8 at a recombination frequency (theta) of 0. 03 was obtained. Tight linkage was also demonstrated for CKMM and the gene for apolipoprotein C2 (ApoC2). This establishes CKMM as a useful marker for myotonic dystrophy 2703233 0 18 Myotonic dystrophy SpecificDisease D009223 2703233 194 212 myotonic dystrophy SpecificDisease D009223 2703233 214 216 DM SpecificDisease D009223 2703233 236 254 myotonic dystrophy Modifier D009223 2703233 527 545 myotonic dystrophy SpecificDisease D009223 7767095|t|Discordant clinical outcome in myotonic dystrophy relatives showing (CTG)n > 700 repeats. 7767095|a|A myotonic dystrophy (DM) family is described in which discordant DM phenotypes were found in the children of two affected sisters with similar CTG expansion and clinical manifestations. In this family, congenital as well as early severe childhood and later childhood onset DM coexist. This observation strengthens the limited ability of lymphocytes CTG repeat number analysis in predicting genotype-phenotype correlations in DM patients.. 7767095 31 49 myotonic dystrophy Modifier D009223 7767095 92 110 myotonic dystrophy Modifier D009223 7767095 112 114 DM Modifier D009223 7767095 156 158 DM Modifier D009223 7767095 364 366 DM SpecificDisease D009223 7767095 516 518 DM Modifier D009223 10554035|t|Constitutional von Hippel-Lindau (VHL) gene deletions detected in VHL families by fluorescence in situ hybridization. 10554035|a|von Hippel-Lindau (VHL) disease is an autosomal dominantly inherited cancer syndrome predisposing to a variety of tumor types that include retinal hemangioblastomas, hemangioblastomas of the central nervous system, renal cell carcinomas, pancreatic cysts and tumors, pheochromocytomas, endolymphatic sac tumors, and epididymal cystadenomas [W. M. Linehan et al., J. Am. Med. Assoc., 273 564-570, 1995; E. A. Maher and W. G. Kaelin, Jr., Medicine (Baltimore), 76 381-391, 1997; W. M. Linehan and R. D. Klausner, In B. Vogelstein and K. Kinzler (eds.), The Genetic Basis of Human Cancer, pp. 455-473, McGraw-Hill, 1998]. The VHL gene was localized to chromosome 3p25-26 and cloned [F. Latif et al., Science (Washington DC), 260 1317-1320, 1993]. Germline mutations in the VHL gene have been detected in the majority of VHL kindreds. The reported frequency of detection of VHL germline mutations has varied from 39 to 80% (J. M. Whaley et al., Am. J. Hum. Genet., 55 1092-1102, 1994; Clinical Research Group for Japan, Hum. Mol. Genet., 4 2233-2237, 1995; F. Chen et al., Hum. Mutat., 5 66-75, 1995; E. R. Maher et al., J. Med. Genet., 33 328-332, 1996; B. Zbar, Cancer Surv., 25 219-232, 1995). Recently a quantitative Southern blotting procedure was found to improve this frequency (C. Stolle et al., Hum. Mutat., 12 417-423, 1998). In the present study, we report the use of fluorescence in situ hybridization (FISH) as a method to detect and characterize VHL germline deletions. We reexamined a group of VHL patients shown previously by single-strand conformation and sequencing analysis not to harbor point mutations in the VHL locus. We found constitutional deletions in 29 of 30 VHL patients in this group using cosmid and P1 probes that cover the VHL locus. We then tested six phenotypically normal offspring from four of these VHL families two were found to carry the deletion and the other four were deletion-free. In addition, germline mosaicism of the VHL gene was identified in one family. In sum, FISH was found to be a simple and reliable method to detect VHL germline deletions and practically useful in cases where other methods of screening have failed to detect a VHL gene abnormality 10554035 15 32 von Hippel-Lindau Modifier D006623 10554035 34 37 VHL Modifier D006623 10554035 66 69 VHL Modifier D006623 10554035 118 149 von Hippel-Lindau (VHL) disease SpecificDisease D006623 10554035 156 202 autosomal dominantly inherited cancer syndrome DiseaseClass D009386 10554035 232 237 tumor Modifier D009369 10554035 265 282 hemangioblastomas DiseaseClass D018325 10554035 284 301 hemangioblastomas DiseaseClass D018325 10554035 333 354 renal cell carcinomas SpecificDisease D002292 10554035 356 383 pancreatic cysts and tumors CompositeMention D010181|D010190 10554035 385 402 pheochromocytomas DiseaseClass D010673 10554035 404 428 endolymphatic sac tumors DiseaseClass D036821 10554035 434 457 epididymal cystadenomas DiseaseClass D003537 10554035 744 747 VHL Modifier D006623 10554035 892 895 VHL Modifier D006623 10554035 939 942 VHL Modifier D006623 10554035 992 995 VHL Modifier D006623 10554035 1584 1587 VHL Modifier D006623 10554035 1633 1636 VHL Modifier D006623 10554035 1754 1757 VHL Modifier D006623 10554035 1811 1814 VHL Modifier D006623 10554035 1880 1883 VHL Modifier D006623 10554035 1961 1964 VHL Modifier D006623 10554035 2090 2093 VHL Modifier D006623 10554035 2197 2200 VHL Modifier D006623 10554035 2309 2329 VHL gene abnormality SpecificDisease D006623 7481765|t|Aberrant subcellular localization of BRCA1 in breast cancer. 7481765|a|The BRCA1 gene product was identified as a 220-kilodalton nuclear phosphoprotein in normal cells, including breast ductal epithelial cells, and in 18 of 20 tumor cell lines derived from tissues other than breast and ovary. In 16 of 17 breast and ovarian cancer lines and 17 of 17 samples of cells obtained from malignant effusions, however, BRCA1 localized mainly in cytoplasm. Absence of BRCA1 or aberrant subcellular location was also observed to a variable extent in histological sections of many breast cancer biopsies. These findings suggest that BRCA1 abnormalities may be involved in the pathogenesis of many breast cancers, sporadic as well as familial.. 7481765 46 59 breast cancer SpecificDisease D001943 7481765 217 222 tumor Modifier D009369 7481765 296 321 breast and ovarian cancer Modifier D001943|D010051 7481765 561 574 breast cancer Modifier D001943 7481765 613 632 BRCA1 abnormalities DiseaseClass OMIM:604370 7481765 677 691 breast cancers SpecificDisease D001943 107868|t|HLA B27 and the genetics of ankylosing spondylitis. 107868|a|One hundred and twenty-eight of 145 patients with ankylosing spondylitis (AS) were found to be HLA B27 positive. Five patients had evidence of a sero-negative peripheral arthritis resembling peripheral psoriatic arthritis and 3 of these were B27 negative. One further B27 negative patients had a sister with ankylosing spondylitis and ulcerative colitis and a mother with ulcerative colitis. There was evidence of a somewhat later age of onset of symptoms in B27 negative patients. These findings are interpreted as suggesting some degree of clinical and genetic heterogeneity in ankylosing spondylitis with genes for psoriasis and inflammatory bowel disease being important in some individuals, particularly those who are B27 negative. Twenty-five first-degree relatives with ankylosing spondylitis were all B27 positive. The only instance of disassociation of B27 and spondylitis in a family was where the proband had ulcerative colitis as well as spondylitis. Of 13 B27 positive fathers 3 could be diagnosed as having definite ankylosing spondylitis (23%). These findings are thought to provide evidence against the concept that the gene for ankylosing spondylitis is not B27 but a closely linked gene and favour the occurrence of an environmental event affecting approximately one-fifth of B27 positive males to result in disease.. 107868 28 50 ankylosing spondylitis SpecificDisease D013167 107868 102 124 ankylosing spondylitis SpecificDisease D013167 107868 126 128 AS SpecificDisease D013167 107868 211 231 peripheral arthritis SpecificDisease D001168 107868 243 273 peripheral psoriatic arthritis SpecificDisease D015535 107868 360 382 ankylosing spondylitis SpecificDisease D013167 107868 387 405 ulcerative colitis SpecificDisease D003093 107868 424 442 ulcerative colitis SpecificDisease D003093 107868 632 654 ankylosing spondylitis SpecificDisease D013167 107868 670 679 psoriasis SpecificDisease D011565 107868 684 710 inflammatory bowel disease SpecificDisease D015212 107868 829 851 ankylosing spondylitis SpecificDisease D013167 107868 922 933 spondylitis SpecificDisease D013166 107868 972 990 ulcerative colitis SpecificDisease D003093 107868 1002 1013 spondylitis SpecificDisease D013166 107868 1082 1104 ankylosing spondylitis SpecificDisease D013167 107868 1197 1219 ankylosing spondylitis SpecificDisease D013167 7858169|t|Late infantile metachromatic leukodystrophy in Israel. 7858169|a|Metachromatic Leukodystrophy (MLD) is a neurodegenerative disease in which the lysosomal enzyme, Aryl sulfatase A (ARSA) is deficient. The disease is inherited as an autosomal recessive trait and its frequency is estimated to be 1/40, 000 live births. The gene of ARSA has been cloned and up to now eight mutations causing MLD have been reported. Another mutation, PD, leads to the deficiency of the enzyme in vitro (pseudodeficiency) without any known clinical effect. The PD mutation is frequent in all populations. In Israel, late infantile MLD was found to be very frequent in a small Jewish isolate, the Habbanite Jews (1/75 live births). The molecular analysis demonstrated that in the Habbanite population, the mutation occurred on an allele with the PD mutation. The loss of ARSA activity is due to a point mutation C > T leading to a change of proline to leucine. MLD is also frequent among Moslem Arabs in Jerusalem. The mutation is a transition G > A destroying the splice donor site of exon 2. This mutation has been reported in patients with the late infantile MLD from different ethnic groups. The Christian Arabs in Israel also have a high incidence of the disease (1/10, 000 live births); the mutation in this population is still unknown. Knowledge of the different mutations causing MLD in these defined populations will allow a carrier screening program to be carried out and prevent the birth of additional affected children.. 7858169 15 43 metachromatic leukodystrophy SpecificDisease D007966 7858169 55 83 Metachromatic Leukodystrophy SpecificDisease D007966 7858169 85 88 MLD SpecificDisease D007966 7858169 95 120 neurodegenerative disease DiseaseClass D019636 7858169 378 381 MLD SpecificDisease D007966 7858169 599 602 MLD SpecificDisease D007966 7858169 928 931 MLD SpecificDisease D007966 7858169 1129 1132 MLD SpecificDisease D007966 7858169 1355 1358 MLD SpecificDisease D007966 10982189|t|Genomic rearrangements of the APC tumor-suppressor gene in familial adenomatous polyposis. 10982189|a|Germline mutations of the adenomatous polyposis coli (APC) tumor-suppressor gene result in the hereditary colorectal cancer syndrome familial adenomatous polyposis (FAP). Almost all APC mutations that have been identified are single-nucleotide alterations, small insertions, or small deletions that would truncate the protein product of the gene. No well-characterized intragenic rearrangement of APC has been described, and the prevalence of this type of mutation in FAP patients is not clear. We screened 49 potential FAP families and identified 26 different germline APC mutations in 30 families. Four of these mutations were genomic rearrangements resulting from homologous and nonhomologous recombinations mediated by Alu elements. Two of these four rearrangements were complex, involving deletion and insertion of nucleotides. Of these four rearrangements, one resulted in the deletion of exons 11 and 12 and two others resulted in either complete or partial deletion of exon 14. The fourth rearrangement grossly altered the sequence within intron 14. Although this rearrangement did not affect any coding sequence of APC at the genomic DNA level, it caused inappropriate splicing of exon 14. These rearrangements were initially revealed by analyzing cDNAs and could not have been identified by using mutation detection methods that screened each exon individually. The identification of a rearrangement that did not alter any coding exons yet affected the splicing further underscores the importance of using cDNA for mutation analysis. The identification of four genomic rearrangements among 30 mutations suggests that genomic rearrangements are frequent germline APC mutations.. 10982189 30 39 APC tumor Modifier D011125 10982189 59 89 familial adenomatous polyposis SpecificDisease D011125 10982189 117 155 adenomatous polyposis coli (APC) tumor Modifier D011125 10982189 186 223 hereditary colorectal cancer syndrome SpecificDisease D015179 10982189 224 254 familial adenomatous polyposis SpecificDisease D011125 10982189 256 259 FAP SpecificDisease D011125 10982189 273 276 APC Modifier D011125 10982189 559 562 FAP Modifier D011125 10982189 611 614 FAP Modifier D011125 10982189 661 664 APC Modifier D011125 10982189 1763 1766 APC Modifier D011125 8075631|t|Familial male breast cancer is not linked to the BRCA1 locus on chromosome 17q. 8075631|a|Breast cancer in men is about a hundredfold less common than in women and this has hindered research into its genetic basis. We have examined 22 families with at least one case of male breast cancer for linkage to the hereditary breast and ovarian cancer locus, BRCA1, on chromosome 17q. We found strong evidence against linkage to BRCA1 (lod score-16. 63) and the best estimate of the proportion of linked families was 0% (95% CI 0-18%). Our results indicate that there is a gene (s) other than BRCA1 which predisposes to early-onset breast cancer in women and which confers a higher risk of male breast cancer. Identification of additional pedigrees that include cases of male breast cancer may therefore facilitate the mapping and isolation of this gene. 8075631 0 27 Familial male breast cancer SpecificDisease D018567 8075631 80 93 Breast cancer SpecificDisease D001943 8075631 260 278 male breast cancer SpecificDisease D018567 8075631 298 334 hereditary breast and ovarian cancer Modifier D061325 8075631 615 628 breast cancer SpecificDisease D001943 8075631 673 691 male breast cancer SpecificDisease D018567 8075631 754 772 male breast cancer SpecificDisease D018567 8528200|t|Evidence for inter-generational instability in the CAG repeat in the MJD1 gene and for conserved haplotypes at flanking markers amongst Japanese and Caucasian subjects with Machado-Joseph disease. 8528200|a|The size of the (CAG)n repeat array in the 3' end of the MJD1 gene and the haplotype at a series of microsatellite markers surrounding the MJD1 gene were examined in a large cohort of Japanese and Caucasian subjects affected with Machado-Joseph disease (MJD). Our data provide five novel observations. First, MJD is associated with expansion fo the array from the normal range of 14-37 repeats to 68-84 repeats in most Japanese and Caucasian subjects, but no subjects were observed with expansions intermediate in size between those of the normal and MJD affected groups. Second, the expanded allele associated with MJD displays inter-generational instability, particularly in male meioses, and this instability was associated with the clinical phenomenon of anticipation. Third, the size of the expanded allele is not only inversely correlated with the age-of-onset of MJD (r = -0.738, p < 0.001), but is also correlated with the frequency of other clinical features [e.g. pseudoexophthalmos and pyramidal signs were more frequent in subjects with large repeats (p < 0.001 and p < 0.05 respectively)]. Fourth, the disease phenotype is significantly more severe and had an early age of onset (16 years) in a subject homozygous for the expanded allele, which contrasts with Huntington disease and suggests that the expanded allele in the MJD1 gene could exert its effect either by a dominant negative effect (putatively excluded in HD) or by a gain of function effect as proposed for HD. Finally, Japanese and Caucasian subjects affected with MJD share haplotypes at several markers surrounding the MJD1 gene, which are uncommon in the normal Japanese and Caucasian population, and which suggests the existence either of common founders in these populations or of chromosomes susceptible to pathologic expansion of the CAG repeat in the MJD1 gene. 8528200 173 195 Machado-Joseph disease SpecificDisease D017827 8528200 427 449 Machado-Joseph disease SpecificDisease D017827 8528200 451 454 MJD SpecificDisease D017827 8528200 506 509 MJD SpecificDisease D017827 8528200 748 751 MJD Modifier D017827 8528200 813 816 MJD SpecificDisease D017827 8528200 1067 1070 MJD SpecificDisease D017827 8528200 1470 1488 Huntington disease SpecificDisease D006816 8528200 1628 1630 HD SpecificDisease D006816 8528200 1680 1682 HD SpecificDisease D006816 8528200 1739 1742 MJD SpecificDisease D017827 3674116|t|Familial Prader-Willi syndrome with apparently normal chromosomes. 3674116|a|We report on 4 sibs (2F, 2M) with Prader-Willi syndrome (PWS). Diagnosis was made clinically on the basis of history, behavior, and physical findings in 3 of the sibs. The other child had died at age 10 months with a history and clinical findings typical of first phase of PWS. Results of chromosome studies on the parents and surviving sibs were normal. The implications of this unusual familial occurrence for our understanding of PWS are discussed.. 3674116 9 30 Prader-Willi syndrome SpecificDisease D011218 3674116 101 122 Prader-Willi syndrome SpecificDisease D011218 3674116 124 127 PWS SpecificDisease D011218 3674116 340 343 PWS SpecificDisease D011218 3674116 500 503 PWS SpecificDisease D011218 7759075|t|Age at diagnosis as an indicator of eligibility for BRCA1 DNA testing in familial breast cancer. 7759075|a|We searched for criteria that could indicate breast cancer families with a high prior probability of being caused by the breast/ovarian cancer susceptibility locus BRCA1 on chromosome 17. To this end, we performed a linkage study with 59 consecutively collected Dutch breast cancer families, including 16 with at least one case of ovarian cancer. We used an intake cut-off of at least three first-degree relatives with breast and/or ovarian cancer at any age. Significant evidence for linkage was found only among the 13 breast cancer families with a mean age at diagnosis of less than 45 years. An unexpectedly low proportion of the breast-ovarian cancer families were estimated to be linked to BRCA1, which could be due to a founder effect in the Dutch population. Given the expected logistical problems in clinical management now that BRCA1 has been identified, we propose an interim period in which only families with a strong positive family history for early onset breast and/or ovarian cancer will be offered BRCA1 mutation testing.. 7759075 73 95 familial breast cancer SpecificDisease D001943 7759075 142 155 breast cancer Modifier D001943 7759075 218 239 breast/ovarian cancer Modifier D061325 7759075 365 378 breast cancer Modifier D001943 7759075 428 442 ovarian cancer SpecificDisease D010051 7759075 516 544 breast and/or ovarian cancer CompositeMention D001943|D010051 7759075 618 631 breast cancer Modifier D001943 7759075 731 752 breast-ovarian cancer Modifier D061325 7759075 1068 1096 breast and/or ovarian cancer CompositeMention D001943|D010051 1999339|t|Some Mexican glucose-6-phosphate dehydrogenase variants revisited. 1999339|a|Glucose-6-phosphate dehydrogenase (G6PD) deficiency appears to be fairly common in Mexico. We have now examined the DNA of three previously reported electrophoretically fast Mexican G6PD variants, -G6PD Distrito Federal, G6PD Tepic, and G6PD Castilla. All three of these variants, believed on the basis of biochemical characterization and population origin to be unique, have the G----A transition at nucleotide 202 and the A----G transition at nucleotide 376, mutations that we now recognize to be characteristic of G6PD A-. Two other Mexican males with G6PD deficiency were found to have the same mutation. All five have the (NlaIII/FokI/PvuII/PstI) haplotype characteristic of G6PD A -in Africa. Since the PvuII + genotype seems to be rare in Europe, we conclude that all of these G6PD A - genes had their ancient origin in Africa, although in many of the Mexican patients with G6PD A -202A/376G the gene may have been imported more recently from Spain, where this variant, formerly known as G6PD Betica, is also prevalent.. 1999339 67 118 Glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 1999339 622 637 G6PD deficiency SpecificDisease D005955 10924409|t|Inactivation of germline mutant APC alleles by attenuated somatic mutations: a molecular genetic mechanism for attenuated familial adenomatous polyposis. 10924409|a|Germline mutations of the adenomatous polyposis coli (APC) tumor-suppressor gene result in familial adenomatous polyposis (FAP). Patients with FAP typically develop hundreds to thousands of benign colorectal tumors and early-onset colorectal cancer. A subset of germline APC mutations results in an attenuated FAP (AFAP) phenotype, in which patients develop fewer tumors and develop them at an older age. Although a genotype-phenotype correlation between the locations of APC germline mutations and the development of AFAP has been well documented, the mechanism for AFAP has not been well defined. We investigated the mechanism for AFAP in patients carrying a mutant APC allele (APC (AS9)) that has a mutation in the alternatively spliced region of exon 9. APC (AS9) was found to down-regulate beta-catenin-regulated transcription, the major tumor-suppressor function of APC, as did the wild-type APC. Mutation analysis showed that both APC (AS9) and the wild-type APC alleles were somatically mutated in most colorectal tumors from these patients. Functional analysis showed that 4666insA, a common somatic mutation in APC (AS9) in these tumors, did not inactivate the wild-type APC. Our results indicate that carriers of APC (AS9) develop fewer colorectal tumors than do typical patients with FAP because somatic inactivation of both APC alleles is necessary for colorectal tumorigenesis. However, these patients develop colorectal tumors more frequently than does the general population because APC (AS9) is inactivated by mutations that do not inactivate the wild-type APC.. 10924409 32 35 APC Modifier D011125 10924409 111 152 attenuated familial adenomatous polyposis SpecificDisease C538265 10924409 180 218 adenomatous polyposis coli (APC) tumor Modifier D011125 10924409 245 275 familial adenomatous polyposis SpecificDisease D011125 10924409 277 280 FAP SpecificDisease D011125 10924409 297 300 FAP SpecificDisease D011125 10924409 344 368 benign colorectal tumors SpecificDisease D015179 10924409 385 402 colorectal cancer SpecificDisease D015179 10924409 425 428 APC Modifier D011125 10924409 453 467 attenuated FAP Modifier C538265 10924409 469 473 AFAP Modifier C538265 10924409 518 524 tumors DiseaseClass D009369 10924409 626 629 APC Modifier D011125 10924409 672 676 AFAP SpecificDisease C538265 10924409 721 725 AFAP SpecificDisease C538265 10924409 787 791 AFAP SpecificDisease C538265 10924409 822 825 APC Modifier D011125 10924409 997 1002 tumor Modifier D009369 10924409 1120 1123 APC Modifier D011125 10924409 1165 1182 colorectal tumors SpecificDisease D015179 10924409 1294 1300 tumors DiseaseClass D009369 10924409 1402 1419 colorectal tumors SpecificDisease D015179 10924409 1450 1453 FAP SpecificDisease D011125 10924409 1491 1494 APC Modifier D011125 10924409 1578 1595 colorectal tumors SpecificDisease D015179 1334370|t|Submicroscopic deletions at the WAGR locus, revealed by nonradioactive in situ hybridization. 1334370|a|Fluorescence in situ hybridization (FISH) with biotin-labeled probes mapping to 11p13 has been used for the molecular analysis of deletions of the WAGR (Wilms tumor, aniridia, genitourinary abnormalities, and mental retardation) locus. We have detected a submicroscopic 11p13 deletion in a child with inherited aniridia who subsequently presented with Wilms tumor in a horseshoe kidney, only revealed at surgery. The mother, who has aniridia, was also found to carry a deletion including both the aniridia candidate gene (AN2) and the Wilms tumor predisposition gene (WT1). This is therefore a rare case of an inherited WAGR deletion. Wilms tumor has so far only been associated with sporadic de novo aniridia cases. We have shown that a cosmid probe for a candidate aniridia gene, homologous to the mouse Pax-6 gene, is deleted in cell lines from aniridia patients with previously characterized deletions at 11p13, while another cosmid marker mapping between two aniridia-associated translocation breakpoints (and hence a second candidate marker) is present on both chromosomes. These results support the Pax-6 homologue as a strong candidate for the AN2 gene. FISH with cosmid probes has proved to be a fast and reliable technique for the molecular analysis of deletions. It can be used with limited amounts of material and has strong potential for clinical applications.. 1334370 32 36 WAGR Modifier D017624 1334370 241 245 WAGR Modifier D017624 1334370 247 258 Wilms tumor Modifier D009396 1334370 260 268 aniridia Modifier D015783 1334370 270 297 genitourinary abnormalities Modifier D014564 1334370 303 321 mental retardation Modifier D008607 1334370 405 413 aniridia SpecificDisease D015783 1334370 446 457 Wilms tumor SpecificDisease D009396 1334370 527 535 aniridia SpecificDisease D015783 1334370 591 599 aniridia Modifier D015783 1334370 629 640 Wilms tumor Modifier D009396 1334370 714 718 WAGR Modifier D017624 1334370 729 740 Wilms tumor SpecificDisease D009396 1334370 795 803 aniridia Modifier D015783 1334370 861 869 aniridia Modifier D015783 1334370 942 950 aniridia Modifier D015783 1334370 1058 1066 aniridia Modifier D015783 8279472|t|Haplotype studies in Wilson disease. 8279472|a|In 51 families with Wilson disease, we have studied DNA haplotypes of dinucleotide repeat polymorphisms (CA repeats) in the 13q14. 3 region, to examine these markers for association with the Wilson disease gene (WND). In addition to a marker (D13S133) described elsewhere, we have developed three new highly polymorphic markers (D13S314, D13S315, and D13S316) close to the WND locus. We have examined the distribution of marker alleles at the loci studied and have found that D13S314, D13S133, and D13S316 each show nonrandom distribution on chromosomes carrying the WND mutation. We have studied haplotypes of these three markers and have found that there are highly significant differences between WND and normal haplotypes in northern European families. These findings have important implications for mutation detection and molecular diagnosis in families with Wilson disease. 8279472 21 35 Wilson disease SpecificDisease D006527 8279472 57 71 Wilson disease SpecificDisease D006527 8279472 228 242 Wilson disease Modifier D006527 8279472 249 252 WND SpecificDisease D006527 8279472 410 413 WND SpecificDisease D006527 8279472 604 607 WND SpecificDisease D006527 8279472 737 740 WND SpecificDisease D006527 8279472 901 915 Wilson disease SpecificDisease D006527 10541953|t|Growth hormone treatment increases CO(2) response, ventilation and central inspiratory drive in children with Prader-Willi syndrome. 10541953|a|We studied whether the beneficial effects of growth hormone (GH) treatment on growth and body composition in PWS are accompanied by an improvement in respiratory function. We measured resting ventilation, airway occlusion pressure (P (0. 1)) and ventilatory response to CO (2) in nine children, aged 7-14 years, before and 6-9 months after the start of GH treatment. During GH treatment, resting ventilation increased by 26%, P (0. 1) by 72% and the response to CO (2) by 65% (P < 0. 002, < 0. 04 and < 0. 02, respectively). This observed increase in ventilatory output was not correlated to changes in body mass index. CONCLUSION Treatment of children with Prader-Willi syndrome (PWS) seems to have a stimulatory effect on central respiratory structures. The observed increase in ventilation and inspiratory drive may contribute to the improved activity level reported by parents of PWS children during growth hormone therapy 10541953 110 131 Prader-Willi syndrome SpecificDisease D011218 10541953 242 245 PWS SpecificDisease D011218 10541953 792 813 Prader-Willi syndrome SpecificDisease D011218 10541953 815 818 PWS SpecificDisease D011218 10541953 1018 1021 PWS Modifier D011218 8522307|t|Somatic von Hippel-Lindau mutation in clear cell papillary cystadenoma of the epididymis. 8522307|a|Papillary cystadenoma of the epididymis is an uncommon benign lesion that may occur sporadically or as a manifestation of von Hippel-Lindau (VHL) disease. Neither immunohistochemical studies nor molecular genetic analyses of the VHL gene have been reported previously for this lesion. The authors describe two cases of clear cell papillary cystadenoma of the epididymis, both of which were initially confused with metastatic renal cell carcinoma. Both lesions showed positive immunohistochemical staining for low and intermediate molecular weight keratins (Cam 5. 2 and AE1/AE3), EMA, vimentin, alpha 1-antitrypsin, and alpha 1-antichymotrypsin. Each was negative for CEA. Because clear cell papillary cystadenoma is similar to renal cell carcinoma histologically, and because both occur as components of the von Hippel-Lindau disease complex, the authors analyzed both cases for the presence of mutations in the VHL gene. A somatic VHL gene mutation was detected in one of the two tumors by polymerase chain reaction followed by single-strand conformation polymorphism analysis. Direct sequencing revealed a cytosine to thymine transition at nucleotide 694, resulting in the replacement of an arginine with a stop codon after the sixth amino acid of exon 3. As the VHL gene is believed to function as a tumor suppressor gene, VHL gene mutations may play a role in the initiation of tumorigenesis in sporadic cystadenomas of the epididymis. 8522307 8 25 von Hippel-Lindau Modifier D006623 8522307 49 88 papillary cystadenoma of the epididymis SpecificDisease D018292 8522307 90 129 Papillary cystadenoma of the epididymis SpecificDisease D018292 8522307 212 243 von Hippel-Lindau (VHL) disease SpecificDisease D006623 8522307 319 322 VHL Modifier D006623 8522307 420 459 papillary cystadenoma of the epididymis SpecificDisease D018292 8522307 504 535 metastatic renal cell carcinoma SpecificDisease C538445 8522307 782 803 papillary cystadenoma SpecificDisease D018292 8522307 818 838 renal cell carcinoma SpecificDisease D002292 8522307 899 924 von Hippel-Lindau disease Modifier D006623 8522307 1003 1006 VHL Modifier D006623 8522307 1023 1026 VHL Modifier D006623 8522307 1072 1078 tumors DiseaseClass D009369 8522307 1356 1359 VHL Modifier D006623 8522307 1394 1399 tumor Modifier D009369 8522307 1417 1420 VHL Modifier D006623 8522307 1499 1529 cystadenomas of the epididymis SpecificDisease D003537 7825586|t|An evaluation of genetic heterogeneity in 145 breast-ovarian cancer families. Breast Cancer Linkage Consortium. 7825586|a|The breast-ovary cancer-family syndrome is a dominant predisposition to cancer of the breast and ovaries which has been mapped to chromosome region 17q12-q21. The majority, but not all, of breast-ovary cancer families show linkage to this susceptibility locus, designated BRCA1. We report here the results of a linkage analysis of 145 families with both breast and ovarian cancer. These families contain either a total of three or more cases of early-onset (before age 60 years) breast cancer or ovarian cancer. All families contained at least one case of ovarian cancer. Overall, an estimated 76% of the 145 families are linked to the BRCA1 locus. None of the 13 families with cases of male breast cancer appear to be linked, but it is estimated that 92% (95% confidence interval 76% -100%) of families with no male breast cancer and with two or more ovarian cancers are linked to BRCA1. These data suggest that the breast-ovarian cancer-family syndrome is genetically heterogeneous. However, the large majority of families with early-onset breast cancer and with two or more cases of ovarian cancer are likely to be due to BRCA1 mutations.. 7825586 46 67 breast-ovarian cancer Modifier D061325 7825586 78 91 Breast Cancer Modifier D001943 7825586 116 151 breast-ovary cancer-family syndrome SpecificDisease D061325 7825586 184 216 cancer of the breast and ovaries CompositeMention D001943|D010051 7825586 301 320 breast-ovary cancer Modifier D061325 7825586 466 491 breast and ovarian cancer CompositeMention D001943|D010051 7825586 591 604 breast cancer SpecificDisease D001943 7825586 608 622 ovarian cancer SpecificDisease D010051 7825586 668 682 ovarian cancer SpecificDisease D010051 7825586 799 817 male breast cancer SpecificDisease D018567 7825586 924 942 male breast cancer SpecificDisease D018567 7825586 964 979 ovarian cancers SpecificDisease D010051 7825586 1029 1066 breast-ovarian cancer-family syndrome SpecificDisease D061325 7825586 1154 1167 breast cancer SpecificDisease D001943 7825586 1198 1212 ovarian cancer SpecificDisease D010051 4019732|t|Genetic analysis in families with van der Woude syndrome. 4019732|a|We have brought together information on 864 affected individuals in 164 families (including three new pedigrees) reported in the 137 year period since 1845 when Demarquay first described a family with what was later called van der Woude syndrome (VWS). Both types of oral cleft, cleft palate (CP) and cleft lip with or without CP (CLP), segregate in these families together with lower lip pits or fistulae in an autosomal dominant mode with high penetrance estimated to be K =. 89 and. 99 by different methods. Cleft types (CLP and CP) occur in VWS in the same proportions as in the general non-VWS population, ie, about twice as many cleft-bearing individuals have CLP as have CP. On the other hand, we do not find the usually observed excess of females with CP and excess of males with CLP; in VWS the sex ratios are more nearly equal. Lip pits also are equally distributed between the sexes. Affected males and females are equally likely to transmit VWS. However, there is an excess of less severely affected individuals among transmitters and a deficiency of more severely affected, brought about by a proband bias and differential fecundity. The expression of VWS is significantly modified by the genetic background More extreme phenotypes in parents tend to produce more extreme expression in their children. For a VWS gene carrier the relative risk of transmitting a cleft is 26. 45%; that of transmitting lower lip pits is 23. 55%. Three pedigrees of lip pits in the literature show no clefts among a significant number of affected individuals. Control of gene expression in VWS in the three target tissues appears to be independent and separately designated. Mutation rate of the VWS gene is calculated to be 1. 8 X 10 (-5) 4019732 34 56 van der Woude syndrome SpecificDisease C536528 4019732 281 303 van der Woude syndrome SpecificDisease C536528 4019732 305 308 VWS SpecificDisease C536528 4019732 325 335 oral cleft SpecificDisease D002971|D002972 4019732 337 349 cleft palate SpecificDisease D002972 4019732 351 353 CP SpecificDisease D002972 4019732 359 368 cleft lip SpecificDisease D002971 4019732 385 387 CP SpecificDisease D002972 4019732 389 392 CLP SpecificDisease D002971 4019732 443 451 lip pits SpecificDisease C536528 4019732 455 463 fistulae SpecificDisease D005402 4019732 582 585 CLP SpecificDisease D002971 4019732 590 592 CP SpecificDisease D002972 4019732 603 606 VWS SpecificDisease C536528 4019732 724 727 CLP SpecificDisease D002971 4019732 736 738 CP SpecificDisease D002972 4019732 818 820 CP SpecificDisease D002972 4019732 846 849 CLP SpecificDisease D002971 4019732 854 857 VWS SpecificDisease C536528 4019732 896 904 Lip pits SpecificDisease C536528 4019732 1011 1014 VWS SpecificDisease C536528 4019732 1223 1226 VWS SpecificDisease C536528 4019732 1380 1383 VWS Modifier C536528 4019732 1478 1486 lip pits SpecificDisease C536528 4019732 1518 1526 lip pits SpecificDisease C536528 4019732 1642 1645 VWS SpecificDisease C536528 4019732 1748 1751 VWS Modifier C536528 2491010|t|Molecular and phenotypic analysis of patients with deletions within the deletion-rich region of the Duchenne muscular dystrophy (DMD) gene. 2491010|a|Eighty unrelated individuals with Duchenne muscular dystrophy (DMD)or Becker muscular dystrophy (BMD) were found to have deletions in the major deletion-rich region of the DMD locus. This region includes the last five exons detected by cDNA5b-7, all exons detected by cDNA8, and the first two exons detected by cDNA9. These 80 individuals account for approximately 75% of 109 deletions of the gene, detected among 181 patients analyzed with the entire dystrophin cDNA. Endpoints for many of these deletions were further characterized using two genomic probes, p20 (DXS269; Wapenaar et al.) and GMGX11 (DXS239; present paper). Clinical findings are presented for all 80 patients allowing a correlation of phenotypic severity with the genotype. Thirty-eight independent patients were old enough to be classified as DMD, BMD, or intermediate phenotype and had deletions of exons with sequenced intron/exon boundaries. Of these, eight BMD patients and one intermediate patient had gene deletions predicted to leave the reading frame intact, while 21 DMD patients, 7 intermediate patients, and 1 BMD patient had gene deletions predicted to disrupt the reading frame. Thus, with two exceptions, frameshift deletions of the gene resulted in more severe phenotype than did in-frame deletions. This is in agreement with recent findings by Baumbach et al. and Koenig et al. but is in contrast to findings, by Malhotra et al. at the 5 ' end of the gene. 2491010 100 127 Duchenne muscular dystrophy Modifier D020388 2491010 129 132 DMD Modifier D020388 2491010 174 201 Duchenne muscular dystrophy SpecificDisease D020388 2491010 203 206 DMD SpecificDisease D020388 2491010 210 235 Becker muscular dystrophy SpecificDisease C537666 2491010 237 240 BMD SpecificDisease C537666 2491010 312 315 DMD Modifier D020388 2491010 953 956 DMD SpecificDisease D020388 2491010 958 961 BMD SpecificDisease C537666 2491010 1071 1074 BMD Modifier C537666 2491010 1186 1189 DMD Modifier D020388 2491010 1231 1234 BMD Modifier C537666 10417279|t|Proteolipoprotein gene analysis in 82 patients with sporadic Pelizaeus-Merzbacher Disease: duplications, the major cause of the disease, originate more frequently in male germ cells, but point mutations do not. The Clinical European Network on Brain Dysmyelinating Disease. 10417279|a|Pelizaeus-Merzbacher Disease (PMD) is an X-linked developmental defect of myelination affecting the central nervous system and segregating with the proteolipoprotein (PLP) locus. Investigating 82 strictly selected sporadic cases of PMD, we found PLP mutations in 77%; complete PLP-gene duplications were the most frequent abnormality (62%), whereas point mutations in coding or splice-site regions of the gene were involved less frequently (38%). We analyzed the maternal status of 56 cases to determine the origin of both types of PLP mutation, since this is relevant to genetic counseling. In the 22 point mutations, 68% of mothers were heterozygous for the mutation, a value identical to the two-thirds of carrier mothers that would be expected if there were an equal mutation rate in male and female germ cells. In sharp contrast, among the 34 duplicated cases, 91% of mothers were carriers, a value significantly (chi2 = 9. 20, P <. 01) in favor of a male bias, with an estimation of the male/female mutation frequency (k) of 9. 3 3. Moreover, we observed the occurrence of de novo mutations between parental and grandparental generations in 17 three-generation families, which allowed a direct estimation of the k value (k = 11). Again, a significant male mutation imbalance was observed only for the duplications. 10417279 61 89 Pelizaeus-Merzbacher Disease SpecificDisease OMIM:312080 10417279 244 272 Brain Dysmyelinating Disease DiseaseClass D020279 10417279 274 302 Pelizaeus-Merzbacher Disease SpecificDisease OMIM:312080 10417279 304 307 PMD SpecificDisease D020371 10417279 315 359 X-linked developmental defect of myelination DiseaseClass D020279 10417279 506 509 PMD SpecificDisease D020371 2569949|t|Haplotype analysis of the phenylalanine hydroxylase gene in Turkish phenylketonuria families. 2569949|a|We have estimated the haplotype distribution of mutant and normal phenylalanine hydroxylase (PAH) alleles for 17 Turkish phenylketonuria (PKU) families 20 normal and 27 mutated PAH alleles could be identified. Of the latter, the most prevalent were associated with haplotype 6 (29. 6%), 1 (18. 5%) and 36 (11. 1%), while the normal alleles were preferentially associated with haplotype 1 (20%). Of the 19 different haplotypes observed, 5 have not been described previously. The haplotype distribution differed significantly from that of the Northern European population. Two of the eight polymorphic sites were in association with PKU. No deletions of exon sequences were found in the families analysed. 2569949 68 83 phenylketonuria Modifier D010661 2569949 215 230 phenylketonuria Modifier D010661 2569949 232 235 PKU Modifier D010661 2569949 726 729 PKU SpecificDisease D010661 7668252|t|Cloning of human very-long-chain acyl-coenzyme A dehydrogenase and molecular characterization of its deficiency in two patients. 7668252|a|Two overlapping cDNA clones (1, 991 bp and 736 bp, respectively) encoding the precursor of human mitochondrial very-long-chain acyl-coenzyme A dehydrogenase (VLCAD) were cloned and sequenced. The cDNA inserts of these clones together encompass a region of 2, 177 bases, encoding the entire protein of 655 amino acids, including a 40-amino acid leader peptide and a 615-amino acid mature polypeptide. PCR-amplified VLCAD cDNAs were sequenced in cultured fibroblasts from two VLCAD-deficient patients. In both patients, a 105-bp deletion encompassing bases 1078-1182 in VLCAD cDNA was identified. The deletion seems to occur due to exon skipping during processing of VLCAD pre-mRNA. This is the first demonstration of a mutation causing VLCAD deficiency. Quantitative cDNA expression of normal human VLCAD was performed in the patients fibroblasts, using vaccinia viral system, which demonstrated that the deficiency of the normal VLCAD protein causes impaired long-chain fatty acid beta-oxidation activity in the patients fibroblasts. In patient fibroblasts, raising VLCAD activity to approximately 20% of normal control fibroblast activity raised palmitic acid beta-oxidation flux to the level found in control fibroblasts, which may offer important information for the rational design of future somatic gene therapy for VLCAD deficiency.. 7668252 603 618 VLCAD-deficient Modifier C536353 7668252 864 880 VLCAD deficiency SpecificDisease C536353 7668252 1033 1071 deficiency of the normal VLCAD protein SpecificDisease C536353 7668252 1450 1466 VLCAD deficiency SpecificDisease C536353 2601691|t|Mutations in the RB1 gene and their effects on transcription. 2601691|a|Inactivation of both alleles of the RB1 gene during normal retinal development initiates the formation of a retinoblastoma (RB) tumor. To identify the mutations which inactivate RB1, 21 RB tumors isolated from 19 patients were analyzed with the polymerase chain reaction or an RNase protection assay or both. Mutations were identified in 13 of 21 RB tumors; in 8 tumors, the precise errors in nucleotide sequence were characterized. Each of four germ line mutations involved a small deletion or duplication, while three somatic mutations were point mutations leading to splice alterations and loss of an exon from the mature RB1 mRNA. We were unable to detect expression of the mutant allele in lymphoblasts of three bilaterally affected patients, although the mutation was present in the genomic DNA and transcripts containing the mutations were obvious in the RB tumors in the absence of a normal RB1 allele. The variations in the level of expression of mutant transcripts suggest deregulation of RB1 transcription in the absence of a functional RB1 gene product.. 2601691 170 195 retinoblastoma (RB) tumor SpecificDisease D012175 2601691 248 257 RB tumors SpecificDisease D012175 2601691 409 418 RB tumors SpecificDisease D012175 2601691 425 431 tumors DiseaseClass D009369 2601691 924 933 RB tumors SpecificDisease D012175 1505982|t|Resolution of the two loci for autosomal dominant aniridia, AN1 and AN2, to a single locus on chromosome 11p13. 1505982|a|Two distinct loci have been proposed for aniridia; AN1 for autosomal dominant aniridia on chromosome 2p and AN2 for the aniridia in the WAGR contiguous gene syndrome on chromosome 11p13. In this report, the kindred segregating for autosomal dominant aniridia, which suggested linkage to acid phosphatase-1 (ACP1) and led to the assignment of the AN1 locus on chromosome 2p, has been updated and expanded. Linkage analysis between the aniridia phenotype and ACP1 does not support the original linkage results, excluding linkage up to theta = 0. 17 with Z = -2. Tests for linkage to other chromosome 2p markers. APOB, D2S71, D2S5, and D2S1, also excluded linkage to aniridia. Markers that have been isolated from the chromosome 11p13 region were then analyzed in this aniridia family. Two RFLPs at the D11S323 locus give significant evidence for linkage. The PvuII polymorphism detected by probe p5S1. 6 detects no recombinants, with a maximum lod score of Z = 6. 97 at theta = 0. 00 00. The HaeIII polymorphism detected by the probe p5BE1. 2 gives a maximum lod score of Z = 2. 57 at theta = 0. 00 00. Locus D11S325 gives a lod score of Z = 1. 53 at theta = 0. 00 00. These data suggest that a locus for aniridia (AN1) on chromosome 2p has been misassigned and that this autosomal dominant aniridia family is segregating for an aniridia mutation linked to markers in the 11p13 region. 1505982 50 58 aniridia SpecificDisease D015783 1505982 153 161 aniridia SpecificDisease D015783 1505982 190 198 aniridia SpecificDisease D015783 1505982 232 240 aniridia SpecificDisease D015783 1505982 248 277 WAGR contiguous gene syndrome SpecificDisease D017624 1505982 362 370 aniridia SpecificDisease D015783 1505982 546 554 aniridia SpecificDisease D015783 1505982 776 784 aniridia SpecificDisease D015783 1505982 878 886 aniridia Modifier D015783 1505982 1315 1323 aniridia SpecificDisease D015783 1505982 1401 1409 aniridia Modifier D015783 1505982 1439 1447 aniridia Modifier D015783 3615198|t|GT to AT transition at a splice donor site causes skipping of the preceding exon in phenylketonuria. 3615198|a|Classical Phenylketonuria (PKU) is an autosomal recessive human genetic disorder caused by a deficiency of hepatic phenylalanine hydroxylase (PAH). We isolated several mutant PAH cDNA clones from a PKU carrier individual and showed that they contained an internal 116 base pair deletion, corresponding precisely to exon 12 of the human chromosomal PAH gene. The deletion causes the synthesis of a truncated protein lacking the C-terminal 52 amino acids. Gene transfer and expression studies using the mutant PAH cDNA indicated that the deletion abolishes PAH activity in the cell as a result of protein instability. To determine the molecular basis of the deletion, the mutant chromosomal PAH gene was isolated from this individual and shown to contain a GT-- greater than AT substitution at the 5 splice donor site of intron 12. Thus, the consequence of the splice donor site mutation in the human liver is the skipping of the preceding exon during RNA splicing.. 3615198 84 99 phenylketonuria SpecificDisease D010661 3615198 101 126 Classical Phenylketonuria SpecificDisease D010661 3615198 128 131 PKU SpecificDisease D010661 3615198 139 181 autosomal recessive human genetic disorder DiseaseClass D030342 3615198 194 241 deficiency of hepatic phenylalanine hydroxylase SpecificDisease OMIM:261600 3615198 299 302 PKU Modifier D010661 10521293|t|Clinical and molecular genetic analysis of 19 Wolfram syndrome kindreds demonstrating a wide spectrum of mutations in WFS1. 10521293|a|Wolfram syndrome is an autosomal recessive neurodegenerative disorder characterized by juvenile-onset diabetes mellitus and progressive optic atrophy. mtDNA deletions have been described, and a gene (WFS1) recently has been identified, on chromosome 4p16, encoding a predicted 890 amino acid transmembrane protein. Direct DNA sequencing was done to screen the entire coding region of the WFS1 gene in 30 patients from 19 British kindreds with Wolfram syndrome. DNA was also screened for structural rearrangements (deletions and duplications) and point mutations in mtDNA. No pathogenic mtDNA mutations were found in our cohort. We identified 24 mutations in the WFS1 gene 8 nonsense mutations, 8 missense mutations, 3 in-frame deletions, 1 in-frame insertion, and 4 frameshift mutations. Of these, 23 were novel mutations, and most occurred in exon 8. The majority of patients were compound heterozygotes for two mutations, and there was no common founder mutation. The data were also analyzed for genotype-phenotype relationships. Although some interesting cases were noted, consideration of the small sample size and frequency of each mutation indicated no clear-cut correlations between any of the observed mutations and disease severity. There were no obvious mutation hot spots or clusters. Hence, molecular screening for Wolfram syndrome in affected families and for Wolfram syndrome-carrier status in subjects with psychiatric disorders or diabetes mellitus will require complete analysis of exon 8 and upstream exons.. 10521293 46 62 Wolfram syndrome Modifier OMIM:222300 10521293 124 140 Wolfram syndrome SpecificDisease OMIM:222300 10521293 147 193 autosomal recessive neurodegenerative disorder DiseaseClass D020271 10521293 211 243 juvenile-onset diabetes mellitus SpecificDisease D003922 10521293 260 273 optic atrophy SpecificDisease D009896 10521293 567 583 Wolfram syndrome SpecificDisease OMIM:222300 10521293 1452 1468 Wolfram syndrome SpecificDisease OMIM:222300 10521293 1498 1514 Wolfram syndrome Modifier OMIM:222300 10521293 1547 1568 psychiatric disorders DiseaseClass D001523 10521293 1572 1589 diabetes mellitus SpecificDisease D003920 10528860|t|Maternal uniparental disomy for chromosome 14 in a boy with a normal karyotype. 10528860|a|We report on a boy with a maternal uniparental disomy for chromosome 14 (UPD (14)). At 7 years of age he was referred to us by the paediatrician because of symptoms of Prader-Willi syndrome (PWS). He showed short stature, obesity, mild developmental delay, cryptorchidism, and some mild dysmorphic features. The history further indicated intrauterine growth retardation at the end of the pregnancy. His mother was 44 years of age at the time of his birth. After birth he showed hypotonia with poor sucking, for which gavage feeding was needed. Motor development was delayed. After 1 year he became obese despite a normal appetite. Recurrent middle ear infections, a high pain threshold, and a great skill with jigsaw puzzles were reported. There were no behavioural problems or sleep disturbance. Chromosomal analysis was normal (46, XY). DNA analysis for Prader-Willi syndrome showed no abnormalities. Two years later he was re-examined because we thought his features fitted the PWS-like phenotype associated with maternal UPD (14). At that time precocious puberty was evident. DNA analysis showed maternal heterodisomy for chromosome 14. In all the previously described 11 cases with maternal UPD (14), a Robertsonian translocation involving chromosome 14 was detected cytogenetically before DNA analysis. This is the first report of diagnosis of maternal UPD (14) based on clinical features. This finding underlines the importance of DNA analysis for maternal UPD (14) in patients with a similar PWS-like phenotype even without previous identification of a Robertsonian translocation involving chromosome 14.. 10528860 0 45 Maternal uniparental disomy for chromosome 14 SpecificDisease D024182 10528860 106 151 maternal uniparental disomy for chromosome 14 SpecificDisease D024182 10528860 153 156 UPD SpecificDisease D024182 10528860 248 269 Prader-Willi syndrome SpecificDisease D011218 10528860 271 274 PWS SpecificDisease D011218 10528860 287 300 short stature DiseaseClass D006130 10528860 302 309 obesity SpecificDisease D009765 10528860 316 335 developmental delay DiseaseClass D002658 10528860 337 351 cryptorchidism SpecificDisease D003456 10528860 367 386 dysmorphic features DiseaseClass D000013 10528860 418 449 intrauterine growth retardation DiseaseClass D005317 10528860 558 567 hypotonia DiseaseClass D009123 10528860 678 683 obese Modifier D009765 10528860 721 742 middle ear infections SpecificDisease D010033 10528860 936 957 Prader-Willi syndrome SpecificDisease D011218 10528860 1061 1064 PWS Modifier D011218 10528860 1105 1108 UPD SpecificDisease D024182 10528860 1180 1201 maternal heterodisomy DiseaseClass D024182 10528860 1276 1279 UPD SpecificDisease D024182 10528860 1439 1442 UPD SpecificDisease D024182 10528860 1580 1583 PWS Modifier D011218 7611277|t|Detection of eight BRCA1 mutations in 10 breast/ovarian cancer families, including 1 family with male breast cancer. 7611277|a|Genetic epidemiological evidence suggests that mutations in BRCA1 may be responsible for approximately one half of early onset familial breast cancer and the majority of familial breast/ovarian cancer. The recent cloning of BRCA1 allows for the direct detection of mutations, but the feasibility of presymptomatic screening for cancer susceptibility is unknown. We analyzed genomic DNA from one affected individual from each of 24 families with at least three cases of ovarian or breast cancer, using SSCP assays. Variant SSCP bands were subcloned and sequenced. Allele-specific oligonucleotide hybridization was used to verify sequence changes and to screen DNA from control individuals. Six frameshift and two missense mutations were detected in 10 different families. A frameshift mutation was detected in a male proband affected with both breast and prostate cancer. A 40-bp deletion was detected in a patient who developed intra-abdominal carcinomatosis 1 year after prophylactic oophorectomy. Mutations were detected throughout the gene, and only one was detected in more than a single family. These results provide further evidence that inherited breast and ovarian cancer can occur as a consequence of a wide array of BRCA1 mutations. These results suggests that development of a screening test for BRCA1 mutations will be technically challenging. The finding of a mutation in a family with male breast cancer, not previously thought to be related to BRCA1, also illustrates the potential difficulties of genetic counseling for individuals known to carry mutations.. 7611277 41 62 breast/ovarian cancer Modifier D061325 7611277 97 115 male breast cancer SpecificDisease D018567 7611277 244 266 familial breast cancer SpecificDisease D001943 7611277 287 317 familial breast/ovarian cancer CompositeMention D061325 7611277 445 451 cancer Modifier D009369 7611277 586 610 ovarian or breast cancer CompositeMention D010051|D001943 7611277 960 986 breast and prostate cancer CompositeMention D001943|D011471 7611277 1045 1075 intra-abdominal carcinomatosis DiseaseClass D000008+D002277 7611277 1261 1296 inherited breast and ovarian cancer CompositeMention D061325 7611277 1516 1534 male breast cancer SpecificDisease D018567 2729274|t|Color vision defects in adrenomyeloneuropathy. 2729274|a|The relationship between abnormal color vision and adrenomyeloneuropathy (AMN) was investigated in 27 AMN patients and 31 age-matched controls by using the Farnsworth-Munsell 100 Hue test. Twelve (44%) of 27 patients showed test scores significantly above normal. The axes of bipolarity determined by the testing differed widely between the patients with abnormal scores, compatible with the notion that different alterations in visual pigment genes occur in different AMN kindreds. These observations confirm our earlier impression that the frequency of abnormal color vision is increased in these kindreds, and it supports our contentions that (1) AMN (and its companion, adrenoleukodystrophy) are very closely linked to the visual pigment loci at Xq28 and (2) this proximity might provide the opportunity to observe contiguous gene defects.. 2729274 0 20 Color vision defects DiseaseClass D003117 2729274 24 45 adrenomyeloneuropathy SpecificDisease D000326 2729274 72 93 abnormal color vision DiseaseClass D003117 2729274 98 119 adrenomyeloneuropathy SpecificDisease D000326 2729274 121 124 AMN SpecificDisease D000326 2729274 149 152 AMN Modifier D000326 2729274 516 519 AMN Modifier D000326 2729274 602 623 abnormal color vision DiseaseClass D003117 2729274 697 700 AMN SpecificDisease D000326 2729274 721 741 adrenoleukodystrophy DiseaseClass D000326 2729274 866 889 contiguous gene defects DiseaseClass D025063 3169738|t|Patterns of exon deletions in Duchenne and Becker muscular dystrophy. 3169738|a|A panel of patients with Duchenne and Becker muscular dystrophy (DMD and BMD) has been screened with the cDNA probes Cf56a and Cf23a, which detect exons in the central part of the DMD gene. One or more exons were deleted in 60% of patients. The deletions were mapped and prove to be heterogeneous in size and extent, particularly in DMD. Deletions specific to DMD and to BMD are described. Half of all BMD patients have a deletion of one particular small group of exons; smaller deletions within this same group produce the more severe DMD.. 3169738 30 68 Duchenne and Becker muscular dystrophy CompositeMention D020388|C537666 3169738 95 133 Duchenne and Becker muscular dystrophy CompositeMention D020388|C537666 3169738 135 138 DMD SpecificDisease D020388 3169738 143 146 BMD SpecificDisease C537666 3169738 250 253 DMD Modifier D020388 3169738 403 406 DMD SpecificDisease D020388 3169738 430 433 DMD SpecificDisease D020388 3169738 441 444 BMD SpecificDisease C537666 3169738 472 475 BMD Modifier C537666 3169738 606 609 DMD SpecificDisease D020388 7951327|t|The LEC rat has a deletion in the copper transporting ATPase gene homologous to the Wilson disease gene. 7951327|a|The Long-Evans Cinnamon (LEC) rat shows similarity to Wilson disease in many clinical and biochemical features. We have cloned cDNAs for the rat gene (Atp7b) homologous to the human Wilson disease gene (ATP7B) and have used them to identify a partial deletion in the Atp7b gene in the LEC rat. The deletion removes at least 900 bp of the coding region at the 3 end, includes the crucial ATP binding domain and extends downstream of the gene. Our results provide convincing evidence for defining the LEC rat as an animal model for Wilson disease. This model will be important for studying liver pathophysiology, for developing therapy for Wilson disease and for studying the pathway of copper transport and its possible interaction with other heavy metals.. 7951327 84 98 Wilson disease Modifier D006527 7951327 159 173 Wilson disease SpecificDisease D006527 7951327 287 301 Wilson disease Modifier D006527 7951327 635 649 Wilson disease SpecificDisease D006527 7951327 743 757 Wilson disease SpecificDisease D006527 8440142|t|Detection of a new submicroscopic Norrie disease deletion interval with a novel DNA probe isolated by differential Alu PCR fingerprint cloning. 8440142|a|Differential Alu PCR fingerprint cloning was used to isolate a DNA probe from the Xp11. 4-- > p11. 21 region of the human X chromosome. This novel sequence, cpXr318 (DXS742), detects a new submicroscopic deletion interval at the Norrie disease locus (NDP). Combining our data with the consensus genetic map of the proximal short arm of the X chromosome, we propose the physical order Xcen-DXS14-DXS255- (DXS426, TIMP) - (DXS742- ([MAOB-MAOA-DXS7], NDP) -DXS77-DXS228) -DXS209-DXS148-DXS196- + + + Xpter. The cpXr318 probe and a subclone from a cosmid corresponding to the DXS7 locus were converted into sequence-tagged sites. Finally, DXS742, DSX7, DXS77, and MAOA were integrated into a physical map spanning the Norrie disease locus 8440142 34 48 Norrie disease Modifier C537849 8440142 373 387 Norrie disease Modifier C537849 8440142 858 872 Norrie disease Modifier C537849 1731805|t|Genetic analysis of a Japanese family with normotriglyceridemic abetalipoproteinemia indicates a lack of linkage to the apolipoprotein B gene. 1731805|a|Normotriglyceridemic abetalipoproteinemia is a rare familial disorder characterized by an isolated deficiency of apoB-100. We have previously reported a patient with this disease, who had normal apoB-48 but no apoB-100. To elucidate the genetic abnormalities in this family, we studied the linkage of apoB gene using three genetic markers. The proband and her affected brother showed completely different apoB gene alleles, suggesting that the apoB gene itself is not related to this disorder in this family. By contrast, an American case had a point substitution in the apoB gene generating an in-frame stop codon. These results indicate that this disorder can be caused by defect (s) of either an apoB gene or other genes.. 1731805 43 84 normotriglyceridemic abetalipoproteinemia SpecificDisease D000012 1731805 143 184 Normotriglyceridemic abetalipoproteinemia SpecificDisease D000012 1731805 195 212 familial disorder DiseaseClass D009358 1731805 242 264 deficiency of apoB-100 SpecificDisease OMIM:200100 1731805 380 401 genetic abnormalities DiseaseClass D030342 10788334|t|Founder mutations in the BRCA1 gene in Polish families with breast-ovarian cancer. 10788334|a|We have undertaken a hospital-based study, to identify possible BRCA1 and BRCA2 founder mutations in the Polish population. The study group consisted of 66 Polish families with cancer who have at least three related females affected with breast or ovarian cancer and who had cancer diagnosed, in at least one of the three affected females, at age < 50 years. A total of 26 families had both breast and ovarian cancers, 4 families had ovarian cancers only, and 36 families had breast cancers only. Genomic DNA was prepared from the peripheral blood leukocytes of at least one affected woman from each family. The entire coding region of BRCA1 and BRCA2 was screened for the presence of germline mutations, by use of SSCP followed by direct sequencing of observed variants. Mutations were found in 35 (53%) of the 66 families studied. All but one of the mutations were detected within the BRCA1 gene. BRCA1 abnormalities were identified in all four families with ovarian cancer only, in 67% of 27 families with both breast and ovarian cancer, and in 34% of 35 families with breast cancer only. The single family with a BRCA2 mutation had the breast-ovarian cancer syndrome. Seven distinct mutations were identified; five of these occurred in two or more families. In total, recurrent mutations were found in 33 (94%) of the 35 families with detected mutations. Three BRCA1 abnormalities - 5382insC, C61G, and 4153delA - accounted for 51%, 20%, and 11% of the identified mutations, respectively.. 10788334 60 81 breast-ovarian cancer CompositeMention D061325 10788334 260 266 cancer DiseaseClass D009369 10788334 321 345 breast or ovarian cancer CompositeMention D001943|D010051 10788334 358 364 cancer DiseaseClass D009369 10788334 474 500 breast and ovarian cancers CompositeMention D001943|D010051 10788334 517 532 ovarian cancers SpecificDisease D010051 10788334 559 573 breast cancers SpecificDisease D001943 10788334 982 1001 BRCA1 abnormalities DiseaseClass OMIM:604370 10788334 1044 1058 ovarian cancer SpecificDisease D010051 10788334 1097 1122 breast and ovarian cancer CompositeMention D001943|D010051 10788334 1155 1168 breast cancer SpecificDisease D001943 10788334 1223 1253 breast-ovarian cancer syndrome SpecificDisease D061325 10788334 1448 1467 BRCA1 abnormalities DiseaseClass OMIM:604370 7762560|t|New founder haplotypes at the myotonic dystrophy locus in southern Africa. 7762560|a|The association between normal alleles at the CTG repeat and two nearby polymorphisms in the myotonin protein kinase gene, the Alu insertion/deletion polymorphism and the myotonic dystrophy kinase (DMK) (G/T) intron 9/HinfI polymorphism, has been analyzed in South African Negroids, a population in which myotonic dystrophy (DM) has not been described. South African Negroids have a CTG allelic distribution that is significantly different from that in Caucasoids and Japanese the CTG repeat lengths of > or = 19 are very rare. The striking linkage disequilibrium between specific alleles at the Alu polymorphism (Alu (ins) and Alu (del)), the HinfI polymorphism (HinfI-1 and HinfI-2), and the CTG repeat polymorphism seen in Caucasoid (Europeans and Canadians) populations was also found in the South African Negroid population. Numerous haplotypes, not previously described in Europeans, were, however, found. It thus seems likely that only a small number of these " African " chromosomes were present in the progenitors of all non-African peoples. These data provide support for the " out of Africa " model for the origin of modern humans and suggest that the rare ancestral DM mutation event may have occurred after the migration from Africa, hence the absence of DM in sub-Saharan Negroid peoples.. 7762560 30 48 myotonic dystrophy Modifier D009223 7762560 246 264 myotonic dystrophy Modifier D009223 7762560 380 398 myotonic dystrophy SpecificDisease D009223 7762560 400 402 DM SpecificDisease D009223 7762560 1254 1256 DM Modifier D009223 7762560 1344 1346 DM SpecificDisease D009223 7573040|t|Marked phenotypic heterogeneity associated with expansion of a CAG repeat sequence at the spinocerebellar ataxia 3/Machado-Joseph disease locus. 7573040|a|The spinocerebellar ataxia 3 locus (SCA3) for type I autosomal dominant cerebellar ataxia (ADCA type I), a clinically and genetically heterogeneous group of neurodegenerative disorders, has been mapped to chromosome 14q32. 1 1. ADCA type I patients from families segregating SCA3 share clinical features in common with those with Machado-Joseph disease (MJD), the gene of which maps to the same region. We show here that the disease gene segregating in each of three French ADCA type I kindreds and in a French family with neuropathological findings suggesting the ataxochoreic form of dentatorubropallidoluysian atrophy carries an expanded CAG repeat sequence located at the same locus as that for MJD. Analysis of the mutation in these families shows a strong negative correlation between size of the expanded CAG repeat and age at onset of clinical disease. Instability of the expanded triplet repeat was not found to be affected by sex of the parent transmitting the mutation. Evidence was found for somatic and gonadal mosaicism for alleles carrying expanded trinucleotide repeats. 7573040 90 137 spinocerebellar ataxia 3/Machado-Joseph disease Modifier D017827 7573040 149 173 spinocerebellar ataxia 3 Modifier D017827 7573040 181 185 SCA3 SpecificDisease D017827 7573040 191 234 type I autosomal dominant cerebellar ataxia SpecificDisease OMIM:109150 7573040 236 247 ADCA type I SpecificDisease OMIM:109150 7573040 302 329 neurodegenerative disorders DiseaseClass D019636 7573040 373 384 ADCA type I Modifier OMIM:109150 7573040 420 424 SCA3 SpecificDisease D017827 7573040 475 497 Machado-Joseph disease SpecificDisease D017827 7573040 499 502 MJD SpecificDisease D017827 7573040 619 630 ADCA type I Modifier OMIM:109150 7573040 731 765 dentatorubropallidoluysian atrophy SpecificDisease OMIM:125370 7573040 844 847 MJD SpecificDisease D017827 2309698|t|The red-green visual pigment gene region in adrenoleukodystrophy. 2309698|a|Although recent data established that a specific very-long-chain fatty acyl-CoA synthetase is defective in X-linked adrenoleukodystrophy (ALD), the ALD gene is still unidentified. The ALD locus has been mapped to Xq28, like the red and green color pigment genes. Abnormal color vision has been observed in 12 of 27 patients with adrenomyeloneuropathy (AMN), a milder form of ALD. Furthermore, rearrangements of the color vision gene cluster were found in four of eight ALD kindreds. This led us to propose that a single DNA rearrangement could underlie both ALD and abnormal color vision in these patients. Study of 34 French ALD patients failed to reveal a higher than expected frequency of green/red visual pigment rearrangements 3 to the red/green color vision gene complex. The previous report of such rearrangements was based on small numbers and lack of knowledge that the frequency of " abnormal " color vision arrays on molecular analysis was twice as high as expected on the basis of the frequency of phenotypic color vision defects. The red/green color pigment (R/GCP) region was studied by pulsed-field gel electrophoresis in 14 of these patients, and we did not find any fragment size difference between the patients and normal individuals who have the same number of pigment genes. The R/GCP region was also analyzed in 29 French and seven North American ALD patients by using six genomic DNA probes, isolated from a cosmid walk, that flank the color vision genes. No deletions were found with probes that lie 3 of the green pigment genes. One of the eight previously reported ALD individuals has a long deletion 5 of the red pigment gene, a deletion causing blue cone monochromacy. This finding and the previous findings of a 45% frequency of phenotypic color vision defects in patients with AMN may suggest that the ALD/AMN gene lies 5 to the red pigment gene and that the frequent phenotypic color vision anomalies owe their origin to deleted DNA that includes regulatory genes for color vision. It is possible, however, that phenotypic color vision anomalies in AMN may be phenocopies secondary to retinal or neural involvement by the disease. The single case of blue cone monochromacy may therefore be a fortuitous coincidence of two diseases.. 2309698 44 64 adrenoleukodystrophy SpecificDisease D000326 2309698 173 202 X-linked adrenoleukodystrophy SpecificDisease D000326 2309698 204 207 ALD SpecificDisease D000326 2309698 214 217 ALD Modifier D000326 2309698 250 253 ALD Modifier D000326 2309698 329 350 Abnormal color vision DiseaseClass D003117 2309698 395 416 adrenomyeloneuropathy SpecificDisease D000326 2309698 418 421 AMN SpecificDisease D000326 2309698 441 444 ALD SpecificDisease D000326 2309698 535 538 ALD Modifier D000326 2309698 624 627 ALD SpecificDisease D000326 2309698 632 653 abnormal color vision SpecificDisease D003117 2309698 692 695 ALD Modifier D000326 2309698 1076 1107 phenotypic color vision defects SpecificDisease D003117 2309698 1434 1437 ALD Modifier D000326 2309698 1656 1659 ALD Modifier D000326 2309698 1872 1875 AMN SpecificDisease D000326 2309698 1897 1900 ALD Modifier D000326 2309698 1901 1904 AMN Modifier D000326 2309698 1974 1996 color vision anomalies DiseaseClass D003117 2309698 2119 2141 color vision anomalies DiseaseClass D003117 2309698 2145 2148 AMN SpecificDisease D000326 10662807|t|Haim-Munk syndrome and Papillon-Lefevre syndrome are allelic mutations in cathepsin C. 10662807|a|Of the many palmoplantar keratoderma (PPK) conditions, only Papillon-Lefevre syndrome (PLS) and Haim-Munk syndrome (HMS) are associated with premature periodontal destruction. Although both PLS and HMS share the cardinal features of PPK and severe periodontitis, a number of additional findings are reported in HMS including arachnodactyly, acro-osteolysis, atrophic changes of the nails, and a radiographic deformity of the fingers. While PLS cases have been identified throughout the world, HMS has only been described among descendants of a religious isolate originally from Cochin, India. Parental consanguinity is a characteristic of many cases of both conditions. Although autosomal recessive transmission of PLS is evident, a more " complex " autosomal recessive pattern of inheritance with phenotypic influences from a closely linked modifying locus has been hypothesised for HMS. Recently, mutations of the cathepsin C gene have been identified as the underlying genetic defect in PLS. To determine if a cathepsin C mutation is also responsible for HMS, we sequenced the gene in affected and unaffected subjects from the Cochin isolate in which both the PLS and HMS phenotypes appear. Here we report identification of a mutation of cathepsin C (exon 6, 2127A-- > G) that changes a highly conserved amino acid in the cathepsin C peptide. This mutation segregates with HMS in four nuclear families. Additionally, the existence of a shared common haplotype for genetic loci flanking the cathepsin C gene suggests that affected subjects descended from the Cochin isolate are homozygous for a mutation inherited " identical by descent " from a common ancestor. This finding supports simple autosomal recessive inheritance for HMS in these families. We also report a mutation of the same exon 6 CTSC codon (2126C-- > T) in a Turkish family with classical PLS. These findings provide evidence that PLS and HMS are allelic variants of cathepsin C gene mutations.. 10662807 0 18 Haim-Munk syndrome SpecificDisease C537627 10662807 23 48 Papillon-Lefevre syndrome SpecificDisease D010214 10662807 99 140 palmoplantar keratoderma (PPK) conditions DiseaseClass D007645 10662807 147 172 Papillon-Lefevre syndrome SpecificDisease D010214 10662807 174 177 PLS SpecificDisease D010214 10662807 183 201 Haim-Munk syndrome SpecificDisease C537627 10662807 203 206 HMS SpecificDisease C537627 10662807 277 280 PLS SpecificDisease D010214 10662807 285 288 HMS SpecificDisease C537627 10662807 320 323 PPK DiseaseClass D007645 10662807 335 348 periodontitis SpecificDisease D010518 10662807 398 401 HMS SpecificDisease C537627 10662807 412 426 arachnodactyly SpecificDisease D054119 10662807 428 443 acro-osteolysis SpecificDisease D030981 10662807 445 474 atrophic changes of the nails SpecificDisease D009260 10662807 482 519 radiographic deformity of the fingers SpecificDisease D006226 10662807 527 530 PLS Modifier D010214 10662807 580 583 HMS SpecificDisease C537627 10662807 802 805 PLS SpecificDisease D010214 10662807 971 974 HMS SpecificDisease C537627 10662807 1059 1073 genetic defect DiseaseClass D030342 10662807 1077 1080 PLS SpecificDisease D010214 10662807 1145 1148 HMS SpecificDisease C537627 10662807 1250 1253 PLS Modifier D010214 10662807 1258 1261 HMS Modifier C537627 10662807 1463 1466 HMS SpecificDisease C537627 10662807 1817 1820 HMS SpecificDisease C537627 10662807 1945 1948 PLS SpecificDisease D010214 10662807 1987 1990 PLS SpecificDisease D010214 10662807 1995 1998 HMS SpecificDisease C537627 10377440|t|X inactivation and somatic cell selection rescue female mice carrying a Piga-null mutation. 10377440|a|A somatic mutation in the X linked PIGA gene is responsible for the deficiency of glycosyl phosphatidylinositol (GPI) -anchored proteins on blood cells from patients with paroxysmal nocturnal hemoglobinuria. No inherited form of GPI-anchor deficiency has been described. Because conventional Piga gene knockout is associated with high embryonic lethality in chimeric mice, we used the Cre/loxP system. We generated mice in which two loxP sites flank part of Piga exon 2. After crossbreeding with female mice of the EIIa-cre strain, the floxed allele undergoes Cre-mediated recombination with high efficiency during early embryonic development. Because of X chromosome inactivation, female offspring are mosaic for cells that express or lack GPI-linked proteins. Analysis of mosaic mice showed that in heart, lung, kidney, brain, and liver, mainly wild-type Piga is active, suggesting that these tissues require GPI-linked proteins. The salient exceptions were spleen, thymus, and red blood cells, which had almost equal numbers of cells expressing the wild-type or the recombined allele, implying that GPI-linked proteins are not essential for the derivation of these tissues. PIGA (-) cells had no growth advantage, suggesting that other factors are needed for their clonal dominance in patients with paroxysmal nocturnal hemoglobinuria.. 10377440 160 228 deficiency of glycosyl phosphatidylinositol (GPI) -anchored proteins DiseaseClass C537277 10377440 263 298 paroxysmal nocturnal hemoglobinuria SpecificDisease D006457 10377440 321 342 GPI-anchor deficiency SpecificDisease C537277 10377440 427 446 embryonic lethality SpecificDisease D020964 10377440 1394 1429 paroxysmal nocturnal hemoglobinuria SpecificDisease D006457 1301161|t|The Norrie disease gene maps to a 150 kb region on chromosome Xp11.3. 1301161|a|Norrie disease is a human X-linked recessive disorder of unknown etiology characterized by congenital blindness, sensory neural deafness and mental retardation. This disease gene was previously linked to the DXS7 (L1. 28) locus and the MAO genes in band Xp11. 3 3. We report here fine physical mapping of the obligate region containing the Norrie disease gene (NDP) defined by a recombination and by the smallest submicroscopic chromosomal deletion associated with Norrie disease identified to date. Analysis, using in addition two overlapping YAC clones from this region, allowed orientation of the MAOA and MAOB genes in a 5-3-3-5 configuration. A recombination event between a (GT) n polymorphism in intron 2 of the MAOB gene and the NDP locus, in a family previously reported to have a recombination between DXS7 and NDP, delineates a flanking marker telomeric to this disease gene. An anonymous DNA probe, dc12, present in one of the YACs and in a patient with a submicroscopic deletion which includes MAOA and MAOB but not L1. 28, serves as a flanking marker centromeric to the disease gene. An Alu-PCR fragment from the right arm of the MAO YAC (YMAO. AluR) is not deleted in this patient and also delineates the centromeric extent of the obligate disease region. The apparent order of these loci is telomere. DXS7-MAOA-MAOB-NDP-dc12-YMAO DXS7-MAOA-MAOB-NDP-dc12-YMAO. AluR. centromere. Together these data define the obligate region containing the NDP gene to a chromosomal segment less than 150 kb. 1301161 4 18 Norrie disease Modifier C537849 1301161 70 84 Norrie disease SpecificDisease C537849 1301161 96 123 X-linked recessive disorder DiseaseClass D040181 1301161 161 181 congenital blindness SpecificDisease D057130 1301161 183 206 sensory neural deafness SpecificDisease D006319 1301161 211 229 mental retardation DiseaseClass D008607 1301161 410 424 Norrie disease Modifier C537849 1301161 535 549 Norrie disease SpecificDisease C537849 2393028|t|Molecular genetics of the glucose-6-phosphate dehydrogenase (G6PD) Mediterranean variant and description of a new G6PD mutant, G6PD Andalus1361A. 2393028|a|Glucose-6-phosphate dehydrogenase (G6PD; E. C. 1. 1. 1. 49) deficiency is the most common human enzymopathy; more than 300 different biochemical variants of the enzyme have been described. In many parts of the world the Mediterranean type of G6PD deficiency is prevalent. However, G6PD Mediterranean has come to be regarded as a generic term applied to similar G6PD mutations thought, however, to represent a somewhat heterogeneous group. A C----T mutation at nucleotide 563 of G6PD Mediterranean has been identified by Vulliamy et al., and the same mutation has been found by De Vita et al. in G6PD Mediterranean, G6PD Sassari, and G6PD Cagliari. The latter subjects had an additional mutation, at nucleotide 1311, that did not produce a coding change. We have examined genomic DNA of five patients--four of Spanish origin and one of Jewish origin--having enzymatically documented G6PD Mediterranean. All had both the mutation at nucleotide 563 and that at nucleotide 1311. A sixth sample, resembling G6PD Mediterranean kinetically but with a slightly rapid electrophoretic mobility, was designated G6PD Andalus and was found to have a different mutation, a G----A transition at nucleotide 1361, producing an arginine-to-histidine substitution. These studies suggest that G6PD Mediterranean is, after all, relatively homogeneous. 2393028 146 216 Glucose-6-phosphate dehydrogenase (G6PD; E. C. 1. 1. 1. 49) deficiency SpecificDisease D005955 2393028 242 253 enzymopathy DiseaseClass D008661 2393028 388 403 G6PD deficiency SpecificDisease D005955 1303277|t|Small nuclear ribonucleoprotein polypeptide N (SNRPN), an expressed gene in the Prader-Willi syndrome critical region. 1303277|a|Prader-Willi syndrome (PWS) is associated with paternally derived chromosomal deletions in region 15q11-13 or with maternal disomy for chromosome 15. Therefore, loss of the expressed paternal alleles of maternally imprinted genes must be responsible for the PWS phenotype. We have mapped the gene encoding the small nuclear RNA associated polypeptide SmN (SNRPN) to human chromosome 15q12 and a processed pseudogene SNRPNP1 to chromosome region 6pter-p21. Furthermore, SNRPN was mapped to the minimal deletion interval that is critical for PWS. The fact that the mouse Snrpn gene is maternally imprinted in brain suggests that loss of the paternally derived SNRPN allele may be involved in the PWS phenotype.. 1303277 80 101 Prader-Willi syndrome Modifier D011218 1303277 119 140 Prader-Willi syndrome SpecificDisease D011218 1303277 142 145 PWS SpecificDisease D011218 1303277 234 267 maternal disomy for chromosome 15 SpecificDisease C538037 1303277 377 380 PWS Modifier D011218 1303277 659 662 PWS SpecificDisease D011218 1303277 813 816 PWS Modifier D011218 10064668|t|Increased incidence of cancer in patients with cartilage-hair hypoplasia. 10064668|a|OBJECTIVE Previous reports have suggested an increased risk of cancer among patients with cartilage-hair hypoplasia (CHH). This study was carried out to further evaluate this risk among patients with CHH and their first-degree relatives. STUDY DESIGN One hundred twenty-two patients with CHH were identified through 2 countrywide epidemiologic surveys in 1974 and in 1986. Their parents and nonaffected siblings were identified through the Population Register Center. This cohort underwent follow-up for cancer incidence through the Finnish Cancer Registry to the end of 1995. RESULTS A statistically significant excess risk of cancer was seen among the patients with CHH (standardized incidence ratio 6. 9, 95% confidence interval 2. 3 to 16), which was mainly attributable to non-Hodgkins lymphoma (standardized incidence ratio 90, 95% confidence interval 18 to 264). In addition, a significant excess risk of basal cell carcinoma was seen (standardized incidence ratio 35, 95% confidence interval 7. 2 to 102). The cancer incidence among the siblings or the parents did not differ from the average cancer incidence in the Finnish population. CONCLUSIONS This study confirms an increased risk of cancer, especially non-Hodgkins lymphoma, probably attributable to defective immunity, among patients with CHH. 10064668 23 29 cancer DiseaseClass D009369 10064668 47 72 cartilage-hair hypoplasia SpecificDisease C535916 10064668 138 144 cancer DiseaseClass D009369 10064668 165 190 cartilage-hair hypoplasia SpecificDisease C535916 10064668 192 195 CHH SpecificDisease C535916 10064668 275 278 CHH SpecificDisease C535916 10064668 364 367 CHH SpecificDisease C535916 10064668 580 586 cancer Modifier D009369 10064668 705 711 cancer DiseaseClass D009369 10064668 745 748 CHH SpecificDisease C535916 10064668 855 876 non-Hodgkins lymphoma SpecificDisease D008228 10064668 990 1010 basal cell carcinoma SpecificDisease D002280 10064668 1096 1102 cancer Modifier D009369 10064668 1179 1185 cancer Modifier D009369 10064668 1277 1283 cancer DiseaseClass D009369 10064668 1296 1317 non-Hodgkins lymphoma SpecificDisease D008228 10064668 1384 1387 CHH SpecificDisease C535916 6540752|t|Genetic polymorphism of G6PD in a Bulgarian population. 6540752|a|Considerable genetic heterogeneity in G6PD was found in the Bulgarian population-14 G6PD variants isolated from 117 hemizygous carriers of G6PD deficiency. Of these, G6PD Mediterranean type was a polymorphic variant and G6PD Corinth occurred with high frequency. Two new variants were identified-G6PD Rudosem and G6PD Nedelino. In a selected group of 78 subjects with clinical manifestations, four variants were established G6PD Mediterranian, G6PD Corinth, G6PD Seattle and G6PD Ohut II.. 6540752 195 210 G6PD deficiency SpecificDisease D005955 1349199|t|A genetic etiology for DiGeorge syndrome: consistent deletions and microdeletions of 22q11. 1349199|a|DiGeorge syndrome (DGS), a developmental field defect of the third and fourth pharyngeal pouches, is characterized by aplasia or hypoplasia of the thymus and parathyroid glands and by conotruncal cardiac malformations. Cytogenetic studies support the presence of a DGS critical region in band 22q11. In the present study, we report the results of clinical, cytogenetic, and molecular studies of 14 patients with DGS. Chromosome analysis, utilizing high-resolution banding techniques, detected interstitial deletions in five probands and was inconclusive for a deletion in three probands. The remaining six patients had normal karyotypes. In contrast, molecular analysis detected DNA deletions in all 14 probands. Two of 10 loci tested, D22S75 and D22S259, are deleted in all 14 patients. A third locus, D22S66, is deleted in the eight DGS probands tested. Physical mapping using somatic cell hybrids places D22S66 between D22S75 and D22S259, suggesting that it should be deleted in the remaining six cases. Parent-of-origin studies were performed in five families. Four probands failed to inherit a maternal allele, and one failed to inherit a paternal allele. On the basis of these families, and of six maternally and five paternally derived unbalanced-translocation DGS probands in the literature, parent of origin or imprinting does not appear to play an important role in the pathogenesis of DGS. Deletion of the same three loci in all 14 DGS probands begins to delineate the region of chromosome 22 critical for DGS and confirms the hypothesis that submicroscopic deletions of 22q11 are etiologic in the vast majority of cases.. 1349199 23 40 DiGeorge syndrome SpecificDisease D004062 1349199 92 109 DiGeorge syndrome SpecificDisease D004062 1349199 111 114 DGS SpecificDisease D004062 1349199 210 268 aplasia or hypoplasia of the thymus and parathyroid glands CompositeMention C536288|D007011 1349199 276 309 conotruncal cardiac malformations SpecificDisease C535464 1349199 357 360 DGS Modifier D004062 1349199 504 507 DGS SpecificDisease D004062 1349199 927 930 DGS Modifier D004062 1349199 1360 1363 DGS Modifier D004062 1349199 1488 1491 DGS SpecificDisease D004062 1349199 1535 1538 DGS Modifier D004062 1349199 1609 1612 DGS SpecificDisease D004062 1301146|t|Characterisation of a new rare fragile site easily confused with the fragile X. 1301146|a|A new fragile site (FRAXE) in Xq28 is described. It appears to be a typical folate sensitive fragile site. The fragile site is not associated with mental retardation, it does not give abnormal results when subjected to Southern analysis with probe pfxa3 which detects the unstable DNA sequence characteristic of fragile X syndrome. In situ hybridization mapping locates the fragile site between 150 kb and 600 kb distal to FRAXA. The distinction between the two fragile sites is important clinically since cytogenetic detection of FRAXE, without molecular analysis, could result in misdiagnosis of fragile X syndrome.. 1301146 69 78 fragile X SpecificDisease D005600 1301146 227 245 mental retardation DiseaseClass D008607 1301146 392 410 fragile X syndrome SpecificDisease D005600 1301146 678 696 fragile X syndrome SpecificDisease D005600 7939630|t|BRCA1 mutations in primary breast and ovarian carcinomas. 7939630|a|Loss of heterozygosity data from familial tumors suggest that BRCA1, a gene that confers susceptibility to ovarian and early-onset breast cancer, encodes a tumor suppressor. The BRCA1 region is also subject to allelic loss in sporadic breast and ovarian cancers, an indication that BRCA1 mutations may occur somatically in these tumors. The BRCA1 coding region was examined for mutations in primary breast and ovarian tumors that show allele loss at the BRCA1 locus. Mutations were detected in 3 of 32 breast and 1 of 12 ovarian carcinomas; all four mutations were germline alterations and occurred in early-onset cancers. These results suggest that mutation of BRCA1 may not be critical in the development of the majority of breast and ovarian cancers that arise in the absence of a mutant germline allele.. 7939630 27 56 breast and ovarian carcinomas CompositeMention D001943|D010051 7939630 91 106 familial tumors DiseaseClass D009386 7939630 165 188 ovarian and early-onset CompositeMention D010051 7939630 189 202 breast cancer CompositeMention D001943 7939630 214 219 tumor Modifier D009369 7939630 284 319 sporadic breast and ovarian cancers SpecificDisease D001943|D010051 7939630 387 393 tumors DiseaseClass D009369 7939630 457 482 breast and ovarian tumors CompositeMention D001943|D010051 7939630 579 597 ovarian carcinomas SpecificDisease D010051 7939630 672 679 cancers DiseaseClass D009369 7939630 784 810 breast and ovarian cancers CompositeMention D001943|D010051 8307570|t|Genomic structure of the EWS gene and its relationship to EWSR1, a site of tumor-associated chromosome translocation. 8307570|a|The EWS gene has been identified based on its location at the chromosome 22 breakpoint of the t (11; 22) (q24; q12) translocation that characterizes Ewing sarcoma and related neuroectodermal tumors. The EWS gene spans about 40 kb of DNA and is encoded by 17 exons. The nucleotide sequence of the exons is identical to that of the previously described cDNA. The first 7 exons encode the N-terminal domain of EWS, which consists of a repeated degenerated polypeptide of 7 to 12 residues rich in tyrosine, serine, threonine, glycine, and glutamine. Exons 11, 12, and 13 encode the putative RNA binding domain. The three glycine- and arginine-rich motifs of the gene are mainly encoded by exons 8-9, 14, and 16. The DNA sequence in the 5 region of the gene has features of a CpG-rich island and lacks canonical promoter elements, such as TATA and CCAAT consensus sequences. Positions of the chromosome 22 breakpoints were determined for 19 Ewing tumors. They were localized in introns 7 or 8 in 18 cases and in intron 10 in 1 case.. 8307570 75 80 tumor Modifier D009369 8307570 267 280 Ewing sarcoma SpecificDisease D012512 8307570 293 315 neuroectodermal tumors SpecificDisease D017599 8307570 1054 1066 Ewing tumors DiseaseClass D012512 8364574|t|PAX6 mutations in aniridia. 8364574|a|Aniridia is a congenital malformation of the eye, chiefly characterised by iris hypoplasia, which can cause blindness. The PAX6 gene was isolated as a candidate aniridia gene by positional cloning from the smallest region of overlap of aniridia-associated deletions. Subsequently PAX6 intragenic mutations were demonstrated in Smalleye, a mouse mutant which is an animal model for aniridia, and six human aniridia patients. In this paper we describe four additional PAX6 point mutations in aniridia patients, both sporadic and familial. These mutations highlight regions of the gene which are essential for normal PAX6 function. In addition, the frequency at which we have found PAX6 mutations suggests that lesions in PAX6 will account for most cases of aniridia.. 8364574 18 26 aniridia SpecificDisease D015783 8364574 28 36 Aniridia SpecificDisease D015783 8364574 42 76 congenital malformation of the eye DiseaseClass D005124 8364574 103 118 iris hypoplasia SpecificDisease D007499 8364574 136 145 blindness SpecificDisease D001766 8364574 189 197 aniridia Modifier D015783 8364574 264 272 aniridia Modifier D015783 8364574 409 417 aniridia SpecificDisease D015783 8364574 433 441 aniridia Modifier D015783 8364574 518 526 aniridia Modifier D015783 8364574 783 791 aniridia SpecificDisease D015783 8111379|t|Mutations in the PAX6 gene in patients with hereditary aniridia. 8111379|a|The 14 exons of the PAX6 gene have been analysed exon-by-exon using SSCP in 6 aniridia families. In each family band shifts were observed on the SSCP gels for only one exon and direct PCR-sequencing revealed mutations in each case. Two mutations involved C-- > T transitions in CGAarg codons in exons 9 and 11. Another C-- > T transition converted a CAG-glutamine to a TAG-stop in exon 7. Small insertions created frameshifts which produced downstream stop codons in another two patients and an A-- > T mutation disrupted the splice donor site of exon 5 in the remaining family. Thus, complete inactivation of the PAX6 gene is predicted in all cases. Analysis of other affected members of the families showed that, in each case, all affected individuals carried the same family-specific mutation. One polymorphism was found in exon 7. This data strongly supports the candidature of PAX6 as the gene responsible for hereditary aniridia.. 8111379 55 63 aniridia SpecificDisease D015783 8111379 143 151 aniridia Modifier D015783 8111379 991 999 aniridia SpecificDisease D015783 10735274|t|Locus heterogeneity in Friedreich ataxia. 10735274|a|Friedreich ataxia (FRDA) is the most common form of autosomal recessive ataxia. The disease locus was assigned to chromosome 9 and the disease gene, STM7/X25, has been isolated. To date most data suggest locus homogeneity in FRDA. We now provide strong evidence of a second FRDA locus. Studying two siblings with FRDA from two families we did not detect a mutation in STM7/X25. Haplotype analysis of the STM7/X25 region of chromosome 9 demonstrated that the relevant portion of chromosome 9 differs in the patients. Although the patients studied had typical FRDA, one sibpair had the uncommon symptom of retained tendon reflexes. In order to investigate whether retained tendon reflexes are characteristic of FRDA caused by the second locus, FRDA2, we studied an unrelated FRDA patient with retained tendon reflexes. The observation of typical mutations in STM7/X25 (GAA expansions) in this patient demonstrates that the two genetically different forms of FRDA cannot be distinguished clinically.. 10735274 23 40 Friedreich ataxia SpecificDisease D005621 10735274 42 59 Friedreich ataxia SpecificDisease D005621 10735274 61 65 FRDA SpecificDisease D005621 10735274 94 120 autosomal recessive ataxia DiseaseClass OMIM:610743 10735274 267 271 FRDA SpecificDisease D005621 10735274 316 320 FRDA Modifier D005621 10735274 355 359 FRDA SpecificDisease D005621 10735274 600 604 FRDA SpecificDisease D005621 10735274 751 755 FRDA SpecificDisease D005621 10735274 815 819 FRDA Modifier D005621 10735274 998 1002 FRDA SpecificDisease D005621 10767326|t|Haploinsufficiency of the transcription factors FOXC1 and FOXC2 results in aberrant ocular development. 10767326|a|Anterior segment developmental disorders, including Axenfeld-Rieger anomaly (ARA), variably associate with harmfully elevated intraocular pressure (IOP), which causes glaucoma. Clinically observed dysgenesis does not correlate with IOP, however, and the etiology of glaucoma development is not understood. The forkhead transcription factor genes Foxc1 (formerly Mf1) and Foxc2 (formerly Mfh1) are expressed in the mesenchyme from which the ocular drainage structures derive. Mutations in the human homolog of Foxc1, FKHL7, cause dominant anterior segment defects and glaucoma in various families. We show that Foxc1 (+/-) mice have anterior segment abnormalities similar to those reported in human patients. These abnormalities include small or absent Schlemms canal, aberrantly developed trabecular meshwork, iris hypoplasia, severely eccentric pupils and displaced Schwalbes line. The penetrance of clinically obvious abnormalities varies with genetic background. In some affected eyes, collagen bundles were half normal diameter, or collagen and elastic tissue were very sparse. Thus, abnormalities in extracellular matrix synthesis or organization may contribute to development of the ocular phenotypes. Despite the abnormalities in ocular drainage structures in Foxc1 (+/-) mice, IOP was normal in almost all mice analyzed, on all genetic backgrounds and at all ages. Similar abnormalities were found in Foxc2 (+/-) mice, but no disease-associated mutations were identified in the human homolog FKHL14 in 32 ARA patients. Foxc1 (+/-) and Foxc2 (+/-) mice are useful models for studying anterior segment development and its anomalies, and may allow identification of genes that interact with Foxc1 and Foxc2 (or FKHL7 and FKHL14) to produce a phenotype with elevated IOP and glaucoma.. 10767326 0 63 Haploinsufficiency of the transcription factors FOXC1 and FOXC2 CompositeMention OMIM:602482|OMIM:153400 10767326 104 144 Anterior segment developmental disorders DiseaseClass C537775 10767326 156 179 Axenfeld-Rieger anomaly SpecificDisease C535679 10767326 181 184 ARA SpecificDisease C535679 10767326 271 279 glaucoma SpecificDisease D005901 10767326 370 378 glaucoma Modifier D005901 10767326 642 666 anterior segment defects DiseaseClass C537775 10767326 671 679 glaucoma SpecificDisease D005901 10767326 736 766 anterior segment abnormalities DiseaseClass C537775 10767326 914 929 iris hypoplasia SpecificDisease D007499 10767326 1617 1620 ARA Modifier C535679 10767326 1883 1891 glaucoma SpecificDisease D005901 10446987|t|The dermatofibrosarcoma protuberans-associated collagen type Ialpha1/platelet-derived growth factor (PDGF) B-chain fusion gene generates a transforming protein that is processed to functional PDGF-BB. 10446987|a|Dermatofibrosarcoma protuberans (DFSP) displays chromosomal rearrangements involving chromosome 17 and 22, which fuse the collagen type Ialpha1 (COLIA1) gene to the platelet-derived growth factor (PDGF) B-chain (PDGFB) gene. To characterize the functional and structural properties of the COLIA1/PDGFB fusion protein, we generated a stable NIH3T3 cell line that contained a tumor-derived chimeric gene resulting from a COIA1 intron 7-PDGFB intron 1 fusion. Expression of the fusion protein led to morphological transformation and increased growth rate of these cells. The PDGF receptor kinase inhibitor CGP57148B reversed the transformed phenotype and reduced the growth rate of COLIA1/PDGFB-expressing cells but had no effects on control cells. The presence of dimeric COLIA1/PDGFB precursors was demonstrated through PDGFB immunoprecipitations of metabolically labeled cells and also by PDGFB immunoprecipitations followed by immunoblotting with COLIA1 antibodies. Pulse-chase studies demonstrated that the COLIA1/PDGFB precursor was processed to an end product that was indistinguishable from wild-type PDGF-BB. Finally, COLIA1/PDGFB-expressing cells generated tumors after s. c c. injection into nude mice, and tumor growth was reduced by treatment with CGP57148B. We conclude that the COLIA1/PDGFB fusion associated with DFSP contributes to tumor development through ectopic production of PDGF-BB and the formation of an autocrine loop. Our findings, thus, suggest that PDGF receptors could be a target for pharmacological treatment of DFSP and giant cell fibroblastoma, e. g., through the use of PDGF receptor kinase inhibitors such as CGP57148B. 10446987 4 35 dermatofibrosarcoma protuberans Modifier C538219 10446987 201 232 Dermatofibrosarcoma protuberans SpecificDisease C538219 10446987 234 238 DFSP SpecificDisease C538219 10446987 1365 1371 tumors DiseaseClass D009369 10446987 1416 1421 tumor Modifier D009369 10446987 1527 1531 DFSP SpecificDisease C538219 10446987 1547 1552 tumor Modifier D009369 10446987 1742 1746 DFSP SpecificDisease C538219 10446987 1751 1775 giant cell fibroblastoma SpecificDisease C538219 2006152|t|Founder effect of a prevalent phenylketonuria mutation in the Oriental population. 2006152|a|A missense mutation has been identified in the human phenylalanine hydroxylase [PAH; phenylalanine 4-monooxygenase; L-phenylalanine, tetrahydrobiopterin oxygen oxidoreductase (4-hydroxylating), EC 1. 14. 16. 1] gene in a Chinese patient with classic phenylketonuria (PKU). A G-to-C transition at the second base of codon 413 in exon 12 of the gene results in the substitution of Pro413 for Arg413 in the mutant protein. This mutation (R413P) results in negligible enzymatic activity when expressed in heterologous mammalian cells and is compatible with a classic PKU phenotype in the patient. Population genetic studies reveal that this mutation is tightly linked to restriction fragment length polymorphism haplotype 4, which is the predominant haplotype of the PAH locus in the Oriental population. It accounts for 13. 8% of northern Chinese and 27% of Japanese PKU alleles, but it is rare in southern Chinese (2. 2%) and is absent in the Caucasian population. The data demonstrate unambiguously that the mutation occurred after racial divergence of Orientals and Caucasians and suggest that the allele has spread throughout the Orient by a founder effect. Previous protein polymorphism studies in eastern Asia have led to the hypothesis that " northern Mongoloids " represented a founding population in Asia. Our results are compatible with this hypothesis in that the PKU mutation might have occurred in northern Mongoloids and subsequently spread to the Chinese and Japanese populations. 2006152 30 45 phenylketonuria Modifier D010661 2006152 334 349 phenylketonuria SpecificDisease D010661 2006152 351 354 PKU SpecificDisease D010661 2006152 647 650 PKU Modifier D010661 2006152 948 951 PKU Modifier D010661 2006152 1456 1459 PKU Modifier D010661 10528243|t|Late-onset familial Mediterranean fever (FMF): a subset with distinct clinical, demographic, and molecular genetic characteristics. 10528243|a|To determine the prevalence and characterize demographic, clinical, and genetic features of familial Mediterranean fever (FMF) of late onset, all patients experiencing their first FMF attack at age 40 years or more were identified using the computerized registry of our FMF clinic, and then thoroughly interviewed and examined. The control group consisted of 40 consecutive FMF patients, who arrived at the FMF clinic for their regular follow-up visit and were 40 years of age or older at the time of the examination. The severity of the disease in patients and controls was determined using a modified score, developed previously. Mutational analysis in the FMF gene was performed using a commercial kit. Only 20 of 4000 (0. 5%) patients had late-onset FMF. These patients were mostly men, of non-North African origin, P < 0. 05 compared to controls. All had abdominal attacks and in most these were the only manifestation of their disease, P < 0. 001 001. None had chronic or prolonged manifestations of FMF, for example, amyloidosis, chronic arthritis, or protracted myalgia, P < 0. 001. The response to treatment was good despite using low colchicine dose, P < 0. 05. The overall severity score indicated a mild disease, P < 0. 001. Mutational analysis revealed absence of M694V homozygosity, P < 0. 01, compared to our regular FMF population. We conclude that the onset of FMF in a late age defines a milder form of disease with typical clinical, demographic, and molecular genetic characteristics 10528243 11 39 familial Mediterranean fever SpecificDisease D010505 10528243 41 44 FMF SpecificDisease D010505 10528243 224 252 familial Mediterranean fever SpecificDisease D010505 10528243 254 257 FMF SpecificDisease D010505 10528243 312 315 FMF Modifier D010505 10528243 402 405 FMF Modifier D010505 10528243 506 509 FMF Modifier D010505 10528243 539 542 FMF Modifier D010505 10528243 791 794 FMF Modifier D010505 10528243 886 889 FMF SpecificDisease D010505 10528243 1138 1141 FMF SpecificDisease D010505 10528243 1156 1167 amyloidosis SpecificDisease D000686 10528243 1169 1186 chronic arthritis SpecificDisease D001168 10528243 1202 1209 myalgia SpecificDisease D059352 10528243 1464 1467 FMF Modifier D010505 10528243 1510 1513 FMF SpecificDisease D010505 8195156|t|Structure of the human Na+/glucose cotransporter gene SGLT1. 8195156|a|Intestinal uptake of dietary glucose and galactose is mediated by the SGLT1 Na +/glucose cotransporter of the brush border. An SGLT1 missense mutation underlies hereditary glucose/galactose malabsorption, characterized by potentially fatal diarrhea; conversely, oral rehydration therapy exploits normal transport to alleviate life-threatening diarrhea of infectious origin. We have mapped the entire human SGLT1 Na +/glucose cotransporter gene from cosmid and lambda phage clones representing a genomic region of 112 kilobases. Transcription initiation occurred from a site 27 base pairs 3 of a TATAA sequence. All exon-flanking regions were sequenced, and the entire 112-kilobase region mapped with four restriction enzymes. SGLT1 is comprised of 15 exons (spanning 72 kilobases); a possible evolutionary origin from a six-membrane-span ancestral precursor via a gene duplication event is suggested from comparison of exons against protein secondary structure and from sequence considerations. A new missense mutation in exon 1 causing glucose/galactose malabsorption is also described. This is the first Na (+) -dependent cotransporter gene structure reported. These data facilitate the search for new glucose/galactose malabsorption-related mutations in this important gene and provide a basis for future evolutionary comparisons with other Na (+) -dependent cotransporters.. 8195156 222 264 hereditary glucose/galactose malabsorption SpecificDisease OMIM:606824 8195156 301 309 diarrhea SpecificDisease D003967 8195156 404 412 diarrhea SpecificDisease D003967 8195156 1098 1129 glucose/galactose malabsorption SpecificDisease OMIM:606824 8195156 1265 1296 glucose/galactose malabsorption Modifier OMIM:606824 10766245|t|ATM phosphorylates p95/nbs1 in an S-phase checkpoint pathway. 10766245|a|The rare diseases ataxia-telangiectasia (AT), caused by mutations in the ATM gene, and Nijmegen breakage syndrome (NBS), with mutations in the p95/nbs1 gene, share a variety of phenotypic abnormalities such as chromosomal instability, radiation sensitivity and defects in cell-cycle checkpoints in response to ionizing radiation. The ATM gene encodes a protein kinase that is activated by ionizing radiation or radiomimetic drugs, whereas p95/nbs1 is part of a protein complex that is involved in responses to DNA double-strand breaks. Here, because of the similarities between AT and NBS, we evaluated the functional interactions between ATM and p95/nbs1. Activation of the ATM kinase by ionizing radiation and induction of ATM-dependent responses in NBS cells indicated that p95/nbs1 may not be required for signalling to ATM after ionizing radiation. However, p95/nbs1 was phosphorylated on serine 343 in an ATM-dependent manner in vitro and in vivo after ionizing radiation. A p95/nbs1 construct mutated at the ATM phosphorylation site abrogated an S-phase checkpoint induced by ionizing radiation in normal cells and failed to compensate for this functional deficiency in NBS cells. These observations link ATM and p95/nbs1 in a common signalling pathway and provide an explanation for phenotypic similarities in these two diseases.. 10766245 66 79 rare diseases DiseaseClass D035583 10766245 80 101 ataxia-telangiectasia SpecificDisease D001260 10766245 103 105 AT SpecificDisease D001260 10766245 149 175 Nijmegen breakage syndrome SpecificDisease D049932 10766245 177 180 NBS SpecificDisease D049932 10766245 239 263 phenotypic abnormalities DiseaseClass D004194 10766245 272 295 chromosomal instability DiseaseClass D043171 10766245 640 642 AT SpecificDisease D001260 10766245 647 650 NBS SpecificDisease D049932 10766245 814 817 NBS Modifier D049932 10766245 1239 1242 NBS Modifier D049932 1671881|t|Two distinct mutations at a single BamHI site in phenylketonuria. 1671881|a|Classical phenylketonuria is an autosomal recessive disease caused by a deficiency of hepatic phenylalanine hydroxylase (PAH). The abolition of an invariant BamHI site located in the coding sequence of the PAH gene (exon 7) led to the recognition of two new point mutations at codon 272 and 273 (272gly----stop and 273ser----phe, respectively). Both mutations were detected in north eastern France or Belgium and occurred on the background of RFLP haplotype 7 alleles. The present study supports the view that the clinical heterogeneity in PKU is accounted for by the large variety of mutant genotypes associated with PAH deficiencies.. 1671881 49 64 phenylketonuria SpecificDisease D010661 1671881 66 91 Classical phenylketonuria SpecificDisease D010661 1671881 98 125 autosomal recessive disease DiseaseClass D030342 1671881 138 185 deficiency of hepatic phenylalanine hydroxylase SpecificDisease OMIM:261600 1671881 606 609 PKU SpecificDisease D010661 1671881 684 700 PAH deficiencies SpecificDisease OMIM:261600 10839544|t|Functional link between ataxia-telangiectasia and Nijmegen breakage syndrome gene products. 10839544|a|Ataxia-telangiectasia (A-T) and Nijmegen breakage syndrome (NBS) are recessive genetic disorders with susceptibility to cancer and similar cellular phenotypes. The protein product of the gene responsible for A-T, designated ATM, is a member of a family of kinases characterized by a carboxy-terminal phosphatidylinositol 3-kinase-like domain. The NBS1 protein is specifically mutated in patients with Nijmegen breakage syndrome and forms a complex with the DNA repair proteins Rad50 and Mrel1. Here we show that phosphorylation of NBS1, induced by ionizing radiation, requires catalytically active ATM. Complexes containing ATM and NBS1 exist in vivo in both untreated cells and cells treated with ionizing radiation. We have identified two residues of NBS1, Ser 278 and Ser 343 that are phosphorylated in vitro by ATM and whose modification in vivo is essential for the cellular response to DNA damage. This response includes S-phase checkpoint activation, formation of the NBS1/Mrel1/Rad50 nuclear foci and rescue of hypersensitivity to ionizing radiation. Together, these results demonstrate a biochemical link between cell-cycle checkpoints activated by DNA damage and DNA repair in two genetic diseases with overlapping phenotypes.. 10839544 24 45 ataxia-telangiectasia Modifier D001260 10839544 50 76 Nijmegen breakage syndrome Modifier D049932 10839544 92 113 Ataxia-telangiectasia SpecificDisease D001260 10839544 115 118 A-T SpecificDisease D001260 10839544 124 150 Nijmegen breakage syndrome SpecificDisease D049932 10839544 152 155 NBS SpecificDisease D049932 10839544 161 188 recessive genetic disorders DiseaseClass D030342 10839544 212 218 cancer DiseaseClass D009369 10839544 300 303 A-T SpecificDisease D001260 10839544 493 519 Nijmegen breakage syndrome SpecificDisease D049932 10839544 1111 1149 hypersensitivity to ionizing radiation DiseaseClass D004194 10839544 1283 1299 genetic diseases DiseaseClass D030342 8499920|t|Structure and genomic sequence of the myotonic dystrophy (DM kinase) gene. 8499920|a|The mutation causing myotonic dystrophy (DM) has recently been identified as an unstable CTG trinucleotide repeat located in the 3 untranslated region of a gene encoding for a protein with putative serine-threonine protein kinase activity. In this report we present the genomic sequences of the human and murine DM kinase gene. A comparison of these sequences with each other and with known cDNA sequences from both species, led us to predict a translation initiation codon, as well as determine the organization of the DM kinase gene. Several polymorphisms within the human DM kinase gene have been identified, and PCR assays to detect two of these are described. The complete sequence and characterization of the structure of the DM kinase gene, as well as the identification of novel polymorphisms within the gene, represent an important step in a further understanding of the genetics of myotonic dystrophy and the molecular biology of the gene.. 8499920 38 56 myotonic dystrophy Modifier D009223 8499920 58 60 DM Modifier D009223 8499920 96 114 myotonic dystrophy SpecificDisease D009223 8499920 116 118 DM SpecificDisease D009223 8499920 387 389 DM Modifier D009223 8499920 595 597 DM Modifier D009223 8499920 650 652 DM Modifier D009223 8499920 807 809 DM Modifier D009223 8499920 967 985 myotonic dystrophy SpecificDisease D009223 10712201|t|Identification of novel imprinted transcripts in the Prader-Willi syndrome and Angelman syndrome deletion region: further evidence for regional imprinting control. 10712201|a|Deletions and other abnormalities of human chromosome 15q11-q13 are associated with two developmental disorders, Prader-Willi syndrome (PWS) and Angelman syndrome (AS). Loss of expression of imprinted, paternally expressed genes has been implicated in PWS. However, the number of imprinted genes that contribute to PWS, and the range over which the imprinting signal acts to silence one copy of the gene in a parent-of-origin-specific manner, are unknown. To identify additional imprinted genes that could contribute to the PWS phenotype and to understand the regional control of imprinting in 15q11-q13, we have constructed an imprinted transcript map of the PWS-AS deletion interval. The imprinting status of 22 expressed sequence tags derived from the radiation-hybrid human transcript maps or physical maps was determined in a reverse transcriptase-PCR assay and correlated with the position of the transcripts on the physical map. Seven new paternally expressed transcripts localize to an approximately 1. 5-Mb domain surrounding the SNRPN-associated imprinting center, which already includes four imprinted, paternally expressed genes. All other tested new transcripts in the deletion region were expressed from both alleles. A domain of exclusive paternal expression surrounding the imprinting center suggests strong regional control of the imprinting process. This study provides the means for further investigation of additional genes that cause or modify the phenotypes associated with rearrangements of 15q11-q13. 10712201 53 74 Prader-Willi syndrome Modifier D011218 10712201 79 96 Angelman syndrome Modifier D017204 10712201 277 298 Prader-Willi syndrome SpecificDisease D011218 10712201 300 303 PWS SpecificDisease D011218 10712201 309 326 Angelman syndrome SpecificDisease D017204 10712201 328 330 AS SpecificDisease D017204 10712201 416 419 PWS SpecificDisease D011218 10712201 479 482 PWS SpecificDisease D011218 10712201 688 691 PWS Modifier D011218 10441573|t|Penetrances of BRCA1 1675delA and 1135insA with respect to breast cancer and ovarian cancer. 10441573|a|For genetic counseling and predictive testing in families with inherited breast-ovarian cancer, penetrances and expressions of the underlying mutations should be known. We have previously reported two BRCA1 founder mutations in the Norwegian population. Index cases for the present study were found two different ways through a series of consecutive ovarian cancers (n = 16) and through our family cancer clinic (n = 14). Altogether, 20 of the patients had BRCA1 1675delA, and 10 had 1135insA. Their relatives were described with respect to absence/presence of breast and/or ovarian cancer. Of 133 living female relatives, 83 (62%) were tested for the presence of a mutation. No difference, in penetrance and expression, between the two mutations were found, whereas differences according to method of ascertainment were seen. The overall findings were that disease started to occur at age 30 years and that by age 50 years 48% of the mutation-carrying women had experienced breast and/or ovarian cancer. More ovarian cancers than breast cancers were recorded. Both penetrance and expression (breast cancer vs. ovarian cancer) were different from those in reports of the Ashkenazi founder mutations. Whether the reported differences reflect true differences and/or methodological problems is discussed. An observed excess of mutation carriers could not be accounted for by methodological problems; possible explanations were a " true " low penetrance or preferential segregation.. 10441573 59 72 breast cancer SpecificDisease D001943 10441573 77 91 ovarian cancer SpecificDisease D010051 10441573 156 187 inherited breast-ovarian cancer CompositeMention D061325 10441573 444 459 ovarian cancers SpecificDisease D010051 10441573 492 498 cancer Modifier D009369 10441573 655 683 breast and/or ovarian cancer CompositeMention D001943|D010051 10441573 1069 1097 breast and/or ovarian cancer CompositeMention D001943|D010051 10441573 1104 1119 ovarian cancers SpecificDisease D010051 10441573 1125 1139 breast cancers SpecificDisease D001943 10441573 1187 1200 breast cancer SpecificDisease D001943 10441573 1205 1219 ovarian cancer SpecificDisease D010051 7535801|t|Molecular basis of subtotal complement C6 deficiency. A carboxy-terminally truncated but functionally active C6. 7535801|a|Individuals with subtotal complement C6 deficiency possess a C6 molecule that is 14% shorter than normal C6 and present in low but detectable concentrations (1-2% of the normal mean). We now show that this dysmorphic C6 is bactericidally active and lacks an epitope that was mapped to the most carboxy-terminal part of C6 using C6 cDNA fragments expressed as fusion proteins in the pUEX expression system. We thus predicted that the abnormal C6 molecule might be carboxy-terminally truncated and sought a mutation in an area approximately 14% from the carboxy-terminal end of the coding sequence. By sequencing PCR-amplified products from this region, we found, in three individuals from two families, a mutation that might plausibly be responsible for the defect. All three have an abnormal 5 splice donor site of intron 15, which would probably prevent splicing. An in-frame stop codon is found 17 codons downstream from the intron boundary, which would lead to a truncated polypeptide 13. 5% smaller than normal C6. This result was unexpected, as earlier studies mapped the C5b binding site, or a putative enzymatic region, to this part of C6. Interestingly, all three subjects were probably heterozygous for both subtotal C6 and complete C6 deficiency. 7535801 19 52 subtotal complement C6 deficiency SpecificDisease OMIM:612446 7535801 130 163 subtotal complement C6 deficiency SpecificDisease OMIM:612446 7535801 1330 1368 subtotal C6 and complete C6 deficiency CompositeMention OMIM:612446 10408776|t|Mutations of the VHL gene in sporadic renal cell carcinoma: definition of a risk factor for VHL patients to develop an RCC. 10408776|a|To investigate the nature of somatic von Hippel-Lindau (VHL) mutations, we analyzed 173 primary sporadic human renal cell carcinomas for mutations of the VHL tumor suppressor gene, using polymerase chain reaction (PCR) and single-strand conformational polymorphism analysis (SSCP) of DNA. We detected abnormal SSCP pattern in 73 samples. After sequencing, we identified microdeletions in 58% of cases, microinsertions in 17%, nonsense mutations in 8%, and missense mutations in 17%. Among these mutations, 50% correspond to new mutations. VHL mutations were found only in the nonpapillary renal cell carcinoma (RCC) subtype, as previously reported. To compare somatic and germline mutations, we used the VHL database, which includes 507 mutations. The study of mutational events revealed a significant difference between somatic and germline mutations with mutations leading to truncated proteins observed in 78% of somatic mutations vs only 37% in germline mutations (P < 0. 001). We postulated that a specific pattern of VHL mutations is associated with sporadic RCC. This pattern corresponds to mutations leading mainly to truncated proteins with few specific missense mutations. We then analyzed the occurrence of RCC in VHL families, based on the nature of mutations. We observed RCC in at least one member of the VHL families in 77% of cases with mutations leading to truncated proteins versus 55% in cases with missense mutations (P < 0. 05). Thus, mutations resulting in truncated proteins may lead to a higher risk of RCC in VHL patients 10408776 17 20 VHL Modifier D006623 10408776 29 58 sporadic renal cell carcinoma SpecificDisease D002292 10408776 92 95 VHL Modifier D006623 10408776 119 122 RCC SpecificDisease D002292 10408776 161 178 von Hippel-Lindau Modifier D006623 10408776 180 183 VHL Modifier D006623 10408776 235 256 renal cell carcinomas SpecificDisease D002292 10408776 278 287 VHL tumor Modifier D006623 10408776 663 666 VHL Modifier D006623 10408776 700 733 nonpapillary renal cell carcinoma Modifier D002292 10408776 735 738 RCC Modifier D002292 10408776 828 831 VHL Modifier D006623 10408776 1147 1150 VHL Modifier D006623 10408776 1180 1192 sporadic RCC SpecificDisease D002292 10408776 1342 1345 RCC SpecificDisease D002292 10408776 1349 1352 VHL Modifier D006623 10408776 1409 1412 RCC SpecificDisease D002292 10408776 1443 1446 VHL Modifier D006623 10408776 1651 1654 RCC SpecificDisease D002292 10408776 1658 1661 VHL Modifier D006623 8528200|t|Evidence for inter-generational instability in the CAG repeat in the MJD1 gene and for conserved haplotypes at flanking markers amongst Japanese and Caucasian subjects with Machado-Joseph disease. 8528200|a|The size of the (CAG)n repeat array in the 3' end of the MJD1 gene and the haplotype at a series of microsatellite markers surrounding the MJD1 gene were examined in a large cohort of Japanese and Caucasian subjects affected with Machado-Joseph disease (MJD). Our data provide five novel observations. First, MJD is associated with expansion fo the array from the normal range of 14-37 repeats to 68-84 repeats in most Japanese and Caucasian subjects, but no subjects were observed with expansions intermediate in size between those of the normal and MJD affected groups. Second, the expanded allele associated with MJD displays inter-generational instability, particularly in male meioses, and this instability was associated with the clinical phenomenon of anticipation. Third, the size of the expanded allele is not only inversely correlated with the age-of-onset of MJD (r = -0.738, p < 0.001), but is also correlated with the frequency of other clinical features [e.g. pseudoexophthalmos and pyramidal signs were more frequent in subjects with large repeats (p < 0.001 and p < 0.05 respectively)]. Fourth, the disease phenotype is significantly more severe and had an early age of onset (16 years) in a subject homozygous for the expanded allele, which contrasts with Huntington disease and suggests that the expanded allele in the MJD1 gene could exert its effect either by a dominant negative effect (putatively excluded in HD) or by a gain of function effect as proposed for HD. Finally, Japanese and Caucasian subjects affected with MJD share haplotypes at several markers surrounding the MJD1 gene, which are uncommon in the normal Japanese and Caucasian population, and which suggests the existence either of common founders in these populations or of chromosomes susceptible to pathologic expansion of the CAG repeat in the MJD1 gene. 8528200 173 195 Machado-Joseph disease SpecificDisease D017827 8528200 427 449 Machado-Joseph disease SpecificDisease D017827 8528200 451 454 MJD SpecificDisease D017827 8528200 506 509 MJD SpecificDisease D017827 8528200 748 751 MJD Modifier D017827 8528200 813 816 MJD SpecificDisease D017827 8528200 1067 1070 MJD SpecificDisease D017827 8528200 1470 1488 Huntington disease SpecificDisease D006816 8528200 1628 1630 HD SpecificDisease D006816 8528200 1680 1682 HD SpecificDisease D006816 8528200 1739 1742 MJD SpecificDisease D017827 10911990|t|Myotonic dystrophy: the role of the CUG triplet repeats in splicing of a novel DMPK exon and altered cytoplasmic DMPK mRNA isoform ratios. 10911990|a|The mechanism by which (CTG) n expansion in the 3 UTR of the DMPK gene causes myotonic dystrophy (DM) is unknown. We identified four RNA splicing factors--hnRNP C, U2AF (U2 auxiliary factor), PTB (polypyrimidine tract binding protein), and PSF (PTB associated splicing factor) --that bind to two short regions 3 of the (CUG) n, and found a novel 3 DMPK exon resulting in an mRNA lacking the repeats. We propose that the (CUG) n is an essential cis acting element for this splicing event. In contrast to (CUG) n containing mRNAs, the novel isoform is not retained in the nucleus in DM cells, resulting in imbalances in relative levels of cytoplasmic DMPK mRNA isoforms and a new dominant effect of the mutation on DMPK.. 10911990 0 18 Myotonic dystrophy SpecificDisease D009223 10911990 217 235 myotonic dystrophy SpecificDisease D009223 10911990 237 239 DM SpecificDisease D009223 10911990 720 722 DM Modifier D009223 10077651|t|Mechanism of increased iron absorption in murine model of hereditary hemochromatosis : increased duodenal expression of the iron transporter DMT1. 10077651|a|Hereditary hemochromatosis (HH) is a common autosomal recessive disorder characterized by tissue iron deposition secondary to excessive dietary iron absorption. We recently reported that HFE, the protein defective in HH, was physically associated with the transferrin receptor (TfR) in duodenal crypt cells and proposed that mutations in HFE attenuate the uptake of transferrin-bound iron from plasma by duodenal crypt cells, leading to up-regulation of transporters for dietary iron. Here, we tested the hypothesis that HFE-/- mice have increased duodenal expression of the divalent metal transporter (DMT1). By 4 weeks of age, the HFE-/- mice demonstrated iron loading when compared with HFE +/+ littermates, with elevated transferrin saturations (68. 4% vs. 49. 8%) and elevated liver iron concentrations (985 micrograms vs. 381 micrograms). By using Northern blot analyses, we quantitated duodenal expression of both classes of DMT1 transcripts one containing an iron responsive element (IRE), called DMT1 (IRE), and one containing no IRE, called DMT1 (non-IRE). The positive control for DMT1 up-regulation was a murine model of dietary iron deficiency that demonstrated greatly increased levels of duodenal DMT1 (IRE) mRNA. HFE-/- mice also demonstrated an increase in duodenal DMT1 (IRE) mRNA (average 7. 7-fold), despite their elevated transferrin saturation and hepatic iron content. Duodenal expression of DMT1 (non-IRE) was not increased, nor was hepatic expression of DMT1 increased. These data support the model for HH in which HFE mutations lead to inappropriately low crypt cell iron, with resultant stabilization of DMT1 (IRE) mRNA, up-regulation of DMT1, and increased absorption of dietary iron. 10077651 58 84 hereditary hemochromatosis SpecificDisease D006432 10077651 147 173 Hereditary hemochromatosis SpecificDisease D006432 10077651 175 177 HH SpecificDisease D006432 10077651 191 219 autosomal recessive disorder DiseaseClass D030342 10077651 364 366 HH SpecificDisease D006432 10077651 1281 1304 dietary iron deficiency SpecificDisease D018798 1325652|t|Yeast artificial chromosomes for the molecular analysis of the familial polyposis APC gene region. 1325652|a|Two yeast artificial chromosomes (YACs) spanning a total distance of 1. 1 megabase pairs of DNA around the MCC (for mutated in colorectal carcinoma) and APC (for adenomatous polyposis coli) genes at 5q21 have been isolated and characterized. Starting from the MCC gene, a strategy was undertaken to identify constitutional submicroscopic deletions in familial adenomatous polyposis patients that might considerably narrow down the position of the APC gene. To this end, YACs identified by the MCC gene were screened across a chromosome 5-specific cosmid library to provide a source of DNA probes for genomic scanning. The cosmids isolated from these experiments were used to screen a panel of somatic cell hybrids containing chromosome 5 segregated from patients suspected to carry putative interstitial deletions. This screening approach led to the confirmation of a small heterozygous deletion in a polyposis patient that overlaps one of the two isolated YACs. This YAC has been shown to contain the entire APC gene, in addition to a significant portion of DNA flanking the 5 end of the gene, and should therefore prove a valuable resource for functional studies by transfer to colorectal tumor-derived cell lines. 1325652 63 85 familial polyposis APC Modifier D011125 1325652 226 246 colorectal carcinoma SpecificDisease D015179 1325652 252 255 APC Modifier D011125 1325652 261 287 adenomatous polyposis coli Modifier D011125 1325652 450 480 familial adenomatous polyposis Modifier D011125 1325652 546 549 APC Modifier D011125 1325652 1000 1009 polyposis Modifier D011125 1325652 1108 1111 APC Modifier D011125 1325652 1279 1295 colorectal tumor Modifier D015179 1269174|t|Analbuminemia in an American Indian girl. 1269174|a|Analbuminemia was fortuitously detected in a nonedematous 12-year-old American Indian girl with atopic dermatitis, mild bronchial asthma, a mild seizure disorder, and hyperlipoproteinemia with a corneal arcus. Immunologic methods revealed trace amounts (17 mg/100 ml) of apparently normal serum albumin. The patients parents were remotely related. The pedigree and clinical findings were compatible with autosomal recessive transmission of analbuminemia. Heterozygotes had subnormal levels of serum albumin. The Gc-locus is closely linked to the structural albumin locus. Gc-protein levels were normal in the patient and together with normal chromosomal banding studies make it unlikely that a chromosomal deletion caused analbuminemia. Gc-types in the family were compatible with, but did not prove, linkage of analbuminemia to the Gc-locus. These findings suggest a " thalassemia " -like mutation for this disorder.. 1269174 0 13 Analbuminemia SpecificDisease OMIM:103600 1269174 42 55 Analbuminemia SpecificDisease OMIM:103600 1269174 138 155 atopic dermatitis SpecificDisease D003876 1269174 162 178 bronchial asthma SpecificDisease D001249 1269174 187 203 seizure disorder SpecificDisease D004827 1269174 209 229 hyperlipoproteinemia SpecificDisease D006951 1269174 237 250 corneal arcus SpecificDisease D001112 1269174 482 495 analbuminemia SpecificDisease OMIM:103600 1269174 764 777 analbuminemia SpecificDisease OMIM:103600 1269174 854 867 analbuminemia SpecificDisease OMIM:103600 1269174 912 923 thalassemia Modifier D013789 3524231|t|Treatment of Duchenne muscular dystrophy with growth hormone inhibitors. 3524231|a|A controlled, double-blind therapeutic trial with the drug mazindol, a growth hormone inhibitor, was performed in a pair of 7 1/2 year-old monozygotic twins, with Duchenne muscular dystrophy (DMD). The rationale for this trial was based on a patient (reported previously) affected simultaneously with DMD and growth hormone (GH) deficiency, who is showing a benign course of the dystrophic process and is still walking at 18 years. One of the twins received 2 mg of mazindol daily, while the other received a placebo. The assessment, repeated every 2 months, included weight and height measurements, functional and motor ability tests, ergometry and determinations of serum enzymes and GH levels. After one year of trial the code was broken and it was seen that the twin under placebo treatment was strikingly worse than his brother, the progression of whose condition was practically arrested. These results strongly suggest that treatment with a GH inhibitor is beneficial for DMD patients.. 3524231 13 40 Duchenne muscular dystrophy SpecificDisease D020388 3524231 236 263 Duchenne muscular dystrophy SpecificDisease D020388 3524231 265 268 DMD SpecificDisease D020388 3524231 374 377 DMD SpecificDisease D020388 3524231 382 412 growth hormone (GH) deficiency SpecificDisease OMIM:262400 3524231 452 470 dystrophic process DiseaseClass D009136 3524231 1052 1055 DMD Modifier D020388 8528198|t|Identification of WASP mutations in patients with Wiskott-Aldrich syndrome and isolated thrombocytopenia reveals allelic heterogeneity at the WAS locus. 8528198|a|Mutation in the gene encoding the recently isolated WASP protein has now been identified as the genetic defect responsible for the X-linked Wiskott-Aldrich syndrome (WAS), a primary immunodeficiency disease associated with extensive phenotypic variability. To elucidate the range of WASP mutations responsible for WAS, we used PCR-SSCP analysis to screen for WASP gene mutation in 19 unrelated boys with the diagnosis of classical or attenuated WAS or isolated thrombocytopenia. All 19 patients had WASP mutations, each of which localized to the initial three or terminal three exons of the gene, and the majority of which were unique in each case. However, a missense mutation which results in substitution of the arginine at WAS codon 86 was identified in three boys with severe WAS as well as in one boy presenting with thrombocytopenia alone. While the three mutations found in the isolated thrombocytopenia patients leave the reading frame intact, about one-half of the gene alterations detected in both severe and attenuated WAS patients result in frameshifted transcript and premature translation termination. These findings therefore confirm the association of WAS with WASP mutation and identify WASP mutation as a cause for isolated congenital thrombocytopenia in males. While the WASP gene defects responsible for isolated thrombocytopenia and other mild presentations of WAS do not appear distinct from those resulting in severe WAS, these data indicate that analysis of WASP gene mutation provides a valuable tool for distinguishing the spectrum of WAS patients and the subset of males with isolated thrombocytopenia who represent mild cases of WAS.. 8528198 50 74 Wiskott-Aldrich syndrome SpecificDisease D014923 8528198 79 104 isolated thrombocytopenia SpecificDisease OMIM:313900 8528198 142 145 WAS Modifier D014923 8528198 249 263 genetic defect DiseaseClass D030342 8528198 284 317 X-linked Wiskott-Aldrich syndrome SpecificDisease D014923 8528198 319 322 WAS SpecificDisease D014923 8528198 335 359 immunodeficiency disease DiseaseClass D007153 8528198 467 470 WAS SpecificDisease D014923 8528198 598 601 WAS SpecificDisease D014923 8528198 605 630 isolated thrombocytopenia SpecificDisease OMIM:313900 8528198 880 883 WAS Modifier D014923 8528198 934 937 WAS SpecificDisease D014923 8528198 976 992 thrombocytopenia SpecificDisease D013921 8528198 1039 1064 isolated thrombocytopenia Modifier OMIM:313900 8528198 1184 1187 WAS SpecificDisease D014923 8528198 1322 1325 WAS SpecificDisease D014923 8528198 1396 1423 congenital thrombocytopenia SpecificDisease OMIM:313900 8528198 1478 1503 isolated thrombocytopenia SpecificDisease OMIM:313900 8528198 1536 1539 WAS SpecificDisease D014923 8528198 1594 1597 WAS SpecificDisease D014923 8528198 1715 1718 WAS Modifier D014923 8528198 1757 1782 isolated thrombocytopenia SpecificDisease OMIM:313900 8528198 1811 1814 WAS SpecificDisease D014923 8178825|t|Huntington disease without CAG expansion: phenocopies or errors in assignment? 8178825|a|Huntington disease (HD) has been shown to be associated with an expanded CAG repeat within a novel gene on 4p16. 3 (IT15). A total of 30 of 1, 022 affected persons (2. 9% of our cohort) did not have an expanded CAG in the disease range. The reasons for not observing expansion in affected individuals are important for determining the sensitivity of using repeat length both for diagnosis of affected patients and for predictive testing programs and may have biological relevance for the understanding of the molecular mechanism underlying HD. Here we show that the majority (18) of the individuals with normal sized alleles represent misdiagnosis, sample mix-up, or clerical error. The remaining 12 patients represent possible phenocopies for HD. In at least four cases, family studies of these phenocopies excluded 4p16. 3 as the region responsible for the phenotype. Mutations in the HD gene that are other than CAG expansion have not been excluded for the remaining eight cases; however, in as many as seven of these persons, retrospective review of these patients clinical features identified characteristics not typical for HD. This study shows that on rare occasions mutations in other, as-yet-undefined genes can present with a clinical phenotype very similar to that of HD 8178825 0 18 Huntington disease SpecificDisease D006816 8178825 79 97 Huntington disease SpecificDisease D006816 8178825 99 101 HD SpecificDisease D006816 8178825 619 621 HD SpecificDisease D006816 8178825 823 825 HD SpecificDisease D006816 8178825 966 968 HD Modifier D006816 8178825 1209 1211 HD SpecificDisease D006816 8178825 1358 1360 HD SpecificDisease D006816 6101415|t|Allelic exclusion of glucose-6-phosphate dehydrogenase in platelets and T lymphocytes from a Wiskott-Aldrich syndrome carrier. 6101415|a|An obligate carrier of the Wiskott-Aldrich syndrome (WAS) who was also heterozygous for the A and B types of X-linked glucose-6-phosphate dehydrogenase was found. With her it became possible to determine whether allelic exclusion occurs in particular cell-types of the WAS carrier. If so, the remaining cells of a particular cell-type would express only the normal X chromosome and only one glucose-6-phosphate dehydrogenase isoenzyme would be demonstrable. This carrier had only the B isoenzyme of glucose-6-phosphate dehydrogenase in platelets and thymus-derived T lymphocytes, although both isoenzymes A and B were present in erythrocytes and neutrophils. These findings suggest that selection against the WAS gene occurs in platelets and thymus-derived T lymphocytes and that the defects associated with WAS expressed in these cell-types may be implicated in the genesis of the Wiskott-Aldrich syndrome.. 6101415 93 117 Wiskott-Aldrich syndrome Modifier D014923 6101415 154 178 Wiskott-Aldrich syndrome SpecificDisease D014923 6101415 180 183 WAS SpecificDisease D014923 6101415 396 399 WAS Modifier D014923 6101415 836 839 WAS Modifier D014923 6101415 935 938 WAS SpecificDisease D014923 6101415 1009 1033 Wiskott-Aldrich syndrome SpecificDisease D014923 10487695|t|A novel mutation in the sodium/iodide symporter gene in the largest family with iodide transport defect. 10487695|a|We previously reported nine children with an autosomally recessive form of congenital hypothyroidism due to an iodide transport defect in a large Hutterite family with extensive consanguinity living in central Canada. Since the original report, we have diagnosed congenital hypothyroidism by newborn TSH screening in 9 additional children from the family. We performed direct sequencing of the PCR products of each NIS (sodium/iodide symporter) gene exon with flanking introns amplified from genomic DNA extracted from peripheral blood cells of the patients. We identified a novel NIS gene mutation, G395R (Gly395-- > Arg; GGA-- > AGA), in 10 patients examined in the present study. All of the parents tested were heterozygous for the mutation, suggesting that the patients were homozygous. The mutation was located in the 10th transmembrane helix. Expression experiments by transfection of the mutant NIS complimentary DNA into COS-7 cells showed no perchlorate-sensitive iodide uptake, confirming that the mutation is the direct cause of the iodide transport defect in these patients. A patient who showed an intermediate saliva/serum technetium ratio (14. 0; normal, > or = 20) and was considered to have a partial or less severe defect in the previous report (IX-24) did not have a NIS gene mutation. It is now possible to use gene diagnostics of this unique NIS mutation to identify patients with congenital hypothyroidism due to an iodide transport defect in this family and to determine the carrier state of potential parents for genetic counseling and arranging rapid and early diagnosis of their infants. 10487695 80 103 iodide transport defect DiseaseClass OMIM:274400 10487695 180 205 congenital hypothyroidism SpecificDisease D003409 10487695 216 239 iodide transport defect DiseaseClass OMIM:274400 10487695 368 393 congenital hypothyroidism SpecificDisease D003409 10487695 1149 1172 iodide transport defect DiseaseClass OMIM:274400 10487695 1507 1532 congenital hypothyroidism SpecificDisease D003409 10487695 1543 1566 iodide transport defect DiseaseClass OMIM:274400 2071157|t|Determination of the mutations responsible for the Lesch-Nyhan syndrome in 17 subjects. 2071157|a|Hypoxanthine--guanine phosphoribosyltransferase (HPRT) is a purine salvage enzyme that catalyzes the conversion of hypoxanthine to inosine monophosphate and guanine to guanosine monophosphate. Previous studies of mutant HPRT proteins analyzed at the molecular level have shown a significant heterogeneity. This investigation further verifies this heterogeneity and identifies insertions, deletions, and point mutations. The direct sequencing of the polymerase chain reaction-amplified product of reverse-transcribed HPRT mRNA enabled the rapid identification of the mutations found in 17 previously uncharacterized cell lines derived from patients with the Lesch-Nyhan syndrome.. 2071157 51 71 Lesch-Nyhan syndrome SpecificDisease D007926 2071157 745 765 Lesch-Nyhan syndrome SpecificDisease D007926 7444053|t|A new CT pattern in adrenoleukodystrophy. 7444053|a|A new CT pattern was observed in 2 patients with adrenoleukodystrophy (ALD). This pattern, which the authors call Type II, is characterized by the absence of posterior periventricular areas of decreased attenuation around the trigone on non-contrast scans after contrast infusion, however, there is striking enhancement of various white-matter structures (tracts or fiber systems) such as the internal capsules, corpus callosum, corona radiata, forceps major, and cerebral peduncles. This is different from numerous previous descriptions of the CT pattern in ALD. Type II ALD does not appear to have been seen in any other leukoencephalopathy and is probably specific for a phenotypic variant or an evolving stage of ALD.. 7444053 20 40 adrenoleukodystrophy SpecificDisease D000326 7444053 91 111 adrenoleukodystrophy SpecificDisease D000326 7444053 113 116 ALD SpecificDisease D000326 7444053 602 605 ALD SpecificDisease D000326 7444053 607 618 Type II ALD SpecificDisease D000326 7444053 666 685 leukoencephalopathy DiseaseClass D056784 7444053 760 763 ALD SpecificDisease D000326 10706858|t|Genetic analysis, phenotypic diagnosis, and risk of venous thrombosis in families with inherited deficiencies of protein S. 10706858|a|Protein S deficiency is a recognized risk factor for venous thrombosis. Of all the inherited thrombophilic conditions, it remains the most difficult to diagnose because of phenotypic variability, which can lead to inconclusive results. We have overcome this problem by studying a cohort of patients from a single center where the diagnosis was confirmed at the genetic level. Twenty-eight index patients with protein S deficiency and a PROS1 gene defect were studied, together with 109 first-degree relatives. To avoid selection bias, we confined analysis of total and free protein S levels and thrombotic risk to the patients relatives. In this group of relatives, a low free protein S level was the most reliable predictor of a PROS1 gene defect (sensitivity 97. 7%, specificity 100%). First-degree relatives with a PROS1 gene defect had a 5. 0-fold higher risk of thrombosis (95% confidence interval, 1. 5-16. 8) than those with a normal PROS1 gene and no other recognized thrombophilic defect. Although pregnancy/puerperium and immobility/trauma were important precipitating factors for thrombosis, almost half of the events were spontaneous. Relatives with splice-site or major structural defects in the PROS1 gene were more likely to have had a thrombotic event and had significantly lower total and free protein S levels than those relatives having missense mutations. We conclude that persons with PROS1 gene defects and protein S deficiency are at increased risk of thrombosis and that free protein S estimation offers the most reliable way of diagnosing the deficiency. (Blood. 2000; 95 1935-1941). 10706858 52 69 venous thrombosis SpecificDisease D020246 10706858 97 122 deficiencies of protein S SpecificDisease D018455 10706858 124 144 Protein S deficiency SpecificDisease D018455 10706858 177 194 venous thrombosis SpecificDisease D020246 10706858 217 241 thrombophilic conditions DiseaseClass D019851 10706858 533 553 protein S deficiency SpecificDisease D018455 10706858 560 577 PROS1 gene defect SpecificDisease OMIM:612336 10706858 854 871 PROS1 gene defect SpecificDisease OMIM:612336 10706858 942 959 PROS1 gene defect SpecificDisease OMIM:612336 10706858 991 1001 thrombosis SpecificDisease D013927 10706858 1100 1120 thrombophilic defect SpecificDisease D019851 10706858 1167 1173 trauma SpecificDisease D014947 10706858 1215 1225 thrombosis SpecificDisease D013927 10706858 1318 1343 defects in the PROS1 gene SpecificDisease OMIM:612336 10706858 1530 1548 PROS1 gene defects SpecificDisease OMIM:612336 10706858 1553 1573 protein S deficiency SpecificDisease D018455 10706858 1599 1609 thrombosis SpecificDisease D013927 10382910|t|Severe clinical expression in X-linked Emery-Dreifuss muscular dystrophy. 10382910|a|X-linked Emery-Dreifuss muscular dystrophy (EDMD) is a relatively rare benign neuromuscular disorder which can vary remarkably in onset, course and severity. In the present study, a TCTAC deletion spanning the nucleotides 631-635 of the emerin gene caused an unusually severe disease phenotype including loss of ambulation and severe muscle wasting in two affected brothers. The same mutation has been reported previously in an unrelated family showing a significantly milder phenotype. The interfamilial heterogeneity in distribution and in severity of the features in the two families point to environmental or genetic modification as the cause of clinical variability in Emery-Dreifuss muscular dystrophy.. 10382910 30 72 X-linked Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10382910 74 116 X-linked Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10382910 118 122 EDMD SpecificDisease D020389 10382910 145 174 benign neuromuscular disorder DiseaseClass D009468 10382910 378 396 loss of ambulation DiseaseClass D051346 10382910 408 422 muscle wasting DiseaseClass D009133 10382910 748 781 Emery-Dreifuss muscular dystrophy SpecificDisease D020389 7825578|t|The gene for spinal cerebellar ataxia 3 (SCA3) is located in a region of approximately 3 cM on chromosome 14q24.3-q32.2. 7825578|a|SCA3, the gene for spinal cerebellar ataxia 3, was recently mapped to a 15-cM interval between D14S67 and D14S81 on chromosome 14q, by linkage analysis in two families of French ancestry. The SCA3 candidate region has now been refined by linkage analysis with four new microsatellite markers (D14S256, D14S291, D14S280, and AFM343vf1) in the same two families, in which 19 additional individuals were genotyped, and in a third French family. Combined two-point linkage analyses show that the new markers, D14S280 and AFM343vf1, are tightly linked to the SCA3 locus, with maximal lod scores, at recombination fraction, (theta) =. 00, of 7. 05 and 13. 70, respectively. Combined multipoint and recombinant haplotype analyses localize the SCA3 locus to a 3-cM interval flanked by D14S291 and D14S81. The same allele for D14S280 segregates with the disease locus in the three kindreds. This allele is frequent in the French population, however, and linkage disequilibrium is not clearly established. The SCA3 locus remains within the 29-cM region on 14q24. 3-q32. 2 containing the gene for the Machado-Joseph disease, which is clinically related to the phenotype determined by SCA3, but it cannot yet be concluded that both diseases result from alterations of the same gene 7825578 13 37 spinal cerebellar ataxia Modifier D013132 7825578 140 164 spinal cerebellar ataxia Modifier D013132 7825578 1211 1233 Machado-Joseph disease SpecificDisease D017827 1831007|t|Huntington disease and childhood-onset Tourette syndrome. 1831007|a|A 40-year-old man with childhood-onset Tourette syndrome (TS) developed Huntington disease (HD). We believe this to be the first reported case of childhood-onset TS with adult onset HD. Discovery of other cases with both disorders may provide clues to the pathophysiology of both conditions.. 1831007 0 18 Huntington disease SpecificDisease D006816 1831007 39 56 Tourette syndrome SpecificDisease D005879 1831007 97 114 Tourette syndrome SpecificDisease D005879 1831007 116 118 TS SpecificDisease D005879 1831007 130 148 Huntington disease SpecificDisease D006816 1831007 150 152 HD SpecificDisease D006816 1831007 220 222 TS SpecificDisease D005879 1831007 240 242 HD SpecificDisease D006816 7761412|t|The protein deficient in Lowe syndrome is a phosphatidylinositol-4,5-bisphosphate 5-phosphatase. 7761412|a|Lowe syndrome, also known as oculocerebrorenal syndrome, is caused by mutations in the X chromosome-encoded OCRL gene. The OCRL protein is 51% identical to inositol polyphosphate 5-phosphatase II (5-phosphatase II) from human platelets over a span of 744 aa, suggesting that OCRL may be a similar enzyme. We engineered a construct of the OCRL cDNA that encodes amino acids homologous to the platelet 5-phosphatase for expression in baculovirus-infected Sf9 insect cells. This cDNA encodes aa 264-968 of the OCRL protein. The recombinant protein was found to catalyze the reactions also carried out by platelet 5-phosphatase II. Thus OCRL converts inositol 1, 4, 5-trisphosphate to inositol 1, 4-bisphosphate, and it converts inositol 1, 3, 4, 5-tetrakisphosphate to inositol 1, 3, 4-trisphosphate. Most important, the enzyme converts phosphatidylinositol 4, 5-bisphosphate to phosphatidylinositol 4-phosphate. The relative ability of OCRL to catalyze the three reactions is different from that of 5-phosphatase II and from that of another 5-phosphatase isoenzyme from platelets, 5-phosphatase I. The recombinant OCRL protein hydrolyzes the phospholipid substrate 10- to 30-fold better than 5-phosphatase II, and 5-phosphatase I does not cleave the lipid at all. We also show that OCRL functions as a phosphatidylinositol 4, 5-bisphosphate 5-phosphatase in OCRL-expressing Sf9 cells. These results suggest that OCRL is mainly a lipid phosphatase that may control cellular levels of a critical metabolite, phosphatidylinositol 4, 5-bisphosphate. Deficiency of this enzyme apparently causes the protean manifestations of Lowe syndrome.. 7761412 25 38 Lowe syndrome SpecificDisease D009800 7761412 97 110 Lowe syndrome SpecificDisease D009800 7761412 126 152 oculocerebrorenal syndrome SpecificDisease D009800 7761412 1715 1728 Lowe syndrome SpecificDisease D009800 10323740|t|Microdeletions at chromosome bands 1q32-q41 as a cause of Van der Woude syndrome. 10323740|a|Van der Woude syndrome (VWS) is an autosomal dominant disorder comprising cleft lip and/or cleft palate and lip pits. We reported previously a family whose underlying mutation is a 500-800 kb deletion localized to chromosome bands 1q32-q41 [Sander et al., 1994 Hum Mol Genet 3 576-578]. Along with cleft lip/palate and lip pits, affected relatives exhibit developmental delays, suggesting that the function of a gene nearby may also be disrupted. To further localize the VWS gene we searched for other deletions that cause VWS. An allele loss assay was performed using a novel highly polymorphic marker, D1S3753. From a panel of 37 unrelated individuals, we detected an allele loss in one family, indicating the presence of a deletion. In this family, the phenotype in three generations of affected individuals was confined to the cardinal signs of VWS. Surprisingly, mapping of the new deletion showed that it extended 0. 2-1 Mb beyond the proximal breakpoint for the deletion described previously. No deletions were detected in seven cases of popliteal pterygia syndrome, 76 cases of mixed syndromic forms of cleft lip and palate, and 178 cases of nonsyndromic cleft lip and palate. 10323740 58 80 Van der Woude syndrome SpecificDisease C536528 10323740 82 104 Van der Woude syndrome SpecificDisease C536528 10323740 106 109 VWS SpecificDisease C536528 10323740 117 144 autosomal dominant disorder DiseaseClass D030342 10323740 156 165 cleft lip SpecificDisease D002971 10323740 173 185 cleft palate SpecificDisease D002972 10323740 190 198 lip pits SpecificDisease C536528 10323740 382 398 cleft lip/palate CompositeMention D002971|D002972 10323740 403 411 lip pits SpecificDisease C536528 10323740 440 460 developmental delays DiseaseClass D006130 10323740 555 558 VWS Modifier C536528 10323740 607 610 VWS SpecificDisease C536528 10323740 933 936 VWS SpecificDisease C536528 10323740 1129 1156 popliteal pterygia syndrome SpecificDisease OMIM:119500 10323740 1176 1215 syndromic forms of cleft lip and palate CompositeMention D002971|D002972 10323740 1234 1267 nonsyndromic cleft lip and palate CompositeMention OMIM:119530 7607677|t|Mucopolysaccharidosis type IVA: common double deletion in the N-acetylgalactosamine-6-sulfatase gene (GALNS). 7607677|a|Mucopolysaccharidosis IVA (MPS IVA) is an autosomal recessive disorder caused by a deficiency in N-acetylgalactosamine-6-sulfatase (GALNS). We found two separate deletions of nearly 8. 0 and 6. 0 kb in the GALNS gene, including some exons. There are Alu repetitive elements near the breakpoints of the 8. 0-kb deletion, and this deletion resulted from an Alu-Alu recombination. The other 6. 0-kb deletion involved illegitimate recombinational events between incomplete short direct repeats of 8 bp at deletion breakpoints. The same rearrangement has been observed in a heteroallelic state in four unrelated patients. This is the first documentation of a common double deletion a gene that is not a member of a gene cluster. 7607677 0 30 Mucopolysaccharidosis type IVA SpecificDisease OMIM:253000 7607677 110 135 Mucopolysaccharidosis IVA SpecificDisease OMIM:253000 7607677 137 144 MPS IVA SpecificDisease OMIM:253000 7607677 152 180 autosomal recessive disorder DiseaseClass D030342 7607677 193 240 deficiency in N-acetylgalactosamine-6-sulfatase SpecificDisease D009085 2995231|t|Segregation analysis of a marker localised Xp21.2-Xp21.3 in Duchenne and Becker muscular dystrophy families. 2995231|a|A DNA marker C7, localised Xp21. 1-Xp21. 3, has been studied in kindreds segregating for Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). In DMD families four crossovers were observed in 38 informative meioses between C7 and the DMD locus (theta = 0. 12, z max = + 2. 72). In BMD families no recombinants were observed in the 16 informative meioses studied. These data are consistent with the localisation of the mutations in these disorders being in the same region of Xp21. Studies in families also segregating for the DNA marker 754 support the previously reported physical order of these loci as X centromere-754-DMD-BMD-C7-X telomere. A recombination fraction of 0. 11 (z max = + 5. 58) was found between DMD-754 by combining our previously published data with the data presented here. C7 and 754 thus provide good bridging markers for the diagnosis of DMD and BMD 2995231 60 98 Duchenne and Becker muscular dystrophy CompositeMention D020388|C537666 2995231 198 225 Duchenne muscular dystrophy SpecificDisease D020388 2995231 227 230 DMD SpecificDisease D020388 2995231 236 261 Becker muscular dystrophy SpecificDisease C537666 2995231 263 266 BMD SpecificDisease C537666 2995231 272 275 DMD Modifier D020388 2995231 360 363 DMD Modifier D020388 2995231 407 410 BMD Modifier C537666 2995231 989 992 DMD SpecificDisease D020388 2995231 997 1000 BMD SpecificDisease C537666 8281142|t|Genetic analysis of the BRCA1 region in a large breast/ovarian family: refinement of the minimal region containing BRCA1. 8281142|a|We have analyzed a single multi-affected breast/ovarian cancer pedigree (BOV3) and have shown consistent inheritance of markers on chromosome 17q with the disease confirming that this family is due to the BRCA1 gene. Analysis of 17q haplotypes shows a recombination event in a bilateral breast cancer case which suggests that the BRCA1 gene lies distal to D17S857; D17S857 is thus the new proximal boundary for the region containing BRCA1. Combining this information with previously published mapping information suggests that BRCA1 is contained in a region estimated at 1-1. 5 Mb in length. All seven breast tumour/blood pairs examined from this family show loss of heterozygosity in the tumours. The allel retained in each tumour was from the disease-bearing chromosome implicating BRCA1 as a tumour suppressor gene. We have sequenced the 17 beta-oestradiol dehydrogenase genes (EDH17B1 and EDH17B2) which have been suggested as candidate genes for BRCA1 in four members of this family. No germline mutations were detected. 8281142 163 184 breast/ovarian cancer Modifier D061325 8281142 409 422 breast cancer Modifier D001943 8281142 724 737 breast tumour Modifier D001943 8281142 811 818 tumours DiseaseClass D009369 8281142 847 853 tumour DiseaseClass D009369 8281142 917 923 tumour Modifier D009369 10590055|t|A point mutation Thr(799)Met on the alpha(2) integrin leads to the formation of new human platelet alloantigen Sit(a) and affects collagen-induced aggregation. 10590055|a|A new platelet-specific alloantigen, termed Sit (a), was identified in a severe case of neonatal alloimmune thrombocytopenia. The Sit (a) alloantigen is of low frequency (1/400) in the German population. Immunochemical studies demonstrated that the Sit (a) epitopes reside on platelet glycoprotein (GP) Ia. Nucleotide sequence analysis of GPIa cDNA derived from Sit (a) -positive platelets showed C (2531) -- > T (2531) point mutation, resulting in Thr (799) Met dimorphism. Analysis of genomic DNA from 22 Sit (a) -negative normal individuals showed that the Thr (799) is encoded by ACG (2532) (90. 9%) or ACA (2532) (9. 1%). To establish a DNA typing technique, we elucidated the organization of the GPIa gene adjacent to the polymorphic bases. The introns (421 bp and 1. 2 kb) encompass a 142-bp exon with the 2 polymorphic bases 2531 and 2532. Polymerase chain reaction-restriction fragment length polymorphism analysis on DNA derived from 100 donors using the restriction enzyme Mae III showed that the Met (799) form of GPIa is restricted to Sit (a) (+) phenotype. Analysis of stable Chinese hamster ovary transfectants expressing allele-specific recombinant forms of GPIa showed that anti-Sit (a) exclusively reacted with the Glu (505) Met (799), but not with the Glu (505) Thr (799) and the Lys (505) Thr (799) isoforms. In contrast, anti-Br (a) (HPA-5b) only recognized the Lys (505) Thr (799) form, whereas anti-Br (b) (HPA-5a) reacted with both Glu (505) Thr (799) and Glu (505) Met (799) isoforms. These results demonstrated that the Met (799) is responsible for formation of the Sit (a) alloantigenic determinants, whereas amino acid 505 (Lys or Glu) specifically controls the expression of Br (a) and Br (b) epitopes, respectively. Platelet aggregation responses of Sit (a) (+) individuals were diminished in response to collagen, indicating that the Thr (799) Met mutation affects the function of the GPIa/IIa complex 10590055 248 284 neonatal alloimmune thrombocytopenia SpecificDisease D054098 3464560|t|Estimation of the male to female ratio of mutation rates from the segregation of X-chromosomal DNA haplotypes in Duchenne muscular dystrophy families. 3464560|a|A novel procedure is presented to estimate the ratio of male to female mutation rates for Duchenne muscular dystrophy (DMD). X-specific restriction fragment length polymorphisms are used to establish DNA haplotypes in three-generation DMD families. From the proportion of DMD patients who have inherited their maternal grandfathers X chromosome, the ratio of mutation rates can be calculated. In contrast to classical methods, the proposed procedure is not restricted to sporadic or familiar cases nor is any information on the carrier status of female relatives required.. 3464560 113 140 Duchenne muscular dystrophy Modifier D020388 3464560 241 268 Duchenne muscular dystrophy SpecificDisease D020388 3464560 270 273 DMD SpecificDisease D020388 3464560 386 389 DMD Modifier D020388 3464560 423 426 DMD Modifier D020388 3789016|t|Genetic analysis of an inherited deficiency of the third component of complement in Brittany spaniel dogs. 3789016|a|Genetically determined C3 deficiency in Brittany spaniel dogs shares a number of biochemical and clinical characteristics with the human disorder. In humans, the gene for C3 deficiency is a null gene that is allelic to the structural gene for C3 and is not linked to the major histocompatibility locus. The current study used allotype analysis of canine C3 in order to demonstrate that the gene for C3 deficiency in these dogs is also a null gene allelic to the structural gene for C3. In addition, preliminary pedigree analysis suggests that the gene for canine C3 deficiency is apparently not closely linked to the major histocompatibility complex of the dog. Thus, it appears that C3 deficiency in Brittany spaniel dogs not only shares biochemical and clinical features with C3 deficiency in humans, but also shares some genetic characteristics with the human disorder.. 3789016 33 80 deficiency of the third component of complement SpecificDisease OMIM:613779 3789016 130 143 C3 deficiency SpecificDisease OMIM:613779 3789016 278 291 C3 deficiency SpecificDisease OMIM:613779 3789016 506 519 C3 deficiency SpecificDisease OMIM:613779 3789016 670 683 C3 deficiency SpecificDisease OMIM:613779 3789016 791 804 C3 deficiency SpecificDisease OMIM:613779 3789016 885 898 C3 deficiency SpecificDisease OMIM:613779 10426139|t|A novel frameshift mutation in the McLeod syndrome gene in a Japanese family. 10426139|a|We report a novel mutation in the XK gene (XK) in a Japanese patient with McLeod syndrome. A 50-year-old man showed progressive muscular atrophy, choreic movement, elevated level of serum creatinine kinase, and acanthocytosis. The expression level of all the Kell antigens in erythrocyte was decreased and molecular analysis revealed a single-base (T) deletion at the nucleotide position 1095 in XK. This deletion caused a frameshift in translation, leading to a premature stop codon at the amino acid position 408. We conclude this single-base deletion causes defective Kx protein, which is responsible for the McLeod phenotype in this patient.. 10426139 35 50 McLeod syndrome Modifier OMIM:300842 10426139 152 167 McLeod syndrome SpecificDisease OMIM:300842 10426139 206 222 muscular atrophy DiseaseClass D009133 10426139 224 240 choreic movement DiseaseClass D002819 10426139 289 303 acanthocytosis SpecificDisease D000012 10426139 690 696 McLeod Modifier OMIM:300842 10330430|t|Alpha-cardiac actin is a novel disease gene in familial hypertrophic cardiomyopathy. 10330430|a|We identified the alpha-cardiac actin gene (ACTC) as a novel disease gene in a pedigree suffering from familial hypertrophic cardiomyopathy (FHC). Linkage analyses excluded all the previously reported FHC loci as possible disease loci in the family studied, with lod scores varying between -2. 5 and -6. 0 0. Further linkage analyses of plausible candidate genes highly expressed in the adult human heart identified ACTC as the most likely disease gene, showing a maximal lod score of 3. 6 6. Mutation analysis of ACTC revealed an Ala295Ser mutation in exon 5 close to 2 missense mutations recently described to cause the inherited form of idiopathic dilated cardiomyopathy (IDC). 10330430 47 83 familial hypertrophic cardiomyopathy SpecificDisease D024741 10330430 188 224 familial hypertrophic cardiomyopathy SpecificDisease D024741 10330430 226 229 FHC SpecificDisease D024741 10330430 286 289 FHC Modifier D024741 10330430 725 758 idiopathic dilated cardiomyopathy SpecificDisease C536277 10330430 760 763 IDC SpecificDisease C536277 8084618|t|The EWS gene, involved in Ewing family of tumors, malignant melanoma of soft parts and desmoplastic small round cell tumors, codes for an RNA binding protein with novel regulatory domains. 8084618|a|The EWS gene, which maps to band q12 of human chromosome 22, is involved in a wide variety of human solid tumors including Ewing sarcoma, related primitive neuroectodermal tumors, malignant melanoma of soft parts and desmoplastic small round cell tumors. In these tumors, the EWS is fused to genes encoding transcriptional activators/repressors, like Fli-1 or erg or ATF 1 or wt1. To better understand the function of the EWS protein, we cloned the EWS cDNA. Sequence analysis of this cDNA revealed differential splicing involving two exons encoding 72 amino acids. Both alternatively spliced transcripts, EWS and EWS-b, are expressed in a variety of cells. Because EWS proteins contain putative conserved RNA binding motifs, we studied the RNA binding properties of the EWS protein. The EWS-b protein binds to RNA in vitro and, specifically, to poly G and poly U. The RNA binding activity was localized to the carboxy terminal 86 amino acids, which constitute RGG box. Thus the amino terminal domain of EWS (NTD-EWS), which is involved in chromosome translocation may regulate the specificity of RNA binding activity of EWS. An EWS-erg chimeric protein, which is found in Ewings sarcoma cells, functions as a transcriptional activator. Mutational analysis of EWS-erg chimeric protein revealed that NTD-EWS functions as a regulatory domain for the transcriptional activation properties of EWS-erg chimeric protein.. 8084618 26 48 Ewing family of tumors DiseaseClass D012512 8084618 50 82 malignant melanoma of soft parts DiseaseClass D008545 8084618 87 123 desmoplastic small round cell tumors SpecificDisease D058405 8084618 289 301 solid tumors DiseaseClass D009369 8084618 312 325 Ewing sarcoma SpecificDisease D012512 8084618 345 367 neuroectodermal tumors DiseaseClass D017599 8084618 369 401 malignant melanoma of soft parts DiseaseClass D008545 8084618 406 442 desmoplastic small round cell tumors DiseaseClass D058405 8084618 453 459 tumors DiseaseClass D009369 8084618 1362 1376 Ewings sarcoma Modifier D012512 1301938|t|A mutation common in non-Jewish Tay-Sachs disease: frequency and RNA studies. 1301938|a|Tay-Sachs disease (TSD) is an autosomal recessive genetic disorder resulting from mutation of the HEXA gene encoding the alpha-subunit of the lysosomal enzyme, beta-N-acetylhexosaminidase A (Hex A). We have discovered that a Tay-Sachs mutation, IVS-9 + 1 G-- > A, first detected by Akli et al. (Genomics 11 124-134, 1991), is a common disease allele in non-Jewish Caucasians (10/58 alleles examined). A PCR-based diagnostic test, which detects an NlaIII site generated by the mutation, revealed a frequency among enzyme-defined carriers of 9/64 (14%). Most of those carrying the allele trace their origins to the United Kingdom, Ireland, or Western Europe. It was not identified among 12 Black American TSD alleles or in any of 18 Ashkenazi Jewish, enzyme-defined carriers who did not carry any of the mutations common to this population. No normally spliced RNA was detected in PCR products generated from reverse transcription of RNA carrying the IVS-9 mutation. Instead, the low levels of mRNA from this allele were comprised of aberrant species resulting from the use of either of two cryptic donor sites, one truncating exon 9 and the other within IVS-9, spliced to exon 10. Numerous additional splice products were detected, most involving skipping of one or more surrounding exons. Together with a recently identified allele responsible for Hex A pseudodeficiency (Triggs-Raine et al. Am J Hum Genet, 1992), these two alleles accounted for almost 50% (29/64) of TSD or carrier alleles ascertained by enzyme screening tests in non-Jewish Caucasians.. 1301938 32 49 Tay-Sachs disease SpecificDisease D013661 1301938 78 95 Tay-Sachs disease SpecificDisease D013661 1301938 97 100 TSD SpecificDisease D013661 1301938 108 144 autosomal recessive genetic disorder DiseaseClass D030342 1301938 303 312 Tay-Sachs Modifier D013661 1301938 782 785 TSD Modifier D013661 1301938 1548 1551 TSD Modifier D013661 1302022|t|Characterization of the myotonic dystrophy region predicts multiple protein isoform-encoding mRNAs. 1302022|a|The mutation underlying myotonic dystrophy (DM) has been identified as an expansion of a polymorphic CTG-repeat in a gene encoding protein kinase activity. Brain and heart transcripts of the DM-kinase (DMR-B15) gene are subject to alternative RNA splicing in both human and mouse. The unstable [CTG] 5-30 motif is found uniquely in humans, although the flanking nucleotides are also present in mouse. Characterization of the DM region of both species reveals another active gene (DMR-N9) in close proximity to the kinase gene. DMR-N9 transcripts, mainly expressed in brain and testis, possess a single, large open reading frame, but the function of its protein product is unknown. Clinical manifestation of DM may be caused by the expanded CTG-repeat compromising the (alternative) expression of DM-kinase or DMR-N9 proteins.. 1302022 24 42 myotonic dystrophy Modifier D009223 1302022 124 142 myotonic dystrophy SpecificDisease D009223 1302022 144 146 DM SpecificDisease D009223 1302022 525 527 DM Modifier D009223 1302022 807 809 DM SpecificDisease D009223 2310692|t|Paroxysmal nocturnal haemoglobinuria with coexisting deficiency of the ninth component of complement: lack of massive haemolytic attack. 2310692|a|A 47-year-old woman with paroxysmal nocturnal haemoglobinuria (PNH) was found to have an inherited deficiency in the ninth complement component (C9). In complement-sensitivity lysis tests, 80% of her erythrocytes were markedly complement-sensitive (PNH-III). Laser cytofluorimetry with a monoclonal antibody against decay-accelerating factor (DAF) revealed that 95% of her erythrocytes were DAF-negative. Surprisingly, she has suffered only mild haemolysis and has never experienced massive spontaneous haemolysis. Gross haemoglobinuria and jaundice occurred only after receiving postoperative transfusion of whole blood. In her serum, C9 was not detectable either by immunological or by functional assays. Both the Ham test and the sugar water test using normal human serum or plasma yielded marked haemolysis of the patients erythrocytes. When the patients serum or plasma was used, only a trace of lysis was detected. Addition of purified human C9 to her plasma fully restored haemolysis. These observations indicated that C9 may play a critical role in haemolytic attacks in patients with PNH and that characteristic haemolysis in PNH may be tempered by coexisting C9 deficiency.. 2310692 0 36 Paroxysmal nocturnal haemoglobinuria SpecificDisease D006457 2310692 53 100 deficiency of the ninth component of complement SpecificDisease OMIM:613825 2310692 118 135 haemolytic attack SpecificDisease D006461 2310692 162 198 paroxysmal nocturnal haemoglobinuria SpecificDisease D006457 2310692 200 203 PNH SpecificDisease D006457 2310692 226 280 inherited deficiency in the ninth complement component SpecificDisease OMIM:613825 2310692 583 593 haemolysis SpecificDisease D006461 2310692 640 650 haemolysis SpecificDisease D006461 2310692 658 673 haemoglobinuria SpecificDisease D006456 2310692 678 686 jaundice SpecificDisease D007565 2310692 1194 1212 haemolytic attacks DiseaseClass D006461 2310692 1230 1233 PNH SpecificDisease D006457 2310692 1258 1268 haemolysis SpecificDisease D006461 2310692 1272 1275 PNH SpecificDisease D006457 2310692 1306 1319 C9 deficiency SpecificDisease OMIM:613825 10943845|t|BRCA1 is associated with a human SWI/SNF-related complex: linking chromatin remodeling to breast cancer. 10943845|a|Germline mutations in the tumor suppressor gene, BRCA1, predispose individuals to breast and ovarian cancers. Using a combination of affinity- and conventional chromatographic techniques, we have isolated a predominant form of a multiprotein BRCA1-containing complex from human cells displaying chromatin-remodeling activity. Mass spectrometric sequencing of components of this complex indicated that BRCA1 is associated with a SWI/SNF-related complex. We show that BRCA1 can directly interact with the BRG1 subunit of the SWI/SNF complex. Moreover, p53-mediated stimulation of transcription by BRCA1 was completely abrogated by either a dominant-negative mutant of BRG1 or the cancer-causing deletion in exon 11 of BRCA1. These findings reveal a direct function for BRCA1 in transcriptional control through modulation of chromatin structure.. 10943845 90 103 breast cancer SpecificDisease D001943 10943845 131 136 tumor Modifier D009369 10943845 187 213 breast and ovarian cancers CompositeMention D010051|D001943 10943845 783 789 cancer DiseaseClass D009369 8375105|t|Germinal mosaicism in a Duchenne muscular dystrophy family: implications for genetic counselling. 8375105|a|In this study we describe a three-generation family in which two siblings were affected by Duchenne muscular dystrophy (DMD). Immunohistochemical analysis of muscle dystrophin and haplotype analysis of the DMD locus revealed that the X chromosome carrying the DMD gene was transmitted from the healthy maternal grandfather to his three daughters, including the probands mother. These findings indicate that the grandfather was a germinal mosaic for the DMD gene. The definition of the carrier status in two possible carriers led us to give accurate genetic counselling and to prevent the birth of an affected boy. The results of this study demonstrate the usefulness of haplotype analysis and immunohistochemical muscle dystrophin studies to detect hidden germinal mosaicism and to improve genetic counselling.. 8375105 24 51 Duchenne muscular dystrophy Modifier D020388 8375105 189 216 Duchenne muscular dystrophy SpecificDisease D020388 8375105 218 221 DMD SpecificDisease D020388 8375105 304 307 DMD Modifier D020388 8375105 358 361 DMD Modifier D020388 8375105 551 554 DMD Modifier D020388 10198641|t|Centrosome amplification and a defective G2-M cell cycle checkpoint induce genetic instability in BRCA1 exon 11 isoform-deficient cells. 10198641|a|Germline mutations of the Brca1 tumor suppressor gene predispose women to breast and ovarian cancers. To study mechanisms underlying BRCA1-related tumorigenesis, we derived mouse embryonic fibroblast cells carrying a targeted deletion of exon 11 of the Brca1 gene. We show that the mutant cells maintain an intact G1-S cell cycle checkpoint and proliferate poorly. However, a defective G2-M checkpoint in these cells is accompanied by extensive chromosomal abnormalities. Mutant fibroblasts contain multiple, functional centrosomes, which lead to unequal chromosome segregation, abnormal nuclear division, and aneuploidy. These data uncover an essential role of BRCA1 in maintaining genetic stability through the regulation of centrosome duplication and the G2-M checkpoint and provide a molecular basis for the role of BRCA1 in tumorigenesis.. 10198641 169 174 tumor Modifier D009369 10198641 211 237 breast and ovarian cancers CompositeMention D061325 10198641 582 607 chromosomal abnormalities DiseaseClass D002869 10198641 747 757 aneuploidy SpecificDisease D000782 1769645|t|Aberrant splicing of phenylalanine hydroxylase mRNA: the major cause for phenylketonuria in parts of southern Europe. 1769645|a|We report a mutation within the phenylalanine hydroxylase (PAH) gene that causes aberrant splicing of the mRNA and that is in tight association with chromosomal haplotypes 6, 10, and 36. Because of the high frequency of these particular haplotypes in Bulgaria, Italy, and Turkey, it appears to be one of the more frequent defects in the PAH gene causing classical phenylketonuria in this part of Europe. The mutation is a G to A transition at position 546 in intron 10 of the PAH gene, 11 bp upstream from the intron 10/exon 11 boundary. It activates a cryptic splice site and results in an in-frame insertion of 9 nucleotides between exon 10 and exon 11 of the processed mRNA. Normal amounts of liver PAH protein are present in homozygous patients, but no catalytic activity can be detected. This loss of enzyme activity is probably caused by conformational changes resulting from the insertion of three additional amino acids (Gly-Leu-Gln) between the normal sequences encoded by exon 10 and exon 11.. 1769645 73 88 phenylketonuria SpecificDisease D010661 1769645 482 497 phenylketonuria SpecificDisease D010661 10814710|t|Biochemical and structural analysis of missense mutations in N-acetylgalactosamine-6-sulfate sulfatase causing mucopolysaccharidosis IVA phenotypes. 10814710|a|Mucopolysaccharidosis IVA (MPS IVA; OMIM # 253000), a lysosomal storage disorder caused by a deficiency of N -acetylgalactosamine-6-sulfate sulfatase (GALNS), has variable clinical phenotypes. To date we have identified 65 missense mutations in the GALNS gene from MPS IVA patients, but the correlation between genotype and phenotype has remained unclear. We studied 17 missense mutations using biochemical approaches and 32 missense mutations, using structural analyses. Fifteen missense mutations and two newly engineered active site mutations (C79S, C79T) were characterized by transient expression analysis. Mutant proteins, except for C79S and C79T, were destabilized and detected as insoluble precursor forms while the C79S and C79T mutants were of a soluble mature size. Mutants found in the severe phenotype had no activity. Mutants found in the mild phenotype had a considerable residual activity (1. 3-13. 3% of wild-type GALNS activity). Sulfatases, including GALNS, are members of a highly conserved gene family sharing an extensive sequence homology. Thus, a tertiary structural model of human GALNS was constructed from the X-ray crystal structure of N -acetylgalacto-samine-4-sulfatase and arylsulfatase A, using homology modeling, and 32 missense mutations were investigated. Consequently, we propose that there are at least three different reasons for the severe phenotype (i) destruction of the hydrophobic core or modification of the packing; (ii) removal of a salt bridge to destabilize the entire conformation; (iii) modification of the active site. In contrast, mild mutations were mostly located on the surface of the GALNS protein. These studies shed further light on the genotype-phenotype correlation of MPS IVA and structure-function relationship in the sulfatase family. 10814710 111 136 mucopolysaccharidosis IVA Modifier OMIM:253000 10814710 149 174 Mucopolysaccharidosis IVA SpecificDisease OMIM:253000 10814710 176 183 MPS IVA SpecificDisease OMIM:253000 10814710 203 229 lysosomal storage disorder DiseaseClass D016464 10814710 242 298 deficiency of N -acetylgalactosamine-6-sulfate sulfatase SpecificDisease OMIM:253000 10814710 414 421 MPS IVA Modifier OMIM:253000 10814710 1880 1887 MPS IVA SpecificDisease OMIM:253000 7437512|t|Wiskott-Aldrich syndrome: cellular impairments and their implication for carrier detection. 7437512|a|A family in which two male siblings were affected with Wiskott-Aldrich syndrome (WAS) was studied using G-6-PD isoenzymes as an X-linked marker in order to investigate the nature of cellular abnormalities. Isolated peripheral blood cell types from the doubly heterozygous mother of the affected males seemingly failed to express the G-6-PD allele in cis position with the WAS allele while her cultured skin fibroblasts expressed both G-6-PD alleles. Additionally, a histogram analysis of platelet size revealed a single population of abnormally small platelets in the affected propositus, whereas the heterozygous mother had no appreciable small platelet subpopulation. In vitro culture of hemopoietic progenitor cells of the heterozygous mother showed that the majority of progenitor cells did not express the WAS allele. However, a small number of cells expressing the G-6-PD type linked with the WAS allele were detected. The proportion of the latter progenitors was significantly higher among more primitive progenitors (those giving rise to later appearing colonies). This observation suggests that selection against cells expressing the Wiskott-Aldrich defect takes place in the hemopoietic system of the heterozygous female and offers a possible means of carrier detection in some women. Linkage studies in this family revealed one example of probable recombination between the loci for WAS and G-6-PD among three informative subjects, suggesting that these two loci may not be closely linked on the X-chromosome.. 7437512 0 24 Wiskott-Aldrich syndrome SpecificDisease D014923 7437512 147 171 Wiskott-Aldrich syndrome SpecificDisease D014923 7437512 173 176 WAS SpecificDisease D014923 7437512 464 467 WAS Modifier D014923 7437512 903 906 WAS Modifier D014923 7437512 991 994 WAS Modifier D014923 7437512 1235 1257 Wiskott-Aldrich defect SpecificDisease D014923 7437512 1486 1489 WAS SpecificDisease D014923 2792129|t|Familial deficiency of the seventh component of complement associated with recurrent meningococcal infections. 2792129|a|We describe an 11-year-old girl suffering from recurrent meningitis with a complete absence of the seventh component of complement (C7). Diagnosis was established by haemolytic titration and western blotting. The patients serum lacked the 85 kDa C7 chain. Haemolytic activity of serum was reconstituted with either pooled normal human serum or with purified C7. The relatives (parents and one sister) had half-normal levels of both immunochemically and functionally determined C7, indicating a heterozygous state for C7 deficiency.. 2792129 0 58 Familial deficiency of the seventh component of complement SpecificDisease OMIM:610102 2792129 85 109 meningococcal infections SpecificDisease D008589 2792129 158 178 recurrent meningitis SpecificDisease D008581+D012008 2792129 195 240 absence of the seventh component of complemen SpecificDisease OMIM:610102 2792129 628 641 C7 deficiency SpecificDisease OMIM:610102 10190331|t|Non-syndromic hearing loss associated with enlarged vestibular aqueduct is caused by PDS mutations. 10190331|a|Enlarged vestibular aqueduct (EVA), known as the most common form of inner ear abnormality, has recently been of particular genetic interest because this anomaly is inherited in a recessive manner. The locus for non-syndromic sensorineural hearing loss with EVA has been mapped to the same chromosomal region, 7q31, as the Pendred syndrome locus. In the present study, seven mutations in the PDS gene (PDS), the gene responsible for Pendred syndrome, have been found in families of non-syndromic sensorineural hearing loss with EVA. One family is homozygous, three families are compound heterozygotes, and two families are heterozygous but with no other mutation detected. The present results provide evidence that mutations in PDS cause both syndromic and non-syndromic hearing loss.. 10190331 0 26 Non-syndromic hearing loss SpecificDisease C537845 10190331 43 71 enlarged vestibular aqueduct SpecificDisease OMIM:600791 10190331 85 88 PDS Modifier C536648 10190331 100 128 Enlarged vestibular aqueduct SpecificDisease OMIM:600791 10190331 130 133 EVA SpecificDisease OMIM:600791 10190331 169 190 inner ear abnormality DiseaseClass D007759 10190331 312 352 non-syndromic sensorineural hearing loss SpecificDisease C537845 10190331 358 361 EVA SpecificDisease OMIM:600791 10190331 423 439 Pendred syndrome Modifier C536648 10190331 492 495 PDS Modifier C536648 10190331 533 549 Pendred syndrome SpecificDisease C536648 10190331 582 622 non-syndromic sensorineural hearing loss SpecificDisease C537845 10190331 628 631 EVA SpecificDisease OMIM:600791 10190331 843 883 syndromic and non-syndromic hearing loss CompositeMention D034381|C537845 3480530|t|Deletions of a DNA sequence in retinoblastomas and mesenchymal tumors: organization of the sequence and its encoded protein. 3480530|a|Retinoblastoma is a childhood tumor that can arise because of mutant alleles acquired as somatic or germinal mutations. The mutant allele can be carried in the germ line. The mutations creating these alleles act by inactivating copies of a recessive oncogene located within band q14 of chromosome 13 and termed the RB1 locus. We have reported isolation of a cDNA fragment that recognizes chromosomal sequences possessing many of the attributes of the retinoblastoma gene associated with the RB1 locus. We now report that this segment is additionally the target of somatic mutations in mesenchymal tumors among patients having no apparent predisposition to retinoblastoma and no previous evidence of retinoblastoma. These tumors provide additional evidence that the cloned sequences are representative of a gene that is a frequent target of inactivation during tumorigenesis. Sequence analysis of this cDNA provides little insight into its normal functional role.. 3480530 31 46 retinoblastomas SpecificDisease D012175 3480530 51 69 mesenchymal tumors DiseaseClass C535700 3480530 125 139 Retinoblastoma SpecificDisease D012175 3480530 145 160 childhood tumor DiseaseClass D009369 3480530 576 590 retinoblastoma Modifier D012175 3480530 710 728 mesenchymal tumors DiseaseClass C535700 3480530 781 795 retinoblastoma SpecificDisease D012175 3480530 824 838 retinoblastoma SpecificDisease D012175 3480530 846 852 tumors DiseaseClass D009369 2862466|t|Genetically determined low C4: a predisposing factor to autoimmune chronic active hepatitis. 2862466|a|Of 26 patients with autoimmune chronic active hepatitis (CAH) starting in childhood 18 (69%) had low C4 and 5 (19%) had low C3 serum levels. Impaired hepatic synthesis and immune-consumption were unlikely since transferrin levels were normal in all patients, albumin levels were persistently low in only 3, and only 3 had raised levels of activation fragment C3d. C4d was normal in all patients studied. In the families of 12 probands with low C4, 7 parents had low C4 and 2 had levels which were at the lower limit of normal. 5 of 10 siblings from 5 families had low C4. These results suggest that low C4 levels in CAH are genetically determined. C4 phenotyping in 20 patients and in 26 parents showed that 90% and 81%, respectively, had null allotypes at either the C4A or C4B locus compared with 59% in controls, indicating that defective expression of structural genes may contribute to the observed C4 deficiency.. 2862466 56 91 autoimmune chronic active hepatitis SpecificDisease D006521 2862466 113 148 autoimmune chronic active hepatitis SpecificDisease D006521 2862466 150 153 CAH SpecificDisease D006521 2862466 709 712 CAH SpecificDisease D006521 2862466 997 1010 C4 deficiency SpecificDisease OMIM:614380+OMIM:614379 3417303|t|Tight linkage between myotonic dystrophy and apolipoprotein E genes revealed with allele-specific oligonucleotides. 3417303|a|In 16 families with myotonic dystrophy (DM) a novel approach based on use of allele-specific oligonucleotides has been employed to study the linkage relationship between the apolipoprotein E (APOE) gene and DM. Synthetic oligonucleotides, designed to discriminate between APOE alleles epsilon 3 and epsilon 4, enabled us to distinguish heterozygous carriers in a hybridization assay. In a subset of families, the relevant segment of the APOE gene was enzymatically amplified to increase the sensitivity of the method. For DM and APOE, a maximum lod score (zmax of 7. 47 was obtained at a recombination frequency (theta) of 0. 047 (male theta = female theta). No recombination (maximum lod score of 5. 61 at theta = 0. 0) was found between APOE and the apolipoprotein CII (APOC2) gene. These results suggest that, in addition to APOC2, APOE is a useful marker for presymptomatic DM diagnosis. 3417303 22 40 myotonic dystrophy Modifier D009223 3417303 136 154 myotonic dystrophy SpecificDisease D009223 3417303 156 158 DM SpecificDisease D009223 3417303 323 325 DM SpecificDisease D009223 3417303 994 996 DM Modifier D009223 7894493|t|Confirmation of BRCA1 by analysis of germline mutations linked to breast and ovarian cancer in ten families. 7894493|a|We provide genetic evidence supporting the identity of the candidate gene for BRCA1 through the characterization of germline mutations in 63 breast cancer patients and 10 ovarian cancer patients in ten families with cancer linked to chromosome 17q21. Nine different mutations were detected by screening BRCA1 DNA and RNA by single-strand conformation polymorphism analysis and direct sequencing. Seven mutations lead to protein truncations at sites throughout the gene. One missense mutation (which occurred independently in two families) leads to loss of a cysteine in the zinc binding domain. An intronic single basepair substitution destroys an acceptor site and activates a cryptic splice site, leading to a 59 basepair insertion and chain termination. The four families with both breast and ovarian cancer had chain termination mutations in the N-terminal half of the protein.. 7894493 66 91 breast and ovarian cancer CompositeMention D061325 7894493 250 263 breast cancer Modifier D001943 7894493 280 294 ovarian cancer Modifier D010051 7894493 325 331 cancer DiseaseClass D009369 7894493 894 919 breast and ovarian cancer CompositeMention D061325 7937795|t|Genetic instability in human ovarian cancer cell lines. 7937795|a|We have analyzed the stability of microsatellites in cell lines derived from human ovarian cancers and found that 5 out of 10 of the ovarian tumor cell lines are genetically unstable at the majority of the loci analyzed. In clones and subclones derived serially from one of these cell lines (2774; serous cystadenocarcinoma), a very high proportion of microsatellites distributed in many different regions of the genome change their size in a mercurial fashion. We conclude that genomic instability in ovarian tumors is a dynamic and ongoing process whose high frequency may have been previously underestimated by PCR-based allelotyping of bulk tumor tissue. We have identified the source of the genetic instability in one ovarian tumor as a point mutation (R524P) in the human mismatch-repair gene MSH2 (Salmonella MutS homologue), which has recently been shown to be involved in hereditary nonpolyposis colorectal cancer. Patient 2774 was a 38-year-old heterozygote, and her normal tissue carried both mutant and wild-type alleles of the human MSH2 gene. However the wild-type allele was lost at some point early during tumorigenesis so that DNA isolated either from the patients ovarian tumor or from the 2774 cell line carries only the mutant allele of the human MSH2 gene. The genetic instability observed in the tumor and cell line DNA, together with the germ-line mutation in a mismatch-repair gene, suggest that the MSH2 gene is involved in the onset and/or progression in a subset of ovarian cancer.. 7937795 29 43 ovarian cancer Modifier D010051 7937795 139 154 ovarian cancers SpecificDisease D010051 7937795 189 202 ovarian tumor Modifier D010051 7937795 354 379 serous cystadenocarcinoma SpecificDisease D018284 7937795 558 572 ovarian tumors SpecificDisease D010051 7937795 701 706 tumor Modifier D009369 7937795 779 792 ovarian tumor SpecificDisease D010051 7937795 937 978 hereditary nonpolyposis colorectal cancer SpecificDisease D003123 7937795 1238 1251 ovarian tumor SpecificDisease D010051 7937795 1374 1379 tumor DiseaseClass D009369 7937795 1549 1563 ovarian cancer SpecificDisease D010051 3455778|t|Retroviral-mediated gene transfer of human phenylalanine hydroxylase into NIH 3T3 and hepatoma cells. 3455778|a|Phenylketonuria (PKU) is caused by deficiency of the hepatic enzyme phenylalanine hydroxylase (PAH). A full-length human PAH cDNA sequence has been inserted into pzip-neoSV (X), which is a retroviral vector containing the bacterial neo gene. The recombinant has been transfected into psi 2 cells, which provide synthesis of the retroviral capsid. Recombinant virus was detected in the culture medium of the transfected psi 2 cells, which is capable of transmitting the human PAH gene into mouse NIH 3T3 cells by infection leading to stable incorporation of the recombinant provirus. Infected cells express PAH mRNA, immunoreactive PAH protein, and exhibit pterin-dependent phenylalanine hydroxylase activity. The recombinant virus is also capable of infecting a mouse hepatoma cell line that does not normally synthesize PAH. PAH activity is present in the cellular extracts and the entire hydroxylation system is reconstituted in the hepatoma cells infected with the recombinant viruses. Thus, recombinant viruses containing human PAH cDNA provide a means for introducing functional PAH into mammalian cells of hepatic origin and can potentially be introduced into whole animals as a model for somatic gene therapy for PKU.. 3455778 86 94 hepatoma Modifier D006528 3455778 102 117 Phenylketonuria SpecificDisease D010661 3455778 119 122 PKU SpecificDisease D010661 3455778 137 195 deficiency of the hepatic enzyme phenylalanine hydroxylase SpecificDisease OMIM:261600 3455778 870 878 hepatoma Modifier D006528 3455778 1037 1045 hepatoma Modifier D006528 3455778 1322 1325 PKU SpecificDisease D010661 7599636|t|Characterisation of molecular defects in X-linked amelogenesis imperfecta (AIH1). 7599636|a|Amelogenins are an heterogenous family of proteins produced by ameloblasts of the enamel organ during tooth development. Disturbances of enamel formation occur in amelogenesis imperfecta, a clinically heterogenous group of inherited disorders characterised by defective enamel biomineralisation. An amelogenin gene, AMGX, has been mapped to the short of the X chromosome (Xp22. 1-p22. 3) and has been implicated in the molecular pathology of X-linked amelogenesis imperfecta (AIH1). We have identified three families exhibiting AIH1 and screened the AMGX gene for mutations using single-strand conformational polymorphism analysis and DNA sequencing. Three novel mutations were identified a C-T substitution in exon 5, and a G-T substitution and single cytosine deletion in exon 6, confirming the existence of extensive allelic heterogeneity in this condition. The identification of family-specific mutations will enable early identification of affected individuals and correlation of clinical phenotype with genotype will facilitate an objective system of disease classification. 7599636 41 73 X-linked amelogenesis imperfecta SpecificDisease C538243 7599636 245 268 amelogenesis imperfecta DiseaseClass D000567 7599636 305 324 inherited disorders DiseaseClass D030342 7599636 524 556 X-linked amelogenesis imperfecta SpecificDisease C538243 7599636 558 562 AIH1 SpecificDisease C538243 7599636 610 614 AIH1 SpecificDisease C538243 10192399|t|The Pendred syndrome gene encodes a chloride-iodide transport protein. 10192399|a|Pendred syndrome is the most common form of syndromic deafness and characterized by congenital sensorineural hearing loss and goitre. This disorder was mapped to chromosome 7 and the gene causing Pendred syndrome (PDS) was subsequently identified by positional cloning. PDS encodes a putative transmembrane protein designated pendrin. Pendrin is closely related to a family of sulfate transport proteins that includes the rat sulfate-anion transporter (encoded by Sat-1; 29% amino acid sequence identity), the human diastrophic dysplasia sulfate transporter (encoded by DTD; 32%) and the human sulfate transporter downregulated in adenoma (encoded by DRA; 45%). On the basis of this homology and the presence of a slightly modified sulfate-transporter signature sequence comprising its putative second transmembrane domain, pendrin has been proposed to function as a sulfate transporter. We were unable to detect evidence of sulfate transport following the expression of pendrin in Xenopus laevis oocytes by microinjection of PDS cRNA or in Sf9 cells following infection with PDS-recombinant baculovirus. The rates of transport for iodide and chloride were significantly increased following the expression of pendrin in both cell systems. Our results demonstrate that pendrin functions as a transporter of chloride and iodide, but not sulfate, and may provide insight into thyroid physiology and the pathophysiology of Pendred syndrome.. 10192399 4 20 Pendred syndrome Modifier C536648 10192399 71 87 Pendred syndrome SpecificDisease C536648 10192399 115 133 syndromic deafness DiseaseClass D003638 10192399 166 192 sensorineural hearing loss SpecificDisease D006319 10192399 197 203 goitre SpecificDisease D006042 10192399 267 283 Pendred syndrome SpecificDisease C536648 10192399 285 288 PDS SpecificDisease C536648 10192399 587 608 diastrophic dysplasia Modifier C536170 10192399 702 709 adenoma DiseaseClass D000236 10192399 1490 1506 Pendred syndrome SpecificDisease C536648 161677|t|Linkage relationship of C2 deficiency, HLA and glyoxalase I loci. 161677|a|Immunogenetic analysis of a homozygous C2-deficient individual and family members demonstrated linkage of HLA-A25, B18 and C2o. HLA-D typing showed that 5 members typed with homozygous Dw2 typing cells from an individual with C2 deficiency but not with Dw2 typing cells from 2 individuals with normal C2. The homozygous C2-deficient propositus and brother were HLA-A and B homozygous but heterozygous at the HLA-D and glyoxalase I loci. Therefore, in this family, the C2o gene is linked with two distinct haplotypes HLA-A25, B18, Dw2, GLO1 and HLA-A25, B18, D unknown, GL02. These results could be explained by an ancestral recombinant event, which occurred between the C2o locus and HLA-D locus in which C2o segregated with HLA-B. This would suggest that the locus for the C2o gene maps between HLA-B and HLA-D on the sixth chromosome.. 161677 24 37 C2 deficiency SpecificDisease OMIM:217000 161677 105 117 C2-deficient Modifier OMIM:217000 161677 292 305 C2 deficiency SpecificDisease OMIM:217000 161677 386 398 C2-deficient Modifier OMIM:217000 1319059|t|A 71-kilodalton protein is a major product of the Duchenne muscular dystrophy gene in brain and other nonmuscle tissues. 1319059|a|The known Duchenne muscular dystrophy (DMD) gene products, the muscle- and brain-type dystrophin isoforms, are 427-kDa proteins translated from 14-kilobase (kb) mRNAs. Recently we described a 6. 5-kb mRNA that also is transcribed from the DMD gene. Cloning and in vitro transcription and translation of the entire coding region show that the 6. 5-kb mRNA encodes a 70. 8-kDa protein that is a major product of the DMD gene. It contains the C-terminal and the cysteine-rich domains of dystrophin, seven additional amino acids at the N terminus, and some modifications formed by alternative splicing in the C-terminal domain. It lacks the entire large domain of spectrin-like repeats and the actin-binding N-terminal domain of dystrophin. This protein is the major DMD gene product in brain and other nonmuscle tissues but is undetectable in skeletal muscle extracts. 1319059 50 77 Duchenne muscular dystrophy Modifier D020388 1319059 131 158 Duchenne muscular dystrophy Modifier D020388 1319059 160 163 DMD Modifier D020388 1319059 360 363 DMD Modifier D020388 1319059 535 538 DMD Modifier D020388 1319059 884 887 DMD Modifier D020388 1146783|t|Histidinemia. Classical and atypical form in siblings. 1146783|a|Two brothers, 6 and 13 years old, had histidinemia. On the basis of clinical and biochemical observations, the younger boy was considered to have a classical type of the disease, while the older boy had an atypical form characterized by partial impairment of the skin histidase activity and a moderately prolonged half-life of blood histidine. The mother is a heterozygous carrier, while the father and sister seem to be normal.. 1146783 0 12 Histidinemia SpecificDisease C538320 1146783 93 105 histidinemia SpecificDisease C538320 1937471|t|Hypoxanthine-guanine phosphoribosyltransferase deficiency: analysis of HPRT mutations by direct sequencing and allele-specific amplification. 1937471|a|The Lesch-Nyhan syndrome is a severe X chromosome-linked human disease caused by a virtual absence of hypoxanthine-guanine phosphoribosyltransferase (HPRT) activity. A partial deficiency in the activity of this enzyme can result in gouty arthritis. To determine the genetic basis for reduction or loss of enzyme activity, we have amplified and sequenced the coding region of HPRT cDNA from four patients one with Lesch-Nyhan syndrome (HPRTPerth) and three with partial deficiencies of HPRT activity, which have been designated HPRTUrangan, HPRTSwan and HPRTToowong. In all four patients, the only mutation identified was a single base substitution in exons 2 or 3 of the coding region, which in each case predicts a single amino acid substitution in the translated protein. Each base change was confirmed by allele-specific amplification of the patients genomic DNA. It is interesting to note that the mutation found for HPRTPerth is identical to that reported for HPRTFlint. It appears that the two mutations are de novo events.. 1937471 0 57 Hypoxanthine-guanine phosphoribosyltransferase deficiency SpecificDisease OMIM:300323 1937471 146 166 Lesch-Nyhan syndrome SpecificDisease D007926 1937471 179 212 X chromosome-linked human disease DiseaseClass D040181 1937471 233 306 absence of hypoxanthine-guanine phosphoribosyltransferase (HPRT) activity SpecificDisease D007926 1937471 374 389 gouty arthritis SpecificDisease D015210 1937471 556 576 Lesch-Nyhan syndrome SpecificDisease D007926 1937471 604 641 partial deficiencies of HPRT activity DiseaseClass OMIM:300323 3343337|t|Sjogren-Larsson syndrome. Impaired fatty alcohol oxidation in cultured fibroblasts due to deficient fatty alcohol:nicotinamide adenine dinucleotide oxidoreductase activity. 3343337|a|Lipid metabolism was studied in cultured skin fibroblasts from patients with the inherited disorder, Sjogren-Larsson syndrome (SLS). Intact SLS fibroblasts incubated in the presence of [1-14C] palmitate accumulated more radioactive hexadecanol than did normal cells, whereas incorporation of radioactivity into other cellular lipids was unaltered. The hexadecanol content of SLS fibroblasts was abnormally elevated. Hexadecanol accumulation was not due to increased fatty alcohol synthesis nor its deficient utilization for glycerol ether synthesis. The half-life of intracellular hexadecanol loaded into SLS fibroblasts was increased (70 min) compared with normal (15 min), and intact SLS fibroblasts showed impaired oxidation of [14C] -hexadecanol to fatty acid. Fatty alcohol NAD + oxidoreductase, the enzyme catalyzing this reaction, was deficient in SLS fibroblasts. Mean total activity in SLS fibroblasts (n = 5) was 13% of that in normal fibroblasts, and palmitoyl CoA-inhibitable activity was 1% of normal. Fibroblasts from two obligate SLS heterozygotes had enzyme activities intermediate between that in normal fibroblasts and individuals with SLS. These results suggest that the primary defect in SLS is deficiency of fatty alcohol NAD + oxidoreductase. SLS represents the first inherited disorder in man associated with an isolated abnormality in fatty alcohol metabolism.. 3343337 0 24 Sjogren-Larsson syndrome SpecificDisease D016111 3343337 254 272 inherited disorder DiseaseClass D030342 3343337 274 298 Sjogren-Larsson syndrome SpecificDisease D016111 3343337 300 303 SLS SpecificDisease D016111 3343337 313 316 SLS Modifier D016111 3343337 548 551 SLS Modifier D016111 3343337 778 781 SLS Modifier D016111 3343337 859 862 SLS Modifier D016111 3343337 1029 1032 SLS Modifier D016111 3343337 1069 1072 SLS Modifier D016111 3343337 1219 1222 SLS Modifier D016111 3343337 1328 1331 SLS SpecificDisease D016111 3343337 1382 1385 SLS SpecificDisease D016111 3343337 1389 1438 deficiency of fatty alcohol NAD + oxidoreductase SpecificDisease D016111 3343337 1440 1443 SLS SpecificDisease D016111 3343337 1465 1483 inherited disorder DiseaseClass D030342 3343337 1519 1558 abnormality in fatty alcohol metabolism DiseaseClass D005235 10736265|t|Glycerol as a correlate of impaired glucose tolerance: dissection of a complex system by use of a simple genetic trait. 10736265|a|Glycerol kinase (GK) represents the primary entry of glycerol into glucose and triglyceride metabolism. Impaired glucose tolerance (IGT) and hypertriglyceridemia are associated with an increased risk of diabetes mellitus and cardiovascular disease. The relationship between glycerol and the risk of IGT, however, is poorly understood. We therefore undertook the study of fasting plasma glycerol levels in a cohort of 1, 056 unrelated men and women of French-Canadian descent. Family screening in the initial cohort identified 18 men from five families with severe hyperglycerolemia (values above 2. 0 mmol/liter) and demonstrated an X-linked pattern of inheritance. Linkage analysis of the data from 12 microsatellite markers surrounding the Xp21. 3 GK gene resulted in a peak LOD score of 3. 46, centered around marker DXS8039. In addition, since all of the families originated in a population with a proven founder effect-the Saguenay Lac-St. -Jean region of Quebec-a common disease haplotype was sought. Indeed, a six-marker haplotype extending over a region of 5. 5 cM was observed in all families. Resequencing of the GK gene in family members led to the discovery of a N288D missense mutation in exon 10, which resulted in the substitution of a highly conserved asparagine residue by a negatively charged aspartic acid. 10736265 27 53 impaired glucose tolerance SpecificDisease D018149 10736265 224 250 Impaired glucose tolerance SpecificDisease D018149 10736265 252 255 IGT SpecificDisease D018149 10736265 261 281 hypertriglyceridemia SpecificDisease D015228 10736265 323 340 diabetes mellitus SpecificDisease D003920 10736265 345 367 cardiovascular disease SpecificDisease D002318 10736265 419 422 IGT SpecificDisease D018149 10736265 684 701 hyperglycerolemia SpecificDisease C538138 1505217|t|Assignment of the aspartylglucosaminidase gene (AGA) to 4q33----q35 based on decreased activity in a girl with a 46,XX,del(4)(q33) karyotype. 1505217|a|Aspartylglucosaminuria (AGU) is a recessive autosomally inherited lysosomal storage disorder due to deficiency of the enzyme aspartylglucosaminidase (AGA). The structural gene for this human enzyme (AGA) has been assigned to the region 4q21----qter. We determined the AGA activity in cultured fibroblasts of a girl with a 46, XX, del (4) (q33) karyotype. The results indicate that the girl is a hemizygote for AGA, permitting the assignment of human AGA to the region 4q33----qter.. 1505217 142 164 Aspartylglucosaminuria SpecificDisease D054880 1505217 166 169 AGU SpecificDisease D054880 1505217 208 234 lysosomal storage disorder DiseaseClass D016464 1505217 242 290 deficiency of the enzyme aspartylglucosaminidase SpecificDisease D054880 10891444|t|Mutations in the fibrinogen aalpha gene account for the majority of cases of congenital afibrinogenemia. 10891444|a|Congenital afibrinogenemia is a rare, autosomal, recessive disorder characterized by the complete absence of detectable fibrinogen. We previously identified the first causative mutations in a nonconsanguineous Swiss family; the 4 affected persons have homozygous deletions of approximately 11 kb of the fibrinogen alpha (FGA) gene. Haplotype data implied that these deletions occurred on distinct ancestral chromosomes, suggesting that this region may be susceptible to deletion by a common mechanism. We subsequently showed that all the deletions were identical to the base pair and probably resulted from a nonhomologous recombination mediated by 7-bp direct repeats. In this study, we have collected data on 13 additional unrelated patients to identify the causative mutations and to determine the prevalence of the 11-kb deletion. A common recurrent mutation, at the donor splice site of FGA intron 4 (IVS4 + 1 G > T), accounted for 14 of the 26 (54%) alleles. One patient was heterozygous for the previously identified deletion. Three more frameshift mutations, 2 nonsense mutations, and a second splice site mutation were also identified. Consequently, 86% of afibrinogenemia alleles analyzed to date have truncating mutations of FGA, though mutations in all 3 fibrinogen genes, FGG, FGA, and FGB, might be predicted to cause congenital afibrinogenemia.. 10891444 77 103 congenital afibrinogenemia SpecificDisease C531603 10891444 105 131 Congenital afibrinogenemia SpecificDisease C531603 10891444 143 172 autosomal, recessive disorder DiseaseClass D030342 10891444 1271 1286 afibrinogenemia Modifier D000347 10891444 1437 1463 congenital afibrinogenemia SpecificDisease C531603 523196|t|Utilization of purines by an HPRT variant in an intelligent, nonmutilative patient with features of the Lesch-Nyhan syndrome. 523196|a|The patient, H. Chr. B., was among the first reported with hyperuricemia and central nervous system symptoms. He has been found to have a variant of hypoxanthine guanine phosphoribosyl transferase (HPRT; E. C. 2. 4. 2. 8) distinct from the enzyme present in patients with the Lesch-Nyhan syndrome. The patient had chroeoathetosis, spasticity, dysarthric speech, and hyperuricemia. However, his intelligence was normal and he had no evidence of self-mutilation. There was no activity of HPRT in the lysates of erythrocytes and cultured fibroblasts when analyzed in the usual manner. Using a newly developed method for the study of purine metabolism in intact cultured cells, this patient was found to metabolize some 9% of 8-14C-hypoxanthine, and 90% of the isotope utilized was converted to adenine and guanine nucleotides. In contrast, cells from patients with the Lesch-Nyhan syndrome were virtually completely unable to convert hypoxanthine to nucleotides. The patients fibroblasts were even more efficient in the metabolism of 8-14C-guanine, which was utilized to the extent of 27%, over 80% of which was converted to guanine and adenine nucleotides. The growth of the cultured fibroblasts of this patient was intermediate in media containing hypoxanthine aminopterin thymidine (HAT), whereas the growth of Lesch-Nyhan cells was inhibited and normal cells grew normally. Similarly in 8-azaguanine, 6-thioguanine, and 8-azahypoxanthine, the growth of the patients cells was intermediate between normal and Lesch-Nyhan cells. These observations provide further evidence for genetic heterogeneity among patients with disorders in purine metabolism involving the HPRT gene. They document that this famous patient did not have the Lesch-Nyhan syndrome 523196 104 124 Lesch-Nyhan syndrome SpecificDisease D007926 523196 185 198 hyperuricemia SpecificDisease D033461 523196 203 234 central nervous system symptoms DiseaseClass D002493 523196 402 422 Lesch-Nyhan syndrome SpecificDisease D007926 523196 440 455 chroeoathetosis SpecificDisease C537181 523196 457 467 spasticity SpecificDisease D009128 523196 469 486 dysarthric speech SpecificDisease D004401 523196 492 505 hyperuricemia SpecificDisease D033461 523196 992 1012 Lesch-Nyhan syndrome SpecificDisease D007926 523196 1437 1448 Lesch-Nyhan Modifier D007926 523196 1635 1646 Lesch-Nyhan Modifier D007926 523196 1856 1876 Lesch-Nyhan syndrome SpecificDisease D007926 8023850|t|Canavan disease: mutations among Jewish and non-Jewish patients. 8023850|a|Canavan disease is an autosomal recessive leukodystrophy caused by the deficiency of aspartoacylase (ASPA). Sixty-four probands were analyzed for mutations in the ASPA gene. Three point mutations--693C-- > A, 854A-- > C, and 914C-- > A--were identified in the coding sequence. The 693C-- > A and 914C-- > A base changes, resulting in nonsense tyr231-- > ter and missense ala305-- > glu mutations, respectively, lead to complete loss of ASPA activity in in vitro expression studies. The 854A-- > C transversion converted glu to ala in codon 285. The glu285-- > ala mutant ASPA has 2. 5% of the activity expressed by the wild-type enzyme. A fourth mutation, 433 --2 (A-- > G) transition, was identified at the splice-acceptor site in intron 2. The splice-site mutation would lead to skipping of exon 3, accompanied by a frameshift, and thus would produce aberrant ASPA. Of the 128 unrelated Canavan chromosomes analyzed, 88 were from probands of Ashkenazi Jewish descent. The glu285-- > ala mutation was predominant (82. 9%) in this population, followed by the tyr231-- > ter (14. 8%) and 433 --2 (A-- > G) (1. 1%) mutations. The three mutations account for 98. 8% of the Canavan chromosomes of Ashkenazi Jewish origin. The ala305-- > glu mutation was found exclusively in non-Jewish probands of European descent and constituted 60% of the 40 mutant chromosomes. Predominant occurrence of certain mutations among Ashkenazi Jewish and non-Jewish patients with Canavan disease would suggest a founding-father effect in propagation of these mutant chromosomes 8023850 0 15 Canavan disease SpecificDisease D017825 8023850 65 80 Canavan disease SpecificDisease D017825 8023850 87 121 autosomal recessive leukodystrophy DiseaseClass D007966 8023850 136 164 deficiency of aspartoacylase SpecificDisease D017825 8023850 954 961 Canavan Modifier D017825 8023850 1235 1242 Canavan Modifier D017825 8023850 1522 1537 Canavan disease SpecificDisease D017825 1678319|t|Identification of deletion mutations and three new genes at the familial polyposis locus. 1678319|a|Small (100-260 kb), nested deletions were characterized in DNA from two unrelated patients with familial adenomatous polyposis coli (APC). Three candidate genes located within the deleted region were ascertained and a previous candidate gene, MCC, was shown to be located outside the deleted region. One of the new genes contained sequence identical to SRP19, the gene coding for the 19 kd component of the ribosomal signal recognition particle. The second, provisionally designated DP1 (deleted in polyposis 1), was found to be transcribed in the same orientation as MCC. Two other cDNAs, DP2 and DP3, were found to overlap, forming a single gene, DP2. 5, that is transcribed in the same orientation as SRP19. 1678319 64 82 familial polyposis Modifier D011125 1678319 186 221 familial adenomatous polyposis coli SpecificDisease D011125 1678319 223 226 APC SpecificDisease D011125 10830910|t|Genotype-phenotype correlations in families with deletions in the von Hippel-Lindau (VHL) gene. 10830910|a|Von Hippel-Lindau (VHL) disease is a hereditary tumor syndrome characterized by predisposition for bilateral and multi-centric hemangioblastoma in the retina and central nervous system, pheochromocytoma, renal cell carcinoma, and cysts in the kidney, pancreas, and epididymis. We describe five families for which direct sequencing of the coding region of the VHL gene had failed to identify the family-specific mutation. Further molecular analysis revealed deletions involving the VHL gene in each of these families. In four families, partial deletions of one or more exons were detected by Southern blot analysis. In the fifth family, FISH analysis demonstrated the deletion of the entire VHL gene. Our results show that (quantitative) Southern blot analysis is a sensitive method for detecting germline deletions of the VHL gene and should be implemented in routine DNA diagnosis for VHL disease. Our data support the previously established observation that families with a germline deletion have a low risk for pheochromocytoma. Further unraveling of genotype-phenotype correlations in VHL disease has revealed that families with a full or partial deletion of the VHL gene exhibit a phenotype with a preponderance of central nervous system hemangioblastoma.. 10830910 66 83 von Hippel-Lindau Modifier D006623 10830910 85 88 VHL Modifier D006623 10830910 96 127 Von Hippel-Lindau (VHL) disease SpecificDisease D006623 10830910 133 158 hereditary tumor syndrome DiseaseClass D009386 10830910 195 239 bilateral and multi-centric hemangioblastoma CompositeMention D018325 10830910 282 298 pheochromocytoma SpecificDisease D010673 10830910 300 320 renal cell carcinoma SpecificDisease D002292 10830910 326 371 cysts in the kidney, pancreas, and epididymis CompositeMention D052177|D010181|D013088 10830910 455 458 VHL Modifier D006623 10830910 577 580 VHL Modifier D006623 10830910 786 789 VHL Modifier D006623 10830910 918 921 VHL Modifier D006623 10830910 982 993 VHL disease SpecificDisease D006623 10830910 1110 1126 pheochromocytoma SpecificDisease D010673 10830910 1185 1196 VHL disease SpecificDisease D006623 10830910 1263 1266 VHL Modifier D006623 10830910 1316 1355 central nervous system hemangioblastoma SpecificDisease D018325 10767313|t|Understanding the molecular basis of fragile X syndrome. 10767313|a|Fragile X syndrome, a common form of inherited mental retardation, is mainly caused by massive expansion of CGG triplet repeats located in the 5-untranslated region of the fragile X mental retardation-1 (FMR1) gene. In patients with fragile X syndrome, the expanded CGG triplet repeats are hypermethylated and the expression of the FMR1 gene is repressed, which leads to the absence of FMR1 protein (FMRP) and subsequent mental retardation. FMRP is an RNA-binding protein that shuttles between the nucleus and cytoplasm. This protein has been implicated in protein translation as it is found associated with polyribosomes and the rough endoplasmic reticulum. We discuss here the recent progress made towards understanding the molecular mechanism of CGG repeat expansion and physiological function (s) of FMRP. These studies will not only help to illuminate the molecular basis of the general class of human diseases with trinucleotide repeat expansion but also provide an avenue to understand aspects of human cognition and intelligence.. 10767313 37 55 fragile X syndrome SpecificDisease D005600 10767313 57 75 Fragile X syndrome SpecificDisease D005600 10767313 94 122 inherited mental retardation DiseaseClass D038901 10767313 229 257 fragile X mental retardation Modifier OMIM:300624 10767313 290 308 fragile X syndrome SpecificDisease D005600 10767313 478 496 mental retardation DiseaseClass D008607 7076260|t|Glucose-6-phosphate dehydrogenase deficiency in Papua New Guinea. The description of 13 new variants. 7076260|a|A total of 362 males from various regions of Papua New Guinea were screened for red cell glucose-6-phosphate dehydrogenase (G6PD) activity. Twenty-six G6PD deficient individuals were identified. Biochemical characterization of G6PD purified from these subjects has revealed 13 new variants and several copies of previously described forms of G6PD. This study illustrates the extreme heterogeneity of G6PD deficiency among the people of Papua New Guinea.. 7076260 0 44 Glucose-6-phosphate dehydrogenase deficiency SpecificDisease D005955 7076260 253 267 G6PD deficient Modifier D005955 7076260 502 517 G6PD deficiency SpecificDisease D005955 3862128|t|Regional mapping of the phenylalanine hydroxylase gene and the phenylketonuria locus in the human genome. 3862128|a|Phenylketonuria (PKU) is an autosomal recessive disorder of amino acid metabolism caused by a deficiency of the hepatic enzyme phenylalanine hydroxylase (PAH; phenylalanine 4-monooxygenase, EC 1. 14. 16. 1). A cDNA clone for human PAH has previously been used to assign the corresponding gene to human chromosome 12. To define the regional map position of the disease locus and the PAH gene on human chromosome 12, DNA was isolated from human-hamster somatic cell hybrids with various deletions of human chromosome 12 and was analyzed by Southern blot analysis using the human cDNA PAH clone as a hybridization probe. From these results, together with detailed biochemical and cytogenetic characterization of the hybrid cells, the region on chromosome 12 containing the human PAH gene has been defined as 12q14. 3----qter 3----qter. The PAH map position on chromosome 12 was further localized by in situ hybridization of 125I-labeled human PAH cDNA to chromosomes prepared from a human lymphoblastoid cell line. Results of these experiments demonstrated that the region on chromosome 12 containing the PAH gene and the PKU locus in man is 12q22----12q24. 1. These results not only provide a regionalized map position for a major human disease locus but also can serve as a reference point for linkage analysis with other DNA markers on human chromosome 12 3862128 63 78 phenylketonuria Modifier D010661 3862128 106 121 Phenylketonuria SpecificDisease D010661 3862128 123 126 PKU SpecificDisease D010661 3862128 134 162 autosomal recessive disorder DiseaseClass D030342 3862128 200 258 deficiency of the hepatic enzyme phenylalanine hydroxylase SpecificDisease OMIM:261600 3862128 1225 1228 PKU Modifier D010661 2390095|t|Total deficiency of plasma cholesteryl ester transfer protein in subjects homozygous and heterozygous for the intron 14 splicing defect. 2390095|a|The molecular basis of cholesteryl ester transfer protein (CETP) deficiency was investigated in 4 unrelated CETP-deficient families. The high density lipoprotein-cholesterol levels of the probands exceeded 150 mg/dl. The plasma of the probands was totally deficient in CETP activity and mass. The genomic DNA of the patients was amplified by polymerase chain reaction, using two oligonucleotide primers located in the intron 12 and 14 of the CETP gene, and the amplified products were directly sequenced. Two patients were homozygous for a G-to-A change at the 5-splice donor site of the intron 14. The G-to-A change would cause impaired splicing of pre-messenger RNA. The other two probands were heterozygous for the mutation, but totally lacked CETP. Their lipoprotein patterns were also similar to those of the two homozygotes. Thus, other genetic defects or metabolic factors influencing CETP expression are implicated. The data suggest that the G-to-A mutation may be common in human plasma CETP deficiency. Furthermore, there could be compound heterozygotes who totally lack plasma CETP and have lipoprotein profiles similar to those of homozygotes.. 2390095 0 61 Total deficiency of plasma cholesteryl ester transfer protein SpecificDisease OMIM:143470 2390095 160 212 cholesteryl ester transfer protein (CETP) deficiency SpecificDisease OMIM:143470 2390095 245 259 CETP-deficient Modifier OMIM:143470 2390095 980 995 genetic defects DiseaseClass D030342 2390095 1133 1148 CETP deficiency SpecificDisease OMIM:143470 3876122|t|Heterogeneity of type I von Willebrand disease: evidence for a subgroup with an abnormal von Willebrand factor. 3876122|a|Type I von Willebrand disease (vWD) is characterized by equally low plasma concentrations of von Willebrand factor antigen (vWF Ag) and ristocetin cofactor (RiCof) and by the presence of all vWF multimers in sodium dodecyl sulfate (SDS) -agarose gel electrophoresis. For 17 patients (13 kindreds) diagnosed with these criteria, we have studied the platelet contents of vWF Ag and RiCof and the changes of these in plasma after DDAVP infusion. Platelet vWF Ag and RiCof were normal in four kindreds (called " platelet normal " subgroup); following 1-deamino-8-D-arginine vasopressin; plasma vWF Ag, RiCof and the bleeding time (BT) became normal. In six kindreds, platelet vWF Ag and RiCof were equally low (platelet low); after DDAVP, plasma vWF Ag and RiCof remained low, and the BT was prolonged. In three additional kindreds, platelets contained normal concentrations of vWF Ag, but RiCof was very low (platelet discordant); even though a complete set of multimers was found in plasma and platelets, there was a relatively small amount of large multimers. After DDAVP, plasma vWF Ag became normal, but RiCof remained low and the BT was very prolonged. These findings demonstrated that there can be an abnormal vWF (RiCof less than vWF Ag) even in type I vWD, coexisting with a complete set of vWF multimers (platelet discordant); that the abnormal vWF can be shown more clearly in platelets than in plasma or else in plasma after DDAVP infusion; and that DDAVP normalizes the BT only in those patients with normal platelet levels of both vWF Ag and RiCof (platelet normal).. 3876122 17 46 type I von Willebrand disease SpecificDisease D056725 3876122 89 103 von Willebrand Modifier D014842 3876122 112 141 Type I von Willebrand disease SpecificDisease D056725 3876122 143 146 vWD SpecificDisease D014842 3876122 205 219 von Willebrand Modifier D014842 3876122 1371 1381 type I vWD SpecificDisease D056725 1303171|t|Characterization of a YAC containing part or all of the Norrie disease locus. 1303171|a|It has been shown from pulsed-field gel electrophoresis (PFGE) that the monoamine oxidase genes A and B (MAOA & MAOB) and DXS7 loci are physically very close. We have therefore extended studies on their relationship through the characterisation of a 650 kb YAC isolated using L1. 28 (recognising the DXS7 locus) as a probe. Restriction mapping of the YAC indicates that it contains both MAOA and MAOB genes in addition to the DXS7 locus. The map derived from the YL1. 28-YAC is compatible both with the map from an independently derived YAC carrying MAOA and B genes and with the long range genomic map for the region. A series of subclones prepared from a phage library (lambda DASH II) of the YAC have been characterised and have been employed to determine the end point of the deletion of a Norrie disease (NDP) patient who has been shown to lack both DXS7 and MAO coding sequences. The pattern of retention of subclones in the deletion patient place the end point of the deletion within 30-130 kb of the proximal end of the YAC. By combining the data with established recombination analysis, we provide evidence that all or part of the NDP lies in the interval of approximately 250kb within the YAC. 1303171 56 70 Norrie disease Modifier C537849 1303171 872 886 Norrie disease Modifier C537849 1303171 888 891 NDP Modifier C537849 8477262|t|Identification of mutations in Danish choroideremia families. 8477262|a|We have searched for mutations in the choroideremia gene (CHM) in patients from 12 Danish families in which CHM is segregating. Employing polymerase chain reaction (PCR), single strand conformation polymorphism (SSCP) analysis, and direct DNA sequencing, different mutations have been identified in 6 patients. All the mutations will interfere with the correct translation of the mRNA predicting a truncated protein or no gene product at all.. 8477262 38 51 choroideremia Modifier D015794 8477262 100 113 choroideremia Modifier D015794 8477262 120 123 CHM Modifier D015794 8477262 170 173 CHM SpecificDisease D015794 10543403|t|G130V, a common FRDA point mutation, appears to have arisen from a common founder. 10543403|a|Friedreich ataxia (FRDA) is the most common inherited ataxia. About 98% of mutant alleles have an expansion of a GAA trinucleotide repeat in intron 1 of the affected gene, FRDA. The other 2% are point mutations. Of the 17 point mutations so far described, three appear to be more common. One of these is the G130V mutation in exon 4 of FRDA. G130V, when present with an expanded GAA repeat on the other allele, is associated with an atypical FRDA phenotype. Haplotype analysis was undertaken on the four families who have been described with this mutation. The results suggest a common founder for this mutation. Although marked differences in extragenic marker haplotypes were seen in one family, similar intragenic haplotyping suggests the same mutation founder for this family with the differences explicable by two recombination events.. 10543403 16 20 FRDA Modifier D005621 10543403 83 100 Friedreich ataxia SpecificDisease D005621 10543403 102 106 FRDA SpecificDisease D005621 10543403 127 143 inherited ataxia DiseaseClass D013132 10543403 525 529 FRDA Modifier D005621 7605382|t|Frequency of exon 15 missense mutation (442D:G) in cholesteryl ester transfer protein gene in hyperalphalipoproteinemic Japanese subjects. 7605382|a|Cholesteryl ester transfer protein (CETP) transfers cholesteryl ester from high density lipoprotein (HDL) to apo B-containing lipoproteins. The hyperalphalipoproteinemia caused by CETP deficiency is fairly common in Japan and one of the most common mutations in the CETP gene is the splicing defect of the intron 14, the allelic frequency of which has been shown to be 0. 0049 in the Japanese general population. Recently, we have reported a missense mutation in exon 15 of the CETP gene (442D G), showing a dominant effect on the CETP activity and HDL-cholesterol level. In the current study, we determined the frequency of this new mutation in Japanese hyperalphalipoproteinemic (HDL-cholesterol > or = 100 mg/dl) subjects. A rapid and easy screening method for this new mutation was developed using a polymerase chain reaction (PCR) -mediated site-directed mutagenesis. Among 117 Japanese hyperalphalipoproteinemic subjects (HDL-cholesterol; 116. 7 +/- 16. 5 mg/dl, mean +/- S. D.) without the intron 14 splice defect, three homozygotes (2. 5%) and 34 heterozygotes (29. 1%) were found to have the 442D G mutation. The relative allelic frequency of this mutation was calculated to be 0. 17. One of the homozygotes for the 442D G mutation was the patient previously described by us as having hyperalphalipoproteinemia with corneal opacity and coronary heart disease. This was the first reported subject homozygous for the CETP deficiency who also demonstrated atherosclerotic symptoms. In homozygous subjects, CETP activity ranged from 37% to 62% of the normal value, which was consistent with the results obtained from the transient expression experiment previously reported; however, the specific activity of CETP was not as low as expected. (ABSTRACT TRUNCATED AT 250 WORDS) 7605382 94 119 hyperalphalipoproteinemic Modifier OMIM:143470 7605382 283 308 hyperalphalipoproteinemia SpecificDisease OMIM:143470 7605382 319 334 CETP deficiency SpecificDisease OMIM:143470 7605382 795 820 hyperalphalipoproteinemic Modifier OMIM:143470 7605382 1032 1057 hyperalphalipoproteinemic Modifier OMIM:143470 7605382 1436 1461 hyperalphalipoproteinemia SpecificDisease OMIM:143470 7605382 1467 1482 corneal opacity SpecificDisease D003318 7605382 1487 1509 coronary heart disease SpecificDisease D003327 7605382 1566 1581 CETP deficiency SpecificDisease OMIM:143470 7605382 1604 1628 atherosclerotic symptoms DiseaseClass D050197 10071193|t|Fibroblast growth factor homologous factor 2 (FHF2): gene structure, expression and mapping to the Borjeson-Forssman-Lehmann syndrome region in Xq26 delineated by a duplication breakpoint in a BFLS-like patient. 10071193|a|Borjeson-Forssman-Lehmann syndrome (BFLS) is a syndromal X-linked mental retardation, which maps by linkage to the q26 region of the human X chromosome. We have identified a male patient with BFLS-like features and a duplication, 46, Y, dup (X) (q26q28), inherited from his phenotypically normal mother. Fluorescence in situ hybridisation using yeast artificial chromosome clones from Xq26 localised the duplication breakpoint to an approximately 400-kb interval in the Xq26. 3 region between DXS155 and DXS294/DXS730. Database searches and analysis of available genomic DNA sequence from the region revealed the presence of the fibroblast growth factor homologous factor gene, FHF2, within the duplication breakpoint interval. The gene structure of FHF2 was determined and two new exons were identified, including a new 5 end exon, 1B. FHF2 is a large gene extending over approximately 200 kb in Xq26. 3 and is composed of at least seven exons. It shows tissue-specific alternative splicing and alternative transcription starts. Northern blot hybridisation showed highest expression in brain and skeletal muscle. The FHF2 gene localisation and tissue-specific expression pattern suggest it to be a candidate gene for familial cases of the BFLS syndrome and other syndromal and non-specific forms of X-linked mental retardation mapping to the region. 10071193 99 133 Borjeson-Forssman-Lehmann syndrome Modifier C536575 10071193 193 197 BFLS Modifier C536575 10071193 212 246 Borjeson-Forssman-Lehmann syndrome SpecificDisease C536575 10071193 248 252 BFLS SpecificDisease C536575 10071193 269 296 X-linked mental retardation DiseaseClass D038901 10071193 404 408 BFLS Modifier C536575 10071193 1452 1465 BFLS syndrome SpecificDisease C536575 10071193 1512 1539 X-linked mental retardation DiseaseClass D038901 8098245|t|Illegitimate transcription of the phenylalanine hydroxylase gene in lymphocytes for identification of mutations in phenylketonuria. 8098245|a|Taking advantage of the illegitimate transcription of the phenylalanine hydroxylase (PAH) gene, we have been able to analyse the PAH cDNA sequence of hyperphenylalaninemic children in circulating lymphocytes. Using this approach, we have also identified 3 novel mutations in cDNA from liver and lymphocytes of two patients. One mutation, detected by the abnormal pattern of migration of an amplified fragment, is a C to T transition in the splice acceptor site of intron 10, which resulted in the skipping of exon 11 with the premature termination of RNA translation downstream from exon 12 (-3 IVS10). The other two mutations are missense mutations in exons 10 and 11 (respectively, L333F and E390G). The present study supports the view that circulating lymphocytes give easy access to PAH gene transcripts whose nucleotide sequence is identical to that reported in liver and therefore represent a useful tool for molecular genetic studies in phenylketonuria.. 8098245 115 130 phenylketonuria SpecificDisease D010661 8098245 282 303 hyperphenylalaninemic Modifier D010661 8098245 1076 1091 phenylketonuria SpecificDisease D010661 10958786|t|Function of an axonal chemoattractant modulated by metalloprotease activity. 10958786|a|The axonal chemoattractant netrin-1 guides spinal commissural axons by activating its receptor DCC (Deleted in Colorectal Cancer ). We have found that chemical inhibitors of metalloproteases potentiate netrin-mediated axon outgrowth in vitro. We have also found that DCC is a substrate for metalloprotease-dependent ectodomain shedding, and that the inhibitors block proteolytic processing of DCC and cause an increase in DCC protein levels on axons within spinal cord explants. Thus, potentiation of netrin activity by inhibitors may result from stabilization of DCC on the axons, and proteolytic activity may regulate axon migration by controlling the number of functional extracellular axon guidance receptors.. 10958786 188 205 Colorectal Cancer SpecificDisease D015179 2886237|t|Choroideremia: close linkage to DXYS1 and DXYS12 demonstrated by segregation analysis and historical-genealogical evidence. 2886237|a|Linkage studies using restriction fragment length polymorphisms were conducted in the X-linked disorder, choroideremia, designated TCD for Progressive Tapeto-Choroidal Dystrophy. Previously demonstrated close linkage with locus DXYS1 was confirmed (lod 11. 44 at 0 recombination distance). In addition, locus DXYS12 was found to be closely linked with TCD (lod 3. 31 at 0 recombination distance). The disease mainly occurs in three large kindreds in remote Northern Finland. While formal genealogical proof is lacking, all presently living (more than 80 affected males and 120 carrier females) probably originate from a common founder couple born in 1644 and 1646, twelve generations ago. All 36 patients and 48 carriers tested from the three kindreds had the same haplotype (TCD/DXYS1, 11kb/DXYS12, 1. 6kb). Given that at least 105 female meioses transmitting TCD have occurred since 1650 in these kindreds, extremely close linkage between TCD, DXYS1 and DXYS12 is suggested. The above haplotype is a very useful diagnostic tool in these TCD families. We suggest that our historical-genealogical approach to linkage analysis may be possible elsewhere in similar isolated populations 2886237 0 13 Choroideremia SpecificDisease D015794 2886237 210 227 X-linked disorder DiseaseClass D040181 2886237 229 242 choroideremia SpecificDisease D015794 2886237 255 258 TCD SpecificDisease C531652 2886237 263 301 Progressive Tapeto-Choroidal Dystrophy SpecificDisease C531652 2886237 476 479 TCD SpecificDisease C531652 2886237 900 903 TCD SpecificDisease C531652 2886237 985 988 TCD SpecificDisease C531652 2886237 1065 1068 TCD SpecificDisease C531652 2886237 1163 1166 TCD Modifier C531652 10369870|t|Functional consequences of mutations in the early growth response 2 gene (EGR2) correlate with severity of human myelinopathies. 10369870|a|The early growth response 2 gene (EGR2) is a Cys2His2zinc finger transcription factor which is thought to play a role in the regulation of peripheral nervous system myelination. This idea is based partly on the phenotype of homozygous Krox20 (Egr2) knockout mice, which display hypomyelination of the PNS and a block of Schwann cells at an early stage of differentiation. Mutations in the human EGR2 gene have recently been associated with the inherited peripheral neuropathies Charcot-Marie-Tooth type 1, Dejerine-Sottas syndrome and congenital hypomyelinating neuropathy. Three of the four EGR2 mutations are dominant and occur within the zinc finger DNA-binding domain. The fourth mutation is recessive and affects the inhibitory domain (R1) that binds the NAB transcriptional co-repressors. A combination of DNA-binding assays and transcriptional analysis was used to determine the functional consequences of these mutations. The zinc finger mutations affect DNA binding and the amount of residual binding directly correlates with disease severity. The R1 domain mutation prevents interaction of EGR2 with the NAB co-repressors and thereby increases transcriptional activity. These data provide insight into the possible disease mechanisms underlying EGR2 mutations and the reason for varying severity and differences in inheritance patterns.. 10369870 113 127 myelinopathies DiseaseClass D011115 10369870 407 433 hypomyelination of the PNS DiseaseClass D010523 10369870 573 606 inherited peripheral neuropathies DiseaseClass C548028 10369870 607 633 Charcot-Marie-Tooth type 1 SpecificDisease D002607 10369870 635 659 Dejerine-Sottas syndrome SpecificDisease C538392 10369870 664 701 congenital hypomyelinating neuropathy SpecificDisease OMIM:605253 2786201|t|Molecular basis of human von Willebrand disease: analysis of platelet von Willebrand factor mRNA. 2786201|a|von Willebrand disease (vWD), the most common inherited bleeding disorder in humans, can result from either a quantitative or a qualitative defect in the adhesive glycoprotein, von Willebrand factor (vWF). Molecular studies of vWD have been limited by the large size of the vWF gene and difficulty in obtaining the vWF mRNA from patients. By use of an adaptation of the polymerase chain reaction, vWF mRNA was amplified and sequenced from peripheral blood platelets. A silent vWF allele was identified, resulting from a cis defect in vWF mRNA transcription or processing. In two type IIA vWD patients, two different but adjacent missense mutations were identified, the locations of which may identify an important vWF functional domain. Expression in heterologous cells of recombinant vWF containing one of these latter mutations reproduced the characteristic structural abnormality seen in type IIA vWD plasma.. 2786201 25 47 von Willebrand disease SpecificDisease D014842 2786201 70 84 von Willebrand Modifier D014842 2786201 98 120 von Willebrand disease SpecificDisease D014842 2786201 122 125 vWD SpecificDisease D014842 2786201 144 171 inherited bleeding disorder DiseaseClass D025861 2786201 275 289 von Willebrand Modifier D014842 2786201 325 328 vWD SpecificDisease D014842 2786201 677 689 type IIA vWD Modifier D056728 2786201 989 1001 type IIA vWD Modifier D056728 10430930|t|A transgene insertion creating a heritable chromosome deletion mouse model of Prader-Willi and angelman syndromes. 10430930|a|Prader-Willi syndrome (PWS) and Angelman syndrome (AS) result from the loss of function of imprinted genes in human chromosome 15q11-q13. The central part of mouse chromosome 7 is homologous to human 15q11-q13, with conservation of both gene order and imprinted features. We report here the characterization of a transgene insertion (Epstein-Barr virus Latent Membrane Protein 2A, LMP2A) into mouse chromosome 7C, which has resulted in mouse models for PWS and AS dependent on the sex of the transmitting parent. Epigenotype (allelic expression and DNA methylation) and fluorescence in situ hybridization analyses indicate that the transgene-induced mutation has generated a complete deletion of the PWS/AS-homologous region but has not deleted flanking loci. Because the intact chromosome 7, opposite the deleted homolog, maintains the correct imprint in somatic cells of PWS and AS mice and establishes the correct imprint in male and female germ cells of AS mice, homologous association and replication asynchrony are not part of the imprinting mechanism. This heritable-deletion mouse model will be particularly useful for the identification of the etiological genes and mechanisms, phenotypic basis, and investigation of therapeutic approaches for PWS.. 10430930 78 113 Prader-Willi and angelman syndromes CompositeMention D011218|D017204 10430930 115 136 Prader-Willi syndrome SpecificDisease D011218 10430930 138 141 PWS SpecificDisease D011218 10430930 147 164 Angelman syndrome SpecificDisease D017204 10430930 166 168 AS SpecificDisease D017204 10430930 568 571 PWS SpecificDisease D011218 10430930 576 578 AS SpecificDisease D017204 10430930 815 818 PWS Modifier D011218 10430930 819 821 AS Modifier D017204 10430930 988 991 PWS Modifier D011218 10430930 996 998 AS Modifier D017204 10430930 1073 1075 AS Modifier D017204 10430930 1368 1371 PWS SpecificDisease D011218 10747931|t|Transgenic mice expressing a truncated form of the high mobility group I-C protein develop adiposity and an abnormally high prevalence of lipomas. 10747931|a|Chromosomal translocations in human lipomas frequently create fusion transcripts encoding high mobility group (HMG) I-C DNA-binding domains and C-terminal sequences from different presumed transcription factors, suggesting a potential role for HMG I-C in the development of lipomas. To evaluate the role of the HMG I-C component, the three DNA-binding domains of HMG I-C have now been expressed in transgenic mice. Despite the ubiquitous expression of the truncated HMG I-C protein, the transgenic mice develop a selective abundance of fat tissue early in life, show marked adipose tissue inflammation, and have an abnormally high incidence of lipomas. These findings demonstrate that the DNA-binding domains of HMG I-C, in the absence of a C-terminal fusion partner, are sufficient to perturb adipogenesis and predispose to lipomas. We provide data supporting the central utility of this animal model as a tool to understand the molecular mechanisms underlying the development of one of the most common kind of human benign tumors.. 10747931 138 145 lipomas DiseaseClass D008067 10747931 183 190 lipomas DiseaseClass D008067 10747931 421 428 lipomas DiseaseClass D008067 10747931 721 748 adipose tissue inflammation SpecificDisease D007249 10747931 791 798 lipomas DiseaseClass D008067 10747931 972 979 lipomas DiseaseClass D008067 10747931 1165 1178 benign tumors DiseaseClass D009369 10051005|t|A common MSH2 mutation in English and North American HNPCC families: origin, phenotypic expression, and sex specific differences in colorectal cancer. 10051005|a|The frequency, origin, and phenotypic expression of a germline MSH2 gene mutation previously identified in seven kindreds with hereditary non-polyposis cancer syndrome (HNPCC) was investigated. The mutation (A-- > T at nt943 + 3) disrupts the 3 splice site of exon 5 leading to the deletion of this exon from MSH2 mRNA and represents the only frequent MSH2 mutation so far reported. Although this mutation was initially detected in four of 33 colorectal cancer families analysed from eastern England, more extensive analysis has reduced the frequency to four of 52 (8%) English HNPCC kindreds analysed. In contrast, the MSH2 mutation was identified in 10 of 20 (50%) separately identified colorectal families from Newfoundland. To investigate the origin of this mutation in colorectal cancer families from England (n = 4), Newfoundland (n = 10), and the United States (n = 3), haplotype analysis using microsatellite markers linked to MSH2 was performed. Within the English and US families there was little evidence for a recent common origin of the MSH2 splice site mutation in most families. In contrast, a common haplotype was identified at the two flanking markers (CA5 and D2S288) in eight of the Newfoundland families. These findings suggested a founder effect within Newfoundland similar to that reported by others for two MLH1 mutations in Finnish HNPCC families. We calculated age related risks of all, colorectal, endometrial, and ovarian cancers in nt943 + 3 A-- > T MSH2 mutation carriers (n = 76) for all patients and for men and women separately. For both sexes combined, the penetrances at age 60 years for all cancers and for colorectal cancer were 0. 86 and 0. 57, respectively. The risk of colorectal cancer was significantly higher (p < 0. 01) in males than females (0. 63 v 0. 30 and 0. 84 v 0. 44 at ages 50 and 60 years, respectively). For females there was a high risk of endometrial cancer (0. 5 at age 60 years) and premenopausal ovarian cancer (0. 2 at 50 years). These intersex differences in colorectal cancer risks have implications for screening programmes and for attempts to identify colorectal cancer susceptibility modifiers. 10051005 53 58 HNPCC Modifier D003123 10051005 132 149 colorectal cancer SpecificDisease D015179 10051005 278 318 hereditary non-polyposis cancer syndrome SpecificDisease D003123 10051005 320 325 HNPCC SpecificDisease D003123 10051005 594 611 colorectal cancer Modifier D015179 10051005 729 734 HNPCC Modifier D003123 10051005 840 850 colorectal Modifier D015179 10051005 925 942 colorectal cancer Modifier D015179 10051005 1507 1512 HNPCC Modifier D003123 10051005 1563 1607 colorectal, endometrial, and ovarian cancers CompositeMention D010051|D016889|D015179 10051005 1777 1784 cancers DiseaseClass D009369 10051005 1794 1811 colorectal cancer SpecificDisease D015179 10051005 1860 1877 colorectal cancer SpecificDisease D015179 10051005 2047 2065 endometrial cancer SpecificDisease D016889 10051005 2093 2121 premenopausal ovarian cancer SpecificDisease D010051 10051005 2172 2189 colorectal cancer Modifier D015179 10051005 2268 2285 colorectal cancer Modifier D015179 7814011|t|Novel mutation at the initiation codon in the Norrie disease gene in two Japanese families. 7814011|a|We have identified a new mutation of Norrie disease (ND) gene in two Japanese males from unrelated families; they showed typical ocular features of ND but no mental retardation or hearing impairment. A mutation was found in both patients at the initiation codon of exon 2 of the ND gene (ATG to GTG), with otherwise normal nucleotide sequences. Their mothers had the normal and mutant types of the gene, which was expected for heterozygotes of the disease. The mutation of the initiation codon would cause the failure of ND gene expression or a defect in translation thereby truncating the amino terminus of ND protein. In view of the rarity and marked heterogeneity of mutations in the ND gene, the present apparently unrelated Japanese families who have lived in the same area for over two centuries presumably share the origin of the mutation.. 7814011 46 60 Norrie disease Modifier C537849 7814011 129 143 Norrie disease Modifier C537849 7814011 145 147 ND Modifier C537849 7814011 240 242 ND SpecificDisease C537849 7814011 250 268 mental retardation DiseaseClass D008607 7814011 272 290 hearing impairment DiseaseClass D034381 7814011 371 373 ND Modifier C537849 7814011 613 615 ND Modifier C537849 7814011 700 702 ND Modifier C537849 7814011 779 781 ND Modifier C537849 8571953|t|Haplotype and phenotype analysis of six recurrent BRCA1 mutations in 61 families: results of an international study. 8571953|a|Several BRCA1 mutations have now been found to occur in geographically diverse breast and ovarian cancer families. To investigate mutation origin and mutation-specific phenotypes due to BRCA1, we constructed a haplotype of nine polymorphic markers within or immediately flanking the BRCA1 locus in a set of 61 breast/ovarian cancer families selected for having one of six recurrent BRCA1 mutations. Tests of both mutations and family-specific differences in age at diagnosis were not significant. A comparison of the six mutations in the relative proportions of cases of breast and ovarian cancer was suggestive of an effect (P =. 069), with 57% of women presumed affected because of the 1294 del 40 BRCA1 mutation having ovarian cancer, compared with 14% of affected women with the splice-site mutation in intron 5 of BRCA1. For the BRCA1 mutations studied here, the individual mutations are estimated to have arisen 9-170 generations ago. In general, a high degree of haplotype conservation across the region was observed, with haplotype differences most often due to mutations in the short-tandem-repeat markers, although some likely instances of recombination also were observed. For several of the instances, there was evidence for multiple, independent, BRCA1 mutational events. 8571953 196 221 breast and ovarian cancer Modifier D061325 8571953 427 448 breast/ovarian cancer Modifier D061325 8571953 688 713 breast and ovarian cancer CompositeMention D061325 8571953 839 853 ovarian cancer SpecificDisease D010051 7166314|t|A new glucose-6-phosphate dehydrogenase variant (G6PD Nagano) associated with congenital hemolytic anemia. 7166314|a|A new glucose-6-phosphate dehydrogenase (G6PD) variant associated with chronic nonspherocytic hemolytic anemia was reported. The patient, a 6-year-old Japanese male, was noticed to have hemolytic anemia soon after birth, and a diagnosis of G6PD deficiency was made at the age of 2. He had episodes of hemolytic crisis several times after upper respiratory infection. G6PD activity of the patient was 5. 5% of normal. The enzymatic characteristics were examined when he was 5 years old, and his G6PD showed faster-than-normal electrophoretic mobility, low Km G6P, high Km NADP, low Ki NADPH, normal utilization of substrate analogues, heat instability, and a normal pH optimum curve. From these results, this was considered to be a new variant and was designated G6PD Nagano. Infection-induced hemolysis and chronic hemolytic anemia seem to be due to markedly impaired enzyme activity and thermal instability. 7166314 78 105 congenital hemolytic anemia SpecificDisease D000745 7166314 178 217 chronic nonspherocytic hemolytic anemia SpecificDisease D000746 7166314 293 309 hemolytic anemia SpecificDisease D000743 7166314 347 362 G6PD deficiency SpecificDisease D005955 7166314 408 424 hemolytic crisis DiseaseClass D006461 7166314 445 472 upper respiratory infection SpecificDisease D012141 7166314 900 909 hemolysis SpecificDisease D006461 7166314 914 938 chronic hemolytic anemia SpecificDisease D000745 1468459|t|Craniofrontonasal dysplasia. 1468459|a|We report on nine patients with craniofrontonasal dysplasia (CFND). Seven classical cases had facial features suggestive of frontonasal dysplasia and coronal craniosynostosis. Extracranial abnormalities such as brittle nails with prominent longitudinal grooves or syndactyly of fingers and toes were observed in individual patients. In two families the father of classical cases showed a milder pattern of abnormalities, consistent with the diagnosis. We present a 2- to 13-year follow-up on our patients. Hypotonia and laxity of joints are common and may necessitate supportive measures. Mild developmental delay was noted in three out of six classical cases studied in detail. Unlike almost all other X-linked disorders, clinical expression in CFND is generally much more severe in females than in males. In contrast to previous reports of this condition, one of our severely affected cases is a male.. 1468459 0 27 Craniofrontonasal dysplasia SpecificDisease C536456 1468459 61 88 craniofrontonasal dysplasia SpecificDisease C536456 1468459 90 94 CFND SpecificDisease C536456 1468459 153 174 frontonasal dysplasia SpecificDisease C538065 1468459 179 203 coronal craniosynostosis SpecificDisease C537369 1468459 205 231 Extracranial abnormalities DiseaseClass D009139 1468459 240 289 brittle nails with prominent longitudinal grooves SpecificDisease D009260 1468459 293 323 syndactyly of fingers and toes CompositeMention D013576 1468459 535 544 Hypotonia SpecificDisease D009123 1468459 549 565 laxity of joints SpecificDisease C535884 1468459 623 642 developmental delay DiseaseClass D002658 1468459 732 750 X-linked disorders DiseaseClass D040181 1468459 775 779 CFND SpecificDisease C536456 1707231|t|A new mutation in the proteolipid protein (PLP) gene in a German family with Pelizaeus-Merzbacher disease. 1707231|a|A C-to-T transition in exon 4 of the PLP gene was found in 2 affected males and two obligate carriers in a German family with Pelizaeus-Merzbacher disease. The mutation, which causes loss of an HphI site and changes amino acid 155 from threonine to isoleucine, was absent from 108 normal chromosomes. There are 5 concordances and 1 discrepancy between these results and those obtained by magnetic resonance imaging in this family.. 1707231 77 105 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 1707231 233 261 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 7579347|t|The Wiskott-Aldrich syndrome and X-linked congenital thrombocytopenia are caused by mutations of the same gene. 7579347|a|The Wiskott-Aldrich syndrome (WAS) is an X-linked recessive disorder characterized by thrombocytopenia, small platelets, eczema, recurrent infections, and immunodeficiency. Besides the classic WAS phenotype, there is a group of patients with congenital X-linked thrombocytopenia (XLT) who have small platelets but only transient eczema, if any, and minimal immune deficiency. Because the gene responsible for WAS has been sequenced, it was possible to correlate the WAS phenotypes with WAS gene mutations. Using a fingerprinting screening technique, we determined the approximate location of the mutation in 13 unrelated WAS patients with mild to severe clinical symptoms. Direct sequence analysis of cDNA and genomic DNA obtained from patient-derived cell lines showed 12 unique mutations distributed throughout the WAS gene, including insertions, deletions, and point mutations resulting in amino acid substitutions, termination, exon skipping, or splicing defects. Of 4 unrelated patients with the XLT phenotype, 3 had missense mutations affecting exon 2 and 1 had a splice-site mutation affecting exon 9. Patients with classic WAS had more complex mutations, resulting in termination codons, frameshift, and early termination. These findings provide direct evidence that XLT and WAS are caused by mutations of the same gene and suggest that severe clinical phenotypes are associated with complex mutations.. 7579347 4 28 Wiskott-Aldrich syndrome SpecificDisease D014923 7579347 33 69 X-linked congenital thrombocytopenia SpecificDisease OMIM:313900 7579347 116 140 Wiskott-Aldrich syndrome SpecificDisease D014923 7579347 142 145 WAS SpecificDisease D014923 7579347 153 180 X-linked recessive disorder DiseaseClass D040181 7579347 198 214 thrombocytopenia SpecificDisease D013921 7579347 233 239 eczema SpecificDisease D004485 7579347 267 283 immunodeficiency DiseaseClass D007153 7579347 305 308 WAS Modifier D014923 7579347 354 390 congenital X-linked thrombocytopenia SpecificDisease OMIM:313900 7579347 392 395 XLT SpecificDisease OMIM:313900 7579347 441 447 eczema SpecificDisease D004485 7579347 469 486 immune deficiency DiseaseClass D007153 7579347 521 524 WAS SpecificDisease D014923 7579347 578 581 WAS Modifier D014923 7579347 598 601 WAS Modifier D014923 7579347 733 736 WAS Modifier D014923 7579347 929 932 WAS Modifier D014923 7579347 1113 1116 XLT Modifier OMIM:313900 7579347 1243 1246 WAS SpecificDisease D014923 7579347 1387 1390 XLT SpecificDisease OMIM:313900 7579347 1395 1398 WAS SpecificDisease D014923 1709636|t|A 3-base pair in-frame deletion of the phenylalanine hydroxylase gene results in a kinetic variant of phenylketonuria. 1709636|a|Phenylketonuria (PKU) is an autosomal recessive disease due to deficiency of a hepatic enzyme, phenylalanine hydroxylase (PAH). The absence of PAH activity results in typical PKU while persistence of a residual enzyme activity gives rise to variant forms of the disease. We report here a 3-base pair in-frame deletion of the PAH gene (delta 194) in a mild variant, with markedly reduced affinity of the enzyme for phenylalanine (Km = 160 nM), and we provide functional evidence for responsibility of the deletion in the mutant phenotype. Since the deletion was located in the third exon of the gene, which presents no homology with other hydroxylases, we suggest that exon 3 is involved in the specificity of the enzyme for phenylalanine. Finally, since none of the 98 PKU patients tested were found to carry this particular deletion, our study suggests that this molecular event probably occurred recently on the background of a haplotype 2 gene in Portugal.. 1709636 102 117 phenylketonuria SpecificDisease D010661 1709636 119 134 Phenylketonuria SpecificDisease D010661 1709636 136 139 PKU SpecificDisease D010661 1709636 147 174 autosomal recessive disease DiseaseClass D030342 1709636 182 239 deficiency of a hepatic enzyme, phenylalanine hydroxylase SpecificDisease OMIM:261600 1709636 294 297 PKU SpecificDisease D010661 1709636 888 891 PKU Modifier D010661 10633128|t|Friedreich ataxia: an overview. 10633128|a|Friedreich ataxia, an autosomal recessive neurodegenerative disease, is the most common of the inherited ataxias. The recent discovery of the gene that is mutated in this condition, FRDA, has led to rapid advances in the understanding of the pathogenesis of Friedreich ataxia. About 98% of mutant alleles have an expansion of a GAA trinucleotide repeat in intron 1 of the gene. This leads to reduced levels of the protein, frataxin. There is mounting evidence to suggest that Friedreich ataxia is the result of accumulation of iron in mitochondria leading to excess production of free radicals, which then results in cellular damage and death. Currently there is no known treatment that alters the natural course of the disease. The discovery of the FRDA gene and its possible function has raised hope that rational therapeutic strategies will be developed.. 10633128 0 17 Friedreich ataxia SpecificDisease D005621 10633128 32 49 Friedreich ataxia SpecificDisease D005621 10633128 54 99 autosomal recessive neurodegenerative disease DiseaseClass D020271 10633128 127 144 inherited ataxias SpecificDisease D013132 10633128 290 307 Friedreich ataxia SpecificDisease D005621 10633128 508 525 Friedreich ataxia SpecificDisease D005621 10633128 782 786 FRDA Modifier D005621 10737119|t|Clinical and molecular genetics of primary dystonias. 10737119|a|Primary dystonias are movement disorders with dystonia as a major symptom. They are frequently inherited as Mendelian traits. There are at least eight clinically distinct autosomal dominant and two X-linked recessive forms. In addition, pedigree analyses suggest the occurrence of an autosomal recessive variant. The clinical classification is increasingly being replaced by a genetic one. To date gene loci have been identified in at least six autosomal dominant forms, i. e., in idiopathic torsion dystonia (9q34), focal dystonia (18p), adult-onset idiopathic torsion dystonia of mixed type (8p21-q22), dopa-responsive dystonia (14q22. 1-q22. 2), and paroxysmal dystonic choreoathetosis (2q25-q33; 1p21-p13. 3). Gene loci in the X-linked recessive forms have been assigned to Xq13. 1 in the X-linked dystonia parkinsonism syndrome and to Xq22 in X-linked sensorineural deafness, dystonia, and mental retardation. The disease genes have been identified in two autosomal dominant forms and in one X-linked recessive form. Mutations in a gene coding for an ATP-binding protein were detected in idiopathic torsion dystonia (DYT1), and the GTP cyclohydrolase 1 gene is mutated in dopa-responsive dystonia (DYT5). In sensorineural deafness, dystonia, and mental retardation , mutations were found in the gene DDP coding for a polypeptide of unknown function. This article reviews the clinical and molecular genetics of primary dystonias, critically discusses present findings, and proposes referring to the known forms, most of which can be distinguished by genetic criteria, as dystonias 1-12. 10737119 35 52 primary dystonias DiseaseClass D020821 10737119 54 71 Primary dystonias DiseaseClass D020821 10737119 76 94 movement disorders DiseaseClass D009069 10737119 100 108 dystonia DiseaseClass D004421 10737119 535 562 idiopathic torsion dystonia SpecificDisease D004422 10737119 571 585 focal dystonia SpecificDisease D020821 10737119 605 632 idiopathic torsion dystonia SpecificDisease D004422 10737119 659 683 dopa-responsive dystonia SpecificDisease C538007 10737119 707 742 paroxysmal dystonic choreoathetosis SpecificDisease C537181 10737119 848 887 X-linked dystonia parkinsonism syndrome SpecificDisease OMIM:314250 10737119 903 934 X-linked sensorineural deafness DiseaseClass OMIM:304500 10737119 936 944 dystonia DiseaseClass D004421 10737119 950 968 mental retardation DiseaseClass D008607 10737119 1148 1175 idiopathic torsion dystonia SpecificDisease D004422 10737119 1232 1256 dopa-responsive dystonia SpecificDisease C538007 10737119 1268 1290 sensorineural deafness DiseaseClass D006319 10737119 1292 1300 dystonia DiseaseClass D004421 10737119 1306 1324 mental retardation DiseaseClass D008607 10737119 1470 1487 primary dystonias DiseaseClass D020821 10737119 1630 1644 dystonias 1-12 CompositeMention D020821 10533031|t|Clinical and genetic study of Friedreich ataxia in an Australian population. 10533031|a|Friedreich ataxia is an autosomal recessive disorder caused by mutations in the FRDA gene that encodes a 210-amino acid protein called frataxin. An expansion of a GAA trinucleotide repeat in intron 1 of the gene is present in more than 95% of mutant alleles. Of the 83 people we studied who have mutations in FRDA, 78 are homozygous for an expanded GAA repeat; the other five patients have an expansion in one allele and a point mutation in the other. Here we present a detailed clinical and genetic study of a subset of 51 patients homozygous for an expansion of the GAA repeat. We found a correlation between the size of the smaller of the two expanded alleles and age at onset, age into wheelchair, scoliosis, impaired vibration sense, and the presence of foot deformity. There was no significant correlation between the size of the smaller allele and cardiomyopathy, diabetes mellitus, loss of proprioception, or bladder symptoms. The larger allele size correlated with bladder symptoms and the presence of foot deformity. The duration of disease is correlated with wheelchair use and the presence of diabetes, scoliosis, bladder symptoms and impaired proprioception, and vibration sense but no other complications studied.. 10533031 30 47 Friedreich ataxia SpecificDisease D005621 10533031 77 94 Friedreich ataxia SpecificDisease D005621 10533031 101 129 autosomal recessive disorder DiseaseClass D030342 10533031 157 161 FRDA Modifier D005621 10533031 779 788 scoliosis DiseaseClass D012600 10533031 836 850 foot deformity DiseaseClass D005530 10533031 932 946 cardiomyopathy SpecificDisease D009202 10533031 948 965 diabetes mellitus SpecificDisease D003920 10533031 967 989 loss of proprioception DiseaseClass D020886 10533031 994 1010 bladder symptoms DiseaseClass D001745 10533031 1051 1067 bladder symptoms DiseaseClass D001745 10533031 1088 1102 foot deformity DiseaseClass D005530 10533031 1182 1190 diabetes DiseaseClass D003920 10533031 1192 1201 scoliosis DiseaseClass D012600 10533031 1203 1219 bladder symptoms DiseaseClass D001745 10533031 1224 1247 impaired proprioception DiseaseClass D020886 492812|t|Recurrent meningococcal meningitis with absence of the sixth component of complement: an evaluation of underlying immunologic mechanisms. 492812|a|A 51/2-year-old black girl with recurrent meningococcal meningitis and absence of the sixth component of complement (C6) is reported. To explore the pathogenesis of recurrent neisserial infections in C6 deficiency, a detailed analysis of her immune competence was conducted. Her serum had normal chemotactic, opsonic, alternative complement pathway, and specific antibody activity, but lacked complement-mediated bacteriolytic activity. In addition, her C6-deficient serum was indistinguishable from normal serum in a complement-dependent assay of phagocyte bactericidal activity. Absent bacteriolysis remains the only consistent defect associated with recurrent neisserial infections and absence of one of the late-acting complement components.. 492812 10 34 meningococcal meningitis SpecificDisease D008585 492812 40 84 absence of the sixth component of complement SpecificDisease OMIM:612446 492812 180 204 meningococcal meningitis SpecificDisease D008585 492812 209 253 absence of the sixth component of complement SpecificDisease OMIM:612446 492812 313 334 neisserial infections SpecificDisease D016870 492812 338 351 C6 deficiency SpecificDisease OMIM:612446 492812 592 604 C6-deficient Modifier OMIM:612446 492812 801 822 neisserial infections SpecificDisease D016870 10519880|t|Mutation of the sterol 27-hydroxylase gene (CYP27) results in truncation of mRNA expressed in leucocytes in a Japanese family with cerebrotendinous xanthomatosis. 10519880|a|OBJECTIVES A Japanese family with cerebrotendinous xanthomatosis (CTX) was investigated for a sequence alteration in the sterol 27-hydroxylase gene (CYP27). The expression of CYP27 has been mostly explored using cultured fibroblasts, prompting the examination of the transcripts from blood leucocytes as a simple and rapid technique. METHODS An alteration in CYP27 of the proband was searched for by polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP) analysis and subsequent sequencing. Samples of RNA were subjected to reverse transcription PCR (RT-PCR) and the product of the proband was amplified with nested primers and sequenced. RESULTS A homozygous G to A transition at the 5 end of intron 7 was detected in the patient. In RT-PCR analysis, only a truncated transcript was detected in the patient, whereas both normal and truncated transcripts were detected in the siblings. The sequencing of the patients cDNA fragment disclosed a direct conjuction of exon 6 and exon 8. CONCLUSION The mutation at splice donor site and the truncation of mRNA were identical with those of a recently reported Italian patient, although different in symptomatology. The application of blood leucocytes can be a simple technique on analysing a constructive abnormality of CYP27 mRNA.. 10519880 131 161 cerebrotendinous xanthomatosis SpecificDisease D019294 10519880 198 228 cerebrotendinous xanthomatosis SpecificDisease D019294 10519880 230 233 CTX SpecificDisease D019294 10519880 1438 1458 abnormality of CYP27 SpecificDisease OMIM:213700 8187068|t|Frequent detection of codon 877 mutation in the androgen receptor gene in advanced prostate cancers. 8187068|a|Prostatic tissue specimens derived from transurethral resections of patients with metastatic prostate cancer were analyzed for genetic alterations in the hormone-binding domain of the androgen receptor (AR) gene. Direct sequencing of the polymerase chain reaction-derived DNAs of 6 of 24 specimens revealed a codon 877 mutation (ACT-- > GCT, Thr-- > Ala) in the hormone-binding domain of the AR gene. This same AR mutation has been reported previously in a metastatic prostate cancer cell line, LNCaP, where this mutation confers upon the AR an altered ligand-binding specificity which is stimulated by estrogens, progestagens, and antiandrogens. It is possible that analogous to an activated/altered growth factor receptor oncogene, codon 877 mutant AR with altered ligand binding may provide a selective growth advantage in the genesis of a subset of advanced prostate cancer. Although estrogens are used infrequently, antiandrogens are used increasingly in hormonal therapy for patients with advanced prostate cancer. The stimulatory effect of these therapeutic agents on the codon 877 mutant AR further suggests that this frequently observed AR mutation may contribute to the treatment refractory disease.. 8187068 74 99 advanced prostate cancers SpecificDisease D011471 8187068 183 209 metastatic prostate cancer SpecificDisease D011471 8187068 558 584 metastatic prostate cancer Modifier D011471 8187068 954 978 advanced prostate cancer SpecificDisease D011471 8187068 1096 1120 advanced prostate cancer SpecificDisease D011471 10802669|t|ATM-dependent phosphorylation of nibrin in response to radiation exposure. 10802669|a|Mutations in the gene ATM are responsible for the genetic disorder ataxia-telangiectasia (A-T), which is characterized by cerebellar dysfunction, radiosensitivity, chromosomal instability and cancer predisposition. Both the A-T phenotype and the similarity of the ATM protein to other DNA-damage sensors suggests a role for ATM in biochemical pathways involved in the recognition, signalling and repair of DNA double-strand breaks (DSBs). There are strong parallels between the pattern of radiosensitivity, chromosomal instability and cancer predisposition in A-T patients and that in patients with Nijmegen breakage syndrome (NBS). The protein defective in NBS, nibrin (encoded by NBS1), forms a complex with MRE11 and RAD50 (refs 1, 2). This complex localizes to DSBs within 30 minutes after cellular exposure to ionizing radiation (IR) and is observed in brightly staining nuclear foci after a longer period of time. The overlap between clinical and cellular phenotypes in A-T and NBS suggests that ATM and nibrin may function in the same biochemical pathway. Here we demonstrate that nibrin is phosphorylated within one hour of treatment of cells with IR. This response is abrogated in A-T cells that either do not express ATM protein or express near full-length mutant protein. We also show that ATM physically interacts with and phosphorylates nibrin on serine 343 both in vivo and in vitro. Phosphorylation of this site appears to be functionally important because mutated nibrin (S343A) does not completely complement radiosensitivity in NBS cells. ATM phosphorylation of nibrin does not affect nibrin-MRE11-RAD50 association as revealed by radiation-induced foci formation. Our data provide a biochemical explanation for the similarity in phenotype between A-T and NBS.. 10802669 125 141 genetic disorder DiseaseClass D030342 10802669 142 163 ataxia-telangiectasia SpecificDisease D001260 10802669 165 168 A-T SpecificDisease D001260 10802669 197 219 cerebellar dysfunction DiseaseClass D002526 10802669 267 273 cancer Modifier D009369 10802669 299 302 A-T Modifier D001260 10802669 610 616 cancer Modifier D009369 10802669 635 638 A-T SpecificDisease D001260 10802669 674 700 Nijmegen breakage syndrome SpecificDisease D049932 10802669 702 705 NBS SpecificDisease D049932 10802669 733 736 NBS SpecificDisease D049932 10802669 1051 1054 A-T SpecificDisease D001260 10802669 1059 1062 NBS SpecificDisease D049932 10802669 1265 1268 A-T Modifier D001260 10802669 1621 1624 NBS Modifier D049932 10802669 1841 1844 A-T SpecificDisease D001260 10802669 1849 1852 NBS SpecificDisease D049932 1346924|t|Detection of an unstable fragment of DNA specific to individuals with myotonic dystrophy. 1346924|a|Myotonic dystrophy (DM) is the most common form of adult muscular dystrophy, with a prevalence of 2-14 per 100, 000 individuals. The disease is characterized by progressive muscle weakness and sustained muscle contraction, often with a wide range of accompanying symptoms. The age at onset and severity of the disease show extreme variation, both within and between families. Despite its clinical variability, this dominant condition segregates as a single locus at chromosome 19q13. 3 in every population studied. It is flanked by the tightly linked genetic markers ERCC1 proximally and D19S51 distally; these define the DM critical region. We report the isolation of an expressed sequence from this region which detects a DNA fragment that is larger in affected individuals than in normal siblings or unaffected controls. The size of this fragment varies between affected siblings, and increases in size through generations in parallel with increasing severity of the disease. We postulate that this unstable DNA sequence is the molecular feature that underlies DM. 1346924 70 88 myotonic dystrophy SpecificDisease D009223 1346924 90 108 Myotonic dystrophy SpecificDisease D009223 1346924 110 112 DM SpecificDisease D009223 1346924 147 165 muscular dystrophy SpecificDisease D009136 1346924 263 278 muscle weakness DiseaseClass D018908 1346924 283 311 sustained muscle contraction DiseaseClass D009120 1346924 712 714 DM Modifier D009223 1346924 1154 1156 DM SpecificDisease D009223 10987655|t|Detection of a novel missense mutation and second recurrent mutation in the CACNA1A gene in individuals with EA-2 and FHM. 10987655|a|Mutations in the brain specific P/Q type Ca2 + channel alpha1 subunit gene, CACNA1A, have been identified in three clinically distinct disorders, viz. episodic ataxia type 2 (EA-2), familial hemiplegic migraine (FHM) and spinocerebellar ataxia 6 (SCA6). For individuals with EA-2, the mutations described thus far are presumed to result in a truncated protein product. Several different missense mutations have been identified in patients with FHM. At least two of these mutations have been identified on two different chromosome 19p13 haplotypes and thus represent recurrent mutations. In the present study, we have screened several individuals for mutations in all 47 exons in the CACNA1A gene by single-strand conformation analysis. We have characterised a novel missense mutation, G5260A, in exon 32 in a family segregating for EA-2. The consequence of this mutation is an amino acid substitution at a highly conserved position within the CACNA1A gene. This represents the first point mutation not resulting in a proposed truncated protein. Furthermore, this mutation has been detected in a family member with mild clinical signs including only migraine. Additionally, a second previously identified recurrent muta tion, C2272T, in exon 16 has been discovered in a patient with FHM.. 10987655 109 113 EA-2 SpecificDisease C535506 10987655 118 121 FHM SpecificDisease D020325 10987655 274 296 episodic ataxia type 2 SpecificDisease C535506 10987655 298 302 EA-2 SpecificDisease C535506 10987655 305 333 familial hemiplegic migraine SpecificDisease D020325 10987655 335 338 FHM SpecificDisease D020325 10987655 344 368 spinocerebellar ataxia 6 SpecificDisease OMIM:183086 10987655 370 374 SCA6 SpecificDisease OMIM:183086 10987655 398 402 EA-2 SpecificDisease C535506 10987655 567 570 FHM SpecificDisease D020325 10987655 955 959 EA-2 SpecificDisease C535506 10987655 1272 1280 migraine SpecificDisease D008881 10987655 1405 1408 FHM SpecificDisease D020325 10213492|t|Dominant negative effect of the APC1309 mutation: a possible explanation for genotype-phenotype correlations in familial adenomatous polyposis. 10213492|a|Inactivation of the adenomatous polyposis coli (APC) gene product initiates colorectal tumorigenesis. Patients with familial APC (FAP) carry germ-line mutations in the APC gene and develop multiple colorectal adenomas and subsequent carcinomas early in life. The severity of the disease correlates with the position of the inherited APC mutation (genotype-phenotype correlation). Together with the fact that both germ-line and sporadic APC mutations cluster in the central region of the APC gene, this points to a dominant negative effect of certain APC mutants. Loss of APC function was recently shown to result in enhanced beta-catenin-/Tcf-mediated transcription in colon epithelial cells. Here, we provide experimental evidence for a dominant negative effect of APC gene products associated with severe polyposis. Wild-type APC activity in beta-catenin-/Tcf-mediated transcription was strongly inhibited by a mutant APC that is truncated at codon 1309. In contrast, mutant APC gene products that are associated with attenuated polyposis (codon 386 or 1465) interfered only weakly with wild-type APC activity. These results suggest a molecular explanation for the genotype-phenotype correlation in FAP patients and support the idea that colorectal tumor growth might be, in part, driven by selection for a mutation in the mutation cluster region.. 10213492 112 142 familial adenomatous polyposis SpecificDisease D011125 10213492 164 190 adenomatous polyposis coli Modifier D011125 10213492 192 195 APC Modifier D011125 10213492 260 272 familial APC SpecificDisease D011125 10213492 274 277 FAP SpecificDisease D011125 10213492 312 315 APC Modifier D011125 10213492 342 361 colorectal adenomas SpecificDisease D003123 10213492 377 387 carcinomas DiseaseClass D009369 10213492 477 480 APC Modifier D011125 10213492 580 583 APC Modifier D011125 10213492 631 634 APC Modifier D011125 10213492 694 697 APC Modifier D011125 10213492 715 718 APC Modifier D011125 10213492 910 913 APC Modifier D011125 10213492 951 960 polyposis SpecificDisease D044483 10213492 972 975 APC Modifier D011125 10213492 1064 1067 APC Modifier D011125 10213492 1121 1124 APC Modifier D011125 10213492 1164 1184 attenuated polyposis SpecificDisease C538265 10213492 1243 1246 APC Modifier D011125 10213492 1345 1348 FAP Modifier D011125 10213492 1384 1400 colorectal tumor Modifier D015179 1717985|t|Mutation of the KIT (mast/stem cell growth factor receptor) protooncogene in human piebaldism. 1717985|a|Piebaldism is an autosomal dominant genetic disorder characterized by cogenital patches of skin and hair from which melanocytes are completely absent. A similar disorder of mouse, dominant white spotting (W), results from mutations of the c-Kit protooncogene, which encodes and receptor for mast/stem cell growth factor. We identified a KIT gene mutation in a proband with classic autosomal dominant piebaldism. This mutation results in a Gly----Arg substitution at codon 664, within the tyrosine kinase domain. This substitution was not seen in any normal individuals and was completely linked to the piebald phenotype in the probands family. Piebaldism in this family thus appears to be the human homologue to dominant white spotting (W) of the mouse.. 1717985 83 93 piebaldism SpecificDisease D016116 1717985 95 105 Piebaldism SpecificDisease D016116 1717985 112 147 autosomal dominant genetic disorder DiseaseClass D030342 1717985 495 505 piebaldism SpecificDisease D016116 1717985 697 704 piebald Modifier D016116 1717985 739 749 Piebaldism SpecificDisease D016116 8104633|t|An arylsulfatase A (ARSA) missense mutation (T274M) causing late-infantile metachromatic leukodystrophy. 8104633|a|Metachromatic leukodystrophy (MLD) is an autosomal recessive lysosomal storage disorder caused by a deficiency of arylsulfatase A (ARSA; EC 3. 1. 6. 8). The 8 ARSA exons and adjacent intron boundaries from a patient with late-infantile metachromatic leukodystrophy were polymerase chain reaction (PCR) amplified in seven discrete reactions. Amplified ARSA exons were analysed for the presence of sequence alterations by single-strand conformation polymorphism analysis, followed by direct sequencing of PCR products. The patient was found to be homozygous for a C-- > T transition in exon IV that results in the substitution of a highly conserved threonine residue at amino acid 274 with a methionine (T274M). Analysis of a further 29 MLD patients revealed the presence of five additional homozygotes for T274M. All 6 T274M homozygotes (representing four families) were of Lebanese descent, and all were known to be the result of consanguineous marriages. The altered amino acid is rigidly conserved among 10 sulfatases from Escherichia coli to humans; therefore, it is most likely that the resultant mutant protein will have little or no enzyme activity. This is consistent with the very low ARSA activity measured in these patients and their uniformly severe clinical presentation 8104633 60 103 late-infantile metachromatic leukodystrophy SpecificDisease D007966 8104633 105 133 Metachromatic leukodystrophy SpecificDisease D007966 8104633 135 138 MLD SpecificDisease D007966 8104633 146 192 autosomal recessive lysosomal storage disorder DiseaseClass D016464 8104633 205 234 deficiency of arylsulfatase A SpecificDisease D007966 8104633 326 369 late-infantile metachromatic leukodystrophy SpecificDisease D007966 8104633 840 843 MLD Modifier D007966 7717396|t|Mutations in the gene for X-linked adrenoleukodystrophy in patients with different clinical phenotypes. 7717396|a|Recently, the gene for the most common peroxisomal disorder, X-linked adrenoleukodystrophy (X-ALD), has been described encoding a peroxisomal membrane transporter protein. We analyzed the entire protein-coding sequence of this gene by reverse-transcription PCR, SSCP, and DNA sequencing in five patients with different clinical expression of X-ALD and in their female relatives; these clinical expressions were cerebral childhood ALD, adrenomyeloneuropathy (AMN), and " Addison disease only " (ADO) phenotype. In the three patients exhibiting the classical picture of severe childhood ALD we identified in the 5 portion of the X-ALD gene a 38-bp deletion that causes a frameshift mutation, a 3-bp deletion leading to a deletion of an amino acid in the ATP-binding domain of the ALD protein, and a missense mutation. In the patient with the clinical phenotype of AMN, a nonsense mutation in codon 212, along with a second site mutation at codon 178, was observed. Analysis of the patient with the ADO phenotype revealed a further missense mutation at a highly conserved position in the ALDP/PMP70 comparison. The disruptive nature of two mutations (i. e., the frameshift and the nonsense mutation) in patients with biochemically proved childhood ALD and AMN further strongly supports the hypothesis that alterations in this gene play a crucial role in the pathogenesis of X-ALD. Since the current biochemical techniques for X-ALD carrier detection in affected families lack sufficient reliability, our procedure described for systematic mutation scanning is also capable of improving genetic counseling and prenatal diagnosis 7717396 26 55 X-linked adrenoleukodystrophy SpecificDisease D000326 7717396 143 163 peroxisomal disorder DiseaseClass D018901 7717396 165 194 X-linked adrenoleukodystrophy SpecificDisease D000326 7717396 196 201 X-ALD SpecificDisease D000326 7717396 446 451 X-ALD SpecificDisease D000326 7717396 534 537 ALD SpecificDisease D000326 7717396 539 560 adrenomyeloneuropathy SpecificDisease D000326 7717396 562 565 AMN SpecificDisease D000326 7717396 574 594 Addison disease only Modifier D000224 7717396 598 601 ADO Modifier D000224 7717396 689 692 ALD SpecificDisease D000326 7717396 731 736 X-ALD Modifier D000326 7717396 882 885 ALD Modifier D000326 7717396 966 969 AMN SpecificDisease D000326 7717396 1100 1103 ADO Modifier D000224 7717396 1349 1352 ALD SpecificDisease D000326 7717396 1357 1360 AMN SpecificDisease D000326 7717396 1475 1480 X-ALD SpecificDisease D000326 7717396 1527 1532 X-ALD Modifier D000326 8571951|t|Atelosteogenesis type II is caused by mutations in the diastrophic dysplasia sulfate-transporter gene (DTDST): evidence for a phenotypic series involving three chondrodysplasias. 8571951|a|Atelosteogenesis type II (AO II) is a neonatally lethal chondrodysplasia whose clinical and histological characteristics resemble those of another chondrodysplasia, the much less severe diastrophic dysplasia (DTD). The similarity suggests a shared pathogenesis involving lesions in the same biochemical pathway and perhaps the same gene. DTD is caused by mutations in the recently identified diastrophic dysplasia sulfate-transporter gene (DTDST). Here, we report that AOII patients also have DTDST mutations, which lead to defective uptake of inorganic sulfate and insufficient sulfation of macromolecules by patient mesenchymal cells in vitro. Together with our recent observation that a third even more severe chondrodysplasia, achondrogenesis type IB, is also caused by mutations in DTDST, these results demonstrate a phenotypic series of three chondrodysplasias of increasing severity caused by lesions in a single sulfate-transporter gene. The severity of the phenotype appears to be correlated with the predicted effect of the mutations on the residual activity of the DTDST protein.. 8571951 0 24 Atelosteogenesis type II SpecificDisease C535395 8571951 55 76 diastrophic dysplasia Modifier C536170 8571951 160 177 chondrodysplasias DiseaseClass D010009 8571951 179 203 Atelosteogenesis type II SpecificDisease C535395 8571951 205 210 AO II SpecificDisease C535395 8571951 235 251 chondrodysplasia DiseaseClass D010009 8571951 326 342 chondrodysplasia DiseaseClass D010009 8571951 365 386 diastrophic dysplasia SpecificDisease C536170 8571951 388 391 DTD SpecificDisease C536170 8571951 517 520 DTD SpecificDisease C536170 8571951 571 592 diastrophic dysplasia Modifier C536170 8571951 648 652 AOII Modifier C535395 8571951 892 908 chondrodysplasia DiseaseClass D010009 8571951 1028 1045 chondrodysplasias DiseaseClass D010009 10732816|t|Emerin, deficiency of which causes Emery-Dreifuss muscular dystrophy, is localized at the inner nuclear membrane. 10732816|a|X-linked recessive Emery-Dreifuss muscular dystrophy (EDMD) is an inherited muscle disorder characterized by the clinical triad of progressive wasting of humero-peroneal muscles, early contractures of the elbows, Achilles tendons and postcervical muscles, and cardiac conduction block with a high risk of sudden death. The gene for EDMD on Xq28 encodes a novel protein named emerin that localizes at the nuclear membrane of skeletal, cardiac and smooth muscles and some other non-muscle tissues. To investigate a possible physiological role for emerin, we examined the ultrastructural localization of the protein in human skeletal muscle and HeLa cells, using ultrathin cryosections. We found that the immune-labeled colloidal gold particles were localized on the nucleoplasmic surface of the inner nuclear membrane, but not on the nuclear pore. Emerin stayed on the cytoplasmic surface of the nuclear lamina, even after detergent treatment that solubilizes membrane lipids and washes out membrane proteins. These results suggest that emerin anchors at the inner nuclear membrane through the hydrophobic stretch, and protrudes from the hydrophilic region to the nucleoplasm where it interacts with the nuclear lamina. We speculate that emerin contributes to maintain the nuclear structure and stability, as well as nuclear functions, particularly in muscle tissues that have severe stress with rigorous contraction-relaxation movements and calcium flux.. 10732816 0 18 Emerin, deficiency SpecificDisease D020389 10732816 35 68 Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10732816 114 166 X-linked recessive Emery-Dreifuss muscular dystrophy SpecificDisease D020389 10732816 168 172 EDMD SpecificDisease D020389 10732816 180 205 inherited muscle disorder DiseaseClass D009135+D030342 10732816 257 291 wasting of humero-peroneal muscles SpecificDisease D019282 10732816 299 368 contractures of the elbows, Achilles tendons and postcervical muscles CompositeMention D003286 10732816 374 398 cardiac conduction block SpecificDisease D006327 10732816 419 431 sudden death SpecificDisease D003645 10732816 446 450 EDMD SpecificDisease D020389 10447258|t|Identification of a common PEX1 mutation in Zellweger syndrome. 10447258|a|The Zellweger spectrum of disease, encompassing Zellweger syndrome and the progressively milder phenotypes of neonatal adrenoleukodystrophy and infantile Refsum disease, is due to a failure to form functional peroxisomes. Cell fusion complementation studies demonstrated that these diseases are genetically heterogeneous, with two-thirds of all patients lying within a single complementation group, CG1. Molecular genetic and cell biology studies have shown that PEX1 is deficient in many CG1 patients. However, previous studies have focused on mildly affected patients and there is still no report of two mutant PEX1 alleles in any Zellweger syndrome patient. Furthermore, mutations in the PMP70 gene have also been identified in two Zellweger syndrome patients from CG1, raising the possibility that CG1 patients may represent a mixture of PEX1-deficient and PMP70-deficient individuals. To address the molecular basis of disease in Zellweger syndrome patients from CG1, we examined all 24 PEX1 exons in four patients, including both patients that have mutations in PMP70. PEX1 mutations were detected in all four patients, including a 1-bp insertion (c. 2097insT) in exon 13 that was present in three of the four patients. Subsequent studies demonstrated that this mutation is present in one-half of all CG1 patients and correlates with the Zellweger syndrome phenotype. As this mutation leads to a loss of protein function its frequency makes it the most common cause of Zellweger syndrome, helping to explain the high percentage of patients that belong to CG1. 10447258 44 62 Zellweger syndrome SpecificDisease D015211 10447258 68 97 Zellweger spectrum of disease DiseaseClass D015211 10447258 112 130 Zellweger syndrome SpecificDisease D015211 10447258 174 203 neonatal adrenoleukodystrophy SpecificDisease OMIM:202370 10447258 208 232 infantile Refsum disease SpecificDisease D052919 10447258 697 715 Zellweger syndrome Modifier D015211 10447258 799 817 Zellweger syndrome Modifier D015211 10447258 999 1017 Zellweger syndrome Modifier D015211 10447258 1408 1426 Zellweger syndrome Modifier D015211 10447258 1539 1557 Zellweger syndrome SpecificDisease D015211 10861282|t|Polymorphisms of the CYP2D6 gene increase susceptibility to ankylosing spondylitis. 10861282|a|Ankylosing spondylitis (AS) is a common and highly familial rheumatic disorder. The sibling recurrence risk ratio for the disease is 63 and heritability assessed in twins > 90%. Although MHC genes, including HLA-B27, contribute only 20-50% of the genetic risk for the disease, no non-MHC gene has yet been convincingly demonstrated to influence either susceptibility to the disease or its phenotypic expression. Previous linkage and association studies have suggested the presence of a susceptibility gene for AS close to, or within, the cytochrome P450 2D6 gene (CYP2D6, debrisoquine hydroxylase) located at chromosome 22q13.1. We performed a linkage study of chromosome 22 in 200 families with AS affected sibling-pairs. Association of alleles of the CYP2D6 gene was examined by both case-control and within-family means. For case-control studies, 617 unrelated individuals with AS (361 probands from sibling-pair and parent-case trio families and 256 unrelated non-familial sporadic cases) and 402 healthy ethnically matched controls were employed. For within-family association studies, 361 families including 161 parent-case trios and 200 affected sibling-pair families were employed. Homozygosity for poor metabolizer alleles was found to be associated with AS. Heterozygosity for the most frequent poor metabolizer allele (CYP2D6*4) was not associated with increased susceptibility to AS. Significant within-family association of CYP2D6*4 alleles and AS was demonstrated. Weak linkage was also demonstrated between CYP2D6 and AS. We postulate that altered metabolism of a natural toxin or antigen by the CYP2D6 gene may increase susceptibility to AS. 10861282 60 82 ankylosing spondylitis SpecificDisease D013167 10861282 84 106 Ankylosing spondylitis SpecificDisease D013167 10861282 108 110 AS SpecificDisease D013167 10861282 144 162 rheumatic disorder DiseaseClass D012216 10861282 594 596 AS SpecificDisease D013167 10861282 780 782 AS SpecificDisease D013167 10861282 965 967 AS SpecificDisease D013167 10861282 1348 1350 AS SpecificDisease D013167 10861282 1476 1478 AS SpecificDisease D013167 10861282 1542 1544 AS SpecificDisease D013167 10861282 1617 1619 AS SpecificDisease D013167 10861282 1738 1740 AS SpecificDisease D013167 10746568|t|Molecular analysis of the genotype-phenotype relationship in factor X deficiency. 10746568|a|Factor X deficiency is a rare haemorrhagic condition, normally inherited as an autosomal recessive trait, in which a variable clinical presentation correlates poorly with laboratory phenotype. The factor X (F10) genes of 14 unrelated individuals with factor X deficiency (12 familial and two sporadic cases) were sequenced yielding a total of 13 novel mutations. Family studies were performed in order to distinguish the contributions of individual mutant F10 alleles to the clinical and laboratory phenotypes. Missense mutations were studied by means of molecular modelling, whereas single basepair substitutions in splice sites and the 5 flanking region were examined by in vitro splicing assay and luciferase reporter gene assay respectively. The deletion allele of a novel hexanucleotide insertion/deletion polymorphism in the F10 gene promoter region was shown by reporter gene assay, to reduce promoter activity by approximately 20%. One family manifesting an autosomal dominant pattern of inheritance possessed three clinically affected members who were heterozygous for a splice-site mutation that was predicted to lead to the production of a truncated protein product. A model which accounts for the dominant negative effect of this lesion is presented. Variation in the antigen level of heterozygous relatives of probands was found to be significantly higher between families than within families, consistent with the view that the nature of the F10 lesion (s) segregating in a given family is a prime determinant of the laboratory phenotype. By contrast, no such relationship could be discerned between laboratory phenotype and polymorphism genotype.. 10746568 61 80 factor X deficiency SpecificDisease D005171 10746568 82 101 Factor X deficiency SpecificDisease D005171 10746568 112 134 haemorrhagic condition DiseaseClass D006474 10746568 333 352 factor X deficiency SpecificDisease D005171 10888879|t|Restoration of photoreceptor ultrastructure and function in retinal degeneration slow mice by gene therapy. 10888879|a|The gene Prph2 encodes a photoreceptor-specific membrane glycoprotein, peripherin-2 (also known as peripherin/rds), which is inserted into the rims of photoreceptor outer segment discs in a complex with rom-1 (ref. 2). The complex is necessary for the stabilization of the discs, which are renewed constantly throughout life, and which contain the visual pigments necessary for photon capture. Mutations in Prph2 have been shown to result in a variety of photoreceptor dystrophies, including autosomal dominant retinitis pigmentosa and macular dystrophy. A common feature of these diseases is the loss of photoreceptor function, also seen in the retinal degeneration slow (rds or Prph2 Rd2/Rd2) mouse, which is homozygous for a null mutation in Prph2. It is characterized by a complete failure to develop photoreceptor discs and outer segments, downregulation of rhodopsin and apoptotic loss of photoreceptor cells. The electroretinograms (ERGs) of Prph2Rd2/Rd2 mice have greatly diminished a-wave and b-wave amplitudes, which decline to virtually undetectable concentrations by two months. Subretinal injection of recombinant adeno-associated virus (AAV) encoding a Prph2 transgene results in stable generation of outer segment structures and formation of new stacks of discs containing both perpherin-2 and rhodopsin, which in many cases are morphologically similar to normal outer segments. Moreover, the re-establishment of the structural integrity of the photoreceptor layer also results in electrophysiological correction. These studies demonstrate for the first time that a complex ultrastructural cell defect can be corrected both morphologically and functionally by in vivo gene transfer.. 10888879 60 80 retinal degeneration Modifier D012162 10888879 563 588 photoreceptor dystrophies DiseaseClass D058499 10888879 600 639 autosomal dominant retinitis pigmentosa SpecificDisease D012174 10888879 644 661 macular dystrophy SpecificDisease D008268 10888879 754 774 retinal degeneration Modifier D012162 2563633|t|Phenylalanine hydroxylase gene haplotypes in Polynesians: evolutionary origins and absence of alleles associated with severe phenylketonuria. 2563633|a|A total of 630 haplotypes for the phenylalanine hydroxylase (PAH) gene locus were established in five groups of Polynesians comprising Samoans, Tongans, Cook Islanders, Maori, and Niueans. Considerable genetic continuity was demonstrated between these widely dispersed populations, since three common haplotypes (4, 1, and 7) constituted over 95% of alleles. A control group of individuals from Southeast Asia shared the same major haplotypes, 4, 1, and 7, with Polynesians. These data provide further support for the theories of genetic homogeneity and of Asian affinities of the Polynesian precursor populations. The absence of severe phenylketonuria (PKU) in both Polynesians and Southeast Asians is consistent with the lack of PAH haplotypes 2 and 3, on which the severe PKU mutants have arisen among Caucasians.. 2563633 125 140 phenylketonuria SpecificDisease D010661 2563633 779 794 phenylketonuria SpecificDisease D010661 2563633 796 799 PKU SpecificDisease D010661 2563633 917 920 PKU Modifier D010661 10557309|t|cDNA microarrays detect activation of a myogenic transcription program by the PAX3-FKHR fusion oncogene. 10557309|a|Alveolar rhabdomyosarcoma is an aggressive pediatric cancer of striated muscle characterized in 60% of cases by a t (2; 13) (q35; q14). This results in the fusion of PAX3, a developmental transcription factor required for limb myogenesis, with FKHR, a member of the forkhead family of transcription factors. The resultant PAX3-FKHR gene possesses transforming properties; however, the effects of this chimeric oncogene on gene expression are largely unknown. To investigate the actions of these transcription factors, both Pax3 and PAX3-FKHR were introduced into NIH 3T3 cells, and the resultant gene expression changes were analyzed with a murine cDNA microarray containing 2, 225 elements. We found that PAX3-FKHR but not PAX3 activated a myogenic transcription program including the induction of transcription factors MyoD, Myogenin, Six1, and Slug as well as a battery of genes involved in several aspects of muscle function. Notable among this group were the growth factor gene Igf2 and its binding protein Igfbp5. Relevance of this model was suggested by verification that three of these genes (IGFBP5, HSIX1, and Slug) were also expressed in alveolar rhabdomyosarcoma cell lines. This study utilizes cDNA microarrays to elucidate the pattern of gene expression induced by an oncogenic transcription factor and demonstrates the profound myogenic properties of PAX3-FKHR in NIH 3T3 cells.. 10557309 105 130 Alveolar rhabdomyosarcoma SpecificDisease D018232 10557309 148 183 pediatric cancer of striated muscle SpecificDisease D019042 10557309 1254 1279 alveolar rhabdomyosarcoma Modifier D018232 3032521|t|Re-evaluation of the sublocalization of esterase D and its relation to the retinoblastoma locus by in situ hybridization. 3032521|a|In situ hybridization of a cDNA probe for the esterase D gene (ESD) was carried out on human chromosomes. The probe hybridized most strongly to 13q14. 2 and 13q14. 3. This observation raises doubts concerning the most recently published assignment of ESD to 13q14. 1. A deletion in an individual with retinoblastoma was reported to separate the closely linked ESD and retinoblastoma (RB1) loci, placing ESD proximal to RB1. Quantitative in situ hybridization studies of this deletion do not confirm this interpretation. Rather, they suggest that ESD is missing from the deleted chromosome 13 and duplicated on the normal homolog. From these findings, we conclude that the deletion in this individual cannot be used to determine the orientation nor the sublocalization of ESD and RB1 within the 13q14 region. 3032521 75 89 retinoblastoma Modifier D012175 3032521 423 437 retinoblastoma SpecificDisease D012175 3032521 490 504 retinoblastoma Modifier D012175 10612394|t|The DNA double-strand break repair gene hMRE11 is mutated in individuals with an ataxia-telangiectasia-like disorder. 10612394|a|We show that hypomorphic mutations in hMRE11, but not in ATM, are present in certain individuals with an ataxia-telangiectasia-like disorder (ATLD). The cellular features resulting from these hMRE11 mutations are similar to those seen in A-T as well as NBS and include hypersensitivity to ionizing radiation, radioresistant DNA synthesis, and abrogation of ATM-dependent events, such as the activation of Jun kinase following exposure to gamma irradiation. Although the mutant hMre11 proteins retain some ability to interact with hRad50 and Nbs1, formation of ionizing radiation-induced hMre11 and Nbs1 foci was absent in hMRE11 mutant cells. These data demonstrate that ATM and the hMre11/hRad50/Nbs1 protein complex act in the same DNA damage response pathway and link hMre11 to the complex pathology of A-T.. 10612394 81 116 ataxia-telangiectasia-like disorder SpecificDisease OMIM:604391 10612394 223 258 ataxia-telangiectasia-like disorder SpecificDisease OMIM:604391 10612394 260 264 ATLD SpecificDisease OMIM:604391 10612394 356 359 A-T SpecificDisease D001260 10612394 371 374 NBS SpecificDisease D049932 10612394 387 425 hypersensitivity to ionizing radiation SpecificDisease D004194 10612394 924 927 A-T SpecificDisease D001260 2450401|t|Duchenne muscular dystrophy gene expression in normal and diseased human muscle. 2450401|a|A probe for the 5 end of the Duchenne muscular dystrophy (DMD) gene was used to study expression of the gene in normal human muscle, myogenic cell cultures, and muscle from patients with DMD. Expression was found in RNA from normal fetal muscle, adult cardiac and skeletal muscle, and cultured muscle after myoblast fusion. In DMD muscle, expression of this portion of the gene was also revealed by in situ RNA hybridization, particularly in regenerating muscle fibers.. 2450401 0 27 Duchenne muscular dystrophy Modifier D020388 2450401 110 137 Duchenne muscular dystrophy Modifier D020388 2450401 139 142 DMD Modifier D020388 2450401 268 271 DMD SpecificDisease D020388 2450401 408 411 DMD Modifier D020388 6086495|t|Localisation of the Becker muscular dystrophy gene on the short arm of the X chromosome by linkage to cloned DNA sequences. 6086495|a|A linkage study in 30 Becker muscular dystrophy (BMD) kindreds using three cloned DNA sequences from the X chromosome which demonstrate restriction fragment length polymorphisms (RFLPs), suggests that the BMD gene is located on the short arm of the X chromosome, in the p21 region. The genes for Becker and Duchenne dystrophies must therefore be closely linked, if not allelic, and any future DNA probes found to be of practical use in one disorder should be equally applicable to the other. The linkage analysis also provides data on the frequency of recombination along the short arm of the X chromosome, and across the centromeric region.. 6086495 20 45 Becker muscular dystrophy Modifier C537666 6086495 146 171 Becker muscular dystrophy SpecificDisease C537666 6086495 173 176 BMD SpecificDisease C537666 6086495 329 332 BMD Modifier C537666 6086495 420 451 Becker and Duchenne dystrophies CompositeMention D020388|C537666 10830915|t|Age of the intronic GAA triplet repeat expansion mutation in Friedreich ataxia. 10830915|a|Friedreich ataxia (FRDA), the most frequently inherited ataxia, is due in the vast majority of cases to a large expansion of an intronic GAA repeat. Using linkage disequilibrium analysis based on haplotype data of seven polymorphic markers close to the frataxin gene, the age of FRDA founding mutational event (s) is estimated to be at least 682 +/-203 generations (95% confidence interval 564-801 g), a dating which is consistent with little or no negative selection and provides further evidence for an ancient spread of a pre-mutation (at-risk alleles) in western Europe.. 10830915 61 78 Friedreich ataxia SpecificDisease D005621 10830915 80 97 Friedreich ataxia SpecificDisease D005621 10830915 99 103 FRDA SpecificDisease D005621 10830915 126 142 inherited ataxia DiseaseClass D013132 10830915 359 363 FRDA Modifier D005621 10556298|t|The human MAGEL2 gene and its mouse homologue are paternally expressed and mapped to the Prader-Willi region. 10556298|a|Prader-Willi syndrome (PWS) is a complex neurogenetic disorder. The phenotype is likely to be a contiguous gene syndrome involving genes which are paternally expressed only, located in the human 15q11-q13 region. Four mouse models of PWS have been reported but these do not definitively allow the delineation of the critical region and the associated genes involved in the aetiology of PWS. Moreover, targeted mutagenesis of mouse homologues of the human candidate PWS genes does not appear to result in any of the features of PWS. Therefore, the isolation of new genes in this region remains crucial for a better understanding of the molecular basis of PWS. In this manuscript, we report the characterization of MAGEL2 and its mouse homologue Magel2. These are located in the human 15q11-q13 and mouse 7C regions, in close proximity to NDN/Ndn. By northern blot analysis we did not detect any expression of MAGEL2/Magel2 but by RT-PCR analysis, specific expression was detected in fetal and adult brain and in placenta. Both genes are intronless with tandem direct repeat sequences contained within a CpG island in the 5-untranscribed region. The transcripts encode putative proteins that are homologous to the MAGE proteins and NDN. Moreover, MAGEL2/Magel2 are expressed only from the paternal allele in brain, suggesting a potential role in the aetiology of PWS and its mouse model, respectively.. 10556298 89 101 Prader-Willi Modifier D011218 10556298 110 131 Prader-Willi syndrome SpecificDisease D011218 10556298 133 136 PWS SpecificDisease D011218 10556298 151 172 neurogenetic disorder DiseaseClass D020271 10556298 206 230 contiguous gene syndrome DiseaseClass D025063 10556298 344 347 PWS SpecificDisease D011218 10556298 496 499 PWS SpecificDisease D011218 10556298 575 578 PWS Modifier D011218 10556298 637 640 PWS SpecificDisease D011218 10556298 764 767 PWS SpecificDisease D011218 10556298 1471 1474 PWS SpecificDisease D011218 1361100|t|Multiple origins for phenylketonuria in Europe. 1361100|a|Phenylketonuria (PKU), a disorder of amino acid metabolism prevalent among Caucasians and other ethnic groups, is caused primarily by a deficiency of the hepatic enzyme phenylalanine hydroxylase (PAH). PKU is a highly heterogeneous disorder, with more than 60 molecular lesions identified in the PAH gene. The haplotype associations, relative frequencies, and distributions of five prevalent PAH mutations (R158Q, R261Q, IVS10nt546, R408W, and IVS12n1) were established in a comprehensive European sample population and subsequently were examined to determine the potential roles of several genetic mechanisms in explaining the present distribution of the major PKU alleles. Each of these five mutations was strongly associated with only one of the more than 70 chromosomal haplotypes defined by eight RFLPs in or near the PAH gene. These findings suggest that each of these mutations arose through a single founding event that occurred within time periods ranging from several hundred to several thousand years ago. From the significant differences observed in the relative frequencies and distributions of these five alleles throughout Europe, four of these putative founding events could be localized to specific ethnic subgroups. Together, these data suggest that there were multiple, geographically and ethnically distinct origins for PKU within the European population.. 1361100 21 36 phenylketonuria SpecificDisease D010661 1361100 48 63 Phenylketonuria SpecificDisease D010661 1361100 65 68 PKU SpecificDisease D010661 1361100 73 106 disorder of amino acid metabolism DiseaseClass D000592 1361100 184 242 deficiency of the hepatic enzyme phenylalanine hydroxylase SpecificDisease OMIM:261600 1361100 250 253 PKU SpecificDisease D010661 1361100 710 713 PKU Modifier D010661 1361100 1388 1391 PKU SpecificDisease D010661 10724160|t|Autoinhibition and activation mechanisms of the Wiskott-Aldrich syndrome protein. 10724160|a|The Rho-family GTPase, Cdc42, can regulate the actin cytoskeleton through activation of Wiskott-Aldrich syndrome protein (WASP) family members. Activation relieves an autoinhibitory contact between the GTPase-binding domain and the carboxy-terminal region of WASP proteins. Here we report the autoinhibited structure of the GTPase-binding domain of WASP, which can be induced by the C-terminal region or by organic co-solvents. In the autoinhibited complex, intramolecular interactions with the GTPase-binding domain occlude residues of the C terminus that regulate the Arp2/3 actin-nucleating complex. Binding of Cdc42 to the GTPase-binding domain causes a dramatic conformational change, resulting in disruption of the hydrophobic core and release of the C terminus, enabling its interaction with the actin regulatory machinery. These data show that intrinsically unstructured peptides such as the GTPase-binding domain of WASP can be induced into distinct structural and functional states depending on context.. 10724160 48 72 Wiskott-Aldrich syndrome Modifier D014923 10724160 170 194 Wiskott-Aldrich syndrome Modifier D014923 1733838|t|Localisation of the gene for Norrie disease to between DXS7 and DXS426 on Xp. 1733838|a|A highly informative microsatellite marker, DXS426, which maps proximal to DXS7 in the interval Xp11. 4-Xp11 4-Xp11. 23, has been used to refine further the localisation of the gene for Norrie disease (NDP). The results from a multiply informative crossover localize the NDP gene proximal to DXS7. In conjunction with information from 2 NDP patients who have a deletion for DXS7 but not for DSX426, our data indicate that the NDP gene lies between DXS7 and DXS426 on proximal Xp. 1733838 29 43 Norrie disease SpecificDisease C537849 1733838 264 278 Norrie disease SpecificDisease C537849 1733838 280 283 NDP SpecificDisease C537849 1733838 349 352 NDP Modifier C537849 1733838 415 418 NDP Modifier C537849 1733838 504 507 NDP Modifier C537849 7479827|t|Molecular basis of human mitochondrial very-long-chain acyl-CoA dehydrogenase deficiency causing cardiomyopathy and sudden death in childhood. 7479827|a|beta-Oxidation of long-chain fatty acids provides the major source of energy in the heart. Defects in enzymes of the beta-oxidation pathway cause sudden, unexplained death in childhood, acute hepatic encephalopathy or liver failure, skeletal myopathy, and cardiomyopathy. Very-long-chain acyl-CoA dehydrogenase [VLCAD; very-long-chain-acyl-CoA (acceptor) 2, 3-oxidoreductase, EC 1. 3 3. 99 99. 13] catalyzes the first step in beta-oxidation. We have isolated the human VLCAD cDNA and gene and determined the complete nucleotide sequences. Polymerase chain reaction amplification of VLCAD mRNA and genomic exons defined the molecular defects in two patients with VLCAD deficiency who presented with unexplained cardiac arrest and cardiomyopathy. In one, a homozygous mutation in the consensus dinucleotide of the donor splice site (g + 1-- > a) was associated with universal skipping of the prior exon (exon 11). The second patient was a compound heterozygote, with a missense mutation, C1837-- > T, changing the arginine at residue 613 to tryptophan on one allele and a single base deletion at the intron-exon 6 boundary as the second mutation. This initial delineation of human mutations in VLCAD suggests that VLCAD deficiency reduces myocardial fatty acid beta-oxidation and energy production and is associated with cardiomyopathy and sudden death in childhood. 7479827 39 88 very-long-chain acyl-CoA dehydrogenase deficiency SpecificDisease C536353 7479827 97 111 cardiomyopathy SpecificDisease D009202 7479827 116 128 sudden death SpecificDisease D003645 7479827 289 314 sudden, unexplained death SpecificDisease D003645 7479827 335 357 hepatic encephalopathy SpecificDisease D006501 7479827 361 374 liver failure SpecificDisease D017093 7479827 376 393 skeletal myopathy SpecificDisease D009135 7479827 399 413 cardiomyopathy SpecificDisease D009202 7479827 806 822 VLCAD deficiency SpecificDisease C536353 7479827 854 868 cardiac arrest SpecificDisease D006323 7479827 873 887 cardiomyopathy SpecificDisease D009202 7479827 1356 1372 VLCAD deficiency SpecificDisease C536353 7479827 1463 1477 cardiomyopathy SpecificDisease D009202 7479827 1482 1494 sudden death SpecificDisease D003645 1577476|t|An error in dystrophin mRNA processing in golden retriever muscular dystrophy, an animal homologue of Duchenne muscular dystrophy. 1577476|a|Golden retriever muscular dystrophy (GRMD) is a spontaneous, X-linked, progressively fatal disease of dogs and is also a homologue of Duchenne muscular dystrophy (DMD). Two-thirds of DMD patients carry detectable deletions in their dystrophin gene. The defect underlying the remaining one-third of DMD patients is undetermined. Analysis of the canine dystrophin gene in normal and GRMD dogs has failed to demonstrate any detectable loss of exons. Here, we have demonstrated a RNA processing error in GRMD that results from a single base change in the 3 consensus splice site of intron 6. The seventh exon is then skipped, which predicts a termination of the dystrophin reading frame within its N-terminal domain in exon 8. This is the first example of dystrophin deficiency caused by a splice-site mutation.. 1577476 42 77 golden retriever muscular dystrophy SpecificDisease D009136 1577476 102 129 Duchenne muscular dystrophy SpecificDisease D020388 1577476 131 166 Golden retriever muscular dystrophy SpecificDisease D009136 1577476 168 172 GRMD SpecificDisease D009136 1577476 265 292 Duchenne muscular dystrophy SpecificDisease D020388 1577476 294 297 DMD SpecificDisease D020388 1577476 314 317 DMD Modifier D020388 1577476 429 432 DMD Modifier D020388 1577476 512 516 GRMD Modifier D009136 1577476 631 635 GRMD SpecificDisease D009136 1577476 883 904 dystrophin deficiency SpecificDisease OMIM:310200 1981994|t|Serum amyloid A and P protein genes in familial Mediterranean fever. 1981994|a|Two recent studies have suggested the involvement of serum amyloid A (SAA) and P (APCS) genes in familial Mediterranean fever (MEF). To test the role of SAA and APCS in MEF and MEF-amyloidosis, we studied 17 informative families (15 Armenians, 2 non-Ashkenazi Jews) and 8 MEF patients with amyloidosis using a candidate gene approach. No evidence for any MEF-associated polymorphism was found in any of the 41 Armenian and Jewish MEF patients tested. Our family studies allowed us to rule out tight linkage between SAA and MEF (lod score = -2. 16, theta less than or equal to 0. 06). For APCS we found that the allele frequency in the MEF-amyloidosis patients was similar to that in 18 unrelated MEF patients without amyloidosis and their 33 healthy parents. 1981994 39 67 familial Mediterranean fever SpecificDisease D010505 1981994 166 194 familial Mediterranean fever SpecificDisease D010505 1981994 196 199 MEF SpecificDisease D010505 1981994 238 241 MEF SpecificDisease D010505 1981994 246 261 MEF-amyloidosis SpecificDisease D000686|D010505 1981994 341 344 MEF Modifier D010505 1981994 359 370 amyloidosis DiseaseClass D000686 1981994 424 427 MEF Modifier D010505 1981994 499 502 MEF Modifier D010505 1981994 592 595 MEF SpecificDisease D010505 1981994 704 719 MEF-amyloidosis Modifier D000686|D010505 1981994 765 768 MEF Modifier D010505 1981994 786 797 amyloidosis DiseaseClass D000686 1201235|t|Glucose-6-phosphate dehydrogenase variants from Italian subjects associated with severe neonatal jaundice. 1201235|a|Screening for the G6PD deficiency was carried out at the Maternity Division of the Galliera Hospital in Genoa, Italy. Two groups of subjects with hyperbilirubinaemia of non-immunological origin were examined (a) 302 newborn babies of Sardinian extraction (on cord blood) and (b) 201 newborn babies of south Italian ancestry (on peripheral blood). Among 503 subjects, 43 showed an enzyme deficiency; in 39 the defect was of the Mediterranean type. In one case, previously described, the enzyme was of the A- type. In the remaining cases three different variants were identified. In the present work these three cases, each with severe neonatal jaundice, are reported. Their parents originated from Calabria, from Sardinia and from Sicily. The abnormal enzymes are respectively designated as GdDcbrousse-like, GdGallura and GdAgrigento.. 1201235 81 105 severe neonatal jaundice SpecificDisease D007567 1201235 125 140 G6PD deficiency SpecificDisease D005955 1201235 253 272 hyperbilirubinaemia DiseaseClass D006932 1201235 735 759 severe neonatal jaundice SpecificDisease D007567 1351034|t|Loss of normal allele of the APC gene in an adrenocortical carcinoma from a patient with familial adenomatous polyposis. 1351034|a|Endocrine neoplasms have been reported occasionally in patients with familial adenomatous polyposis (FAP). An adrenocorotical carcinoma was studied in a patient with a family history of FAP. Loss of heterozygosity (LOH) in the region close to the adenomatous polyposis coli (APC) gene was detected in this carcinoma, and evidence was obtained that there was a loss of the normal allele of the APC gene. This is the first demonstration of LOH at the APC locus in adrenocortical tumors. The present results and our previous data on LOH in a recurring desmoid tumor suggest that the heterozygous mutant/wild-type condition of the APC gene may give rise to benign tumors, and that functional loss of this gene leads to development of tumors not only in the colon but also in other various parts of the body in FAP patients.. 1351034 29 32 APC Modifier D011125 1351034 44 68 adrenocortical carcinoma SpecificDisease D018268 1351034 89 119 familial adenomatous polyposis SpecificDisease D011125 1351034 121 140 Endocrine neoplasms DiseaseClass D004701 1351034 190 220 familial adenomatous polyposis SpecificDisease D011125 1351034 222 225 FAP SpecificDisease D011125 1351034 231 256 adrenocorotical carcinoma SpecificDisease D018268 1351034 307 310 FAP SpecificDisease D011125 1351034 368 394 adenomatous polyposis coli Modifier D011125 1351034 396 399 APC Modifier D011125 1351034 427 436 carcinoma DiseaseClass D002277 1351034 514 517 APC Modifier D011125 1351034 570 573 APC Modifier D011125 1351034 583 604 adrenocortical tumors SpecificDisease D018268 1351034 670 683 desmoid tumor SpecificDisease C535944 1351034 748 751 APC Modifier D011125 1351034 774 787 benign tumors DiseaseClass D009369 1351034 851 857 tumors DiseaseClass D009369 1351034 927 930 FAP Modifier D011125 7191069|t|Heterozygous C2-deficiency and myasthenia gravis. 7191069|a|Complement deficiency states in myasthenia gravis (MG) have not been reported previously. We describe a 19-year-old woman with typical MG and heterozygous C2 deficiency, along with HLA typing of the patient and her immediate family.. 7191069 13 26 C2-deficiency SpecificDisease OMIM:217000 7191069 31 48 myasthenia gravis SpecificDisease D009157 7191069 50 71 Complement deficiency Modifier D007153 7191069 82 99 myasthenia gravis SpecificDisease D009157 7191069 101 103 MG SpecificDisease D009157 7191069 185 187 MG SpecificDisease D009157 7191069 205 218 C2 deficiency SpecificDisease OMIM:217000 10472529|t|Large heterozygous deletion masquerading as homozygous missense mutation: a pitfall in diagnostic mutation analysis. 10472529|a|The clinical use of molecular analyses in recessive disorders relies on the exact characterization of both mutant alleles in the affected patient. This can be problematic when only part of the gene is examined or when relevant DNA alterations are not recognized by standard methods. We present a child in whom phenylketonuria was apparently caused by homozygosity for the mutation E390G in exon 11 of the phenylalanine hydroxylase (PAH) gene. However, the clinical severity of the disease was not quite as mild as expected, the mutation was not identified in the father despite confirmed paternity, and the paternal allele showed a highly unusual pattern of polymorphic markers in the PAH gene. Presence of a large deletion involving exons 9, 10 and 11 of the phenylalanine hydroxylase gene was confirmed by long-range PCR. Diagnostic DNA analyses should include a comprehensive examination of the whole relevant gene in the patient and confirmation of carrier status in both parents.. 10472529 159 178 recessive disorders DiseaseClass D030342 10472529 427 442 phenylketonuria SpecificDisease D010661 2912069|t|Two point mutations are responsible for G6PD polymorphism in Sardinia. 2912069|a|The human X-linked gene encoding glucose 6-phosphate dehydrogenase (G6PD) is highly polymorphic; more than 300 G6PD variants have been identified. G6PD deficiency in different geographical areas appears to have arisen through independent mutational events, but within the same population it may also be heterogeneous. One example is the island of Sardinia, where careful clinical and biochemical studies have identified four different G6PD variants. We cloned and sequenced the four G6PD variants from Sardinia and found that only two mutations are responsible for G6PD deficiency in this area one mutation is the cause of the G6PD Seattle-like phenotype, a milder form of G6PD deficiency; the other mutation is responsible for all forms of very severe G6PD deficiency in Sardinia and, possibly, in the Mediterranean.. 2912069 218 233 G6PD deficiency SpecificDisease D005955 2912069 636 651 G6PD deficiency SpecificDisease D005955 2912069 699 726 G6PD Seattle-like phenotype SpecificDisease D005955 2912069 745 760 G6PD deficiency SpecificDisease D005955 2912069 825 840 G6PD deficiency SpecificDisease D005955 6902670|t|Familial discoid lupus erythematosus associated with heterozygote C2 deficiency. 6902670|a|Two siblings with chronic discoid lupus erythematosus and several family members were found with heterozygous C2 deficiency. An association with histocompatibility markers HLA-B18 and HLA-Dw2 was demonstrated, and the slow allotype of factor B was present. Linkage studies in this family suggested a close linkage between the C2 deficiency gene and genes coding for B18, Dw2, and BfS antigens. One HLA-ACB/DBf recombinant was observed showing closer linkage between HLA-D and Bf than between HLA-B and Bf.. 6902670 0 36 Familial discoid lupus erythematosus SpecificDisease D008179 6902670 66 79 C2 deficiency SpecificDisease OMIM:217000 6902670 99 134 chronic discoid lupus erythematosus SpecificDisease D008179 6902670 191 204 C2 deficiency SpecificDisease OMIM:217000 6902670 407 420 C2 deficiency Modifier OMIM:217000 7545954|t|A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. 7545954|a|A strong candidate for the 17q-linked BRCA1 gene, which influences susceptibility to breast and ovarian cancer, has been identified by positional cloning methods. Probable predisposing mutations have been detected in five of eight kindreds presumed to segregate BRCA1 susceptibility alleles. The mutations include an 11-base pair deletion, a 1-base pair insertion, a stop codon, a missense substitution, and an inferred regulatory mutation. The BRCA1 gene is expressed in numerous tissues, including breast and ovary, and encodes a predicted protein of 1863 amino acids. This protein contains a zinc finger domain in its amino-terminal region, but is otherwise unrelated to previously described proteins. Identification of BRCA1 should facilitate early diagnosis of breast and ovarian cancer susceptibility in some individuals as well as a better understanding of breast cancer biology.. 7545954 27 52 breast and ovarian cancer Modifier D061325 7545954 165 190 breast and ovarian cancer CompositeMention D061325 7545954 846 871 breast and ovarian cancer Modifier D061325 7545954 944 957 breast cancer Modifier D001943 8198128|t|Recombinations in individuals homozygous by descent localize the Friedreich ataxia locus in a cloned 450-kb interval. 8198128|a|The locus for Friedreich ataxia (FRDA), a severe neurodegenerative disease, is tightly linked to markers D9S5 and D9S15, and analysis of rare recombination events has suggested the order cen-FRDA-D9S5-D9S15-qter. We report here the construction of a YAC contig extending 800 kb centromeric to D9S5 and the isolation of five new microsatellite markers from this region. In order to map these markers with respect to the FRDA locus, all within a 1-cM confidence interval, we sought to increase the genetic information of available FRDA families by considering homozygosity by descent and association with founder haplotypes in isolated populations. This approach allowed us to identify one phase-known recombination and one probable historic recombination on haplotypes from Reunion Island patients, both of which place three of the five markers proximal to FRDA. This represents the first identification of close FRDA flanking markers on the centromeric side. The two other markers allowed us to narrow the breakpoint of a previously identified distal recombination that is > 180 kb from D9S5 (26P). Taken together, the results place the FRDA locus in a 450-kb interval, which is small enough for direct search of candidate genes. A detailed rare cutter restriction map and a cosmid contig covering this interval were constructed and should facilitate the search of genes in this region.. 8198128 65 82 Friedreich ataxia SpecificDisease D005621 8198128 132 149 Friedreich ataxia SpecificDisease D005621 8198128 151 155 FRDA SpecificDisease D005621 8198128 167 192 neurodegenerative disease DiseaseClass D019636 8198128 537 541 FRDA Modifier D005621 8198128 647 651 FRDA Modifier D005621 8198128 974 978 FRDA SpecificDisease D005621 8198128 1030 1034 FRDA Modifier D005621 8198128 1255 1259 FRDA Modifier D005621 2220826|t|Further mapping of an ataxia-telangiectasia locus to the chromosome 11q23 region. 2220826|a|We recently mapped the gene for ataxia-telangiectasia group A (ATA) to chromosome 11q22-23 by linkage analysis, using the genetic markers THY1 and pYNB3. 12 (D11S144). The most likely order was cent-AT-S144-THY1. The present paper describes further mapping of the AT locus by means of a panel of 10 markers that span approximately 60 cM in the 11q22-23 region centered around S144 and THY1. Location scores indicate that three contiguous subsegments within the [S144-THY1] segment, as well as three contiguous segments telomeric to THY1, are each unlikely to contain the AT locus, while the more centromeric [STMY-S144] segment is most likely to contain the AT locus. These data, together with recent refinements in the linkage and physical maps of 11q22-23, place the AT locus at 11q23. 2220826 22 43 ataxia-telangiectasia Modifier D001260 2220826 114 135 ataxia-telangiectasia Modifier D001260 2220826 346 348 AT Modifier D001260 2220826 653 655 AT Modifier D001260 2220826 740 742 AT Modifier D001260 2220826 851 853 AT Modifier D001260 1311721|t|Proliferation-related expression of p19/nm23 nucleoside diphosphate kinase. 1311721|a|High level expression of the nm23-H1 gene, which encodes for a nucleoside diphosphate kinase, has been found to correlate with diminished metastasis in some tumors but not in others. We have previously identified the protein product of the nm23-H1 gene in two-dimensional electrophoretic gels and have designated it p19/nm23. In neuroblastoma, higher levels of p19/nm23, which are associated with amplification of the N-myc oncogene, large tumor mass, and metastasis, were observed in advanced stage tumors compared with limited stage disease. Because of the variable expression of nm23-H1 in different tumors, we have investigated the relationship between amounts of the protein and cell proliferation. The levels of p19/nm23 were compared between resting and mitotically stimulated normal human PBLs and in leukemia cells. The amount of p19/nm23 increased in normal lymphocytes in response to mitotic stimulation and paralleled the increase in DNA synthesis. In leukemia cells obtained from patients with different subtypes of acute leukemia, p19/nm23 levels were also increased relative to resting normal lymphocytes. Treatment of mitotically stimulated lymphocytes with cyclosporin, which inhibits proliferation, blocked the increase in p19/nm23; treatment of the leukemia cell line HL-60 with dimethylsulfoxide, which induces terminal differentiation, resulted in diminished levels of p19/nm23. Our data therefore provide evidence that nm23-H1 expression is related to cell proliferative activity.. 1311721 233 239 tumors DiseaseClass D009369 1311721 405 418 neuroblastoma SpecificDisease D009447 1311721 516 521 tumor Modifier D009369 1311721 576 582 tumors DiseaseClass D009369 1311721 679 685 tumors DiseaseClass D009369 1311721 885 893 leukemia Modifier D007938 1311721 1040 1048 leukemia Modifier D007938 1311721 1105 1119 acute leukemia SpecificDisease D007938 1311721 1344 1352 leukemia Modifier D007938 116187|t|Carrier detection of pyruvate carboxylase deficiency in fibroblasts and lymphocytes. 116187|a|Pyruvate carboxylase (E. C. 6. 4. 1. 1) activity was determined in the circulating peripheral lymphocytes and cultured skin fibroblasts from the family of a patient with hepatic, cerebral, renal cortical, leukocyte, and fibroblast pyruvate carboxylase deficiency (PC Portland deficiency). Lymphocyte activities were mother, 33--39%; father, 11--29%; brother, 82--103%; and sister, 38--48% of the lowest normal. Fibroblasts from the patients mother and father had 42 and 34%, respectively, of the activity of the lowest normal. These data demonstrate that the disease is inherited in an autosomal recessive manner and that lymphocytes and fibroblasts can be used to detect carriers. Neither pyruvate carboxylase nor mitochondrial PEPCK activity in lymphocytes was increased by a 21-hr fast. 116187 21 52 pyruvate carboxylase deficiency SpecificDisease D015324 116187 316 347 pyruvate carboxylase deficiency SpecificDisease D015324 116187 349 371 PC Portland deficiency SpecificDisease D015324 2852474|t|Complex glycerol kinase deficiency: molecular-genetic, cytogenetic, and clinical studies of five Japanese patients. 2852474|a|Five male Japanese patients with complex glycerol kinase deficiency (CGKD) and their relatives were studied clinically, cytogenetically, and molecular-genetically. All patients had muscular dystrophy or muscle weakness, mental retardation, congenital adrenal hypoplasia, and glycerol kinase deficiency. High-resolution GTG-banded chromosomes showed a microdeletion in the Xp21 region in all four patients examined and in all five mothers. Southern hybridizations, after digestions by restriction endonucleases, with various cloned DNAs (D2, 99-6, B24, C7, L1-4, cDMD13-14, J66-HI, P20, J-Bir, ERT87-30, ERT87-15, ERT87-8, ERT87-1, XJ-1. 1, 754, cx5. 7, and OTC-1) that are located around Xp21 also showed a deletion in the genome of all patients and mothers. Although the deletion differed in size among patients, a segment commonly absent was located between the genomic sequences corresponding to L1-4 and cDMD13-14. This finding indicated that the gene coding for glycerol kinase (GK) is located within this segment. A comparison of the clinical manifestations of the present five patients and reported CGKD or Duchenne muscular dystrophy (DMD) patients with DNA deletion suggests the existence of a certain gene responsible for gonadotropin deficiency (GTD). The result of the present study and results of previous studies suggest that genes for ornithine transcarbamylase (OTC), DMD, and GK and putative genes responsible for congenital adrenal hypoplasia (AHC) and GTD are arranged from telomere to centromere as pter--GTD--AHC--GK--DMD--OTC--cen 2852474 0 34 Complex glycerol kinase deficiency SpecificDisease C538138 2852474 149 183 complex glycerol kinase deficiency SpecificDisease C538138 2852474 185 189 CGKD SpecificDisease C538138 2852474 297 315 muscular dystrophy DiseaseClass D009136 2852474 319 334 muscle weakness DiseaseClass D018908 2852474 336 354 mental retardation DiseaseClass D008607 2852474 356 385 congenital adrenal hypoplasia SpecificDisease D000312 2852474 391 417 glycerol kinase deficiency DiseaseClass C538138 2852474 1222 1226 CGKD Modifier C538138 2852474 1230 1257 Duchenne muscular dystrophy Modifier D020388 2852474 1259 1262 DMD Modifier D020388 2852474 1348 1371 gonadotropin deficiency SpecificDisease C535764 2852474 1373 1376 GTD SpecificDisease C535764 2852474 1500 1503 DMD SpecificDisease D020388 2852474 1547 1576 congenital adrenal hypoplasia SpecificDisease D000312 2852474 1578 1581 AHC SpecificDisease D000312 2852474 1587 1590 GTD SpecificDisease C535764 7726234|t|Duchenne muscular dystrophy and myotonic dystrophy in the same patient. 7726234|a|We report on the first patient identified with myotonic dystrophy and Duchenne muscular dystrophy (DMD). The family of the propositus had a strong history of myotonic dystrophy, and there was an intrafamilial pathological expansion of the responsible CTG repeat between the mildly affected mother (160 repeats; normal 27 repeats) and her more severely affected son (650 repeats), and his sister (650 repeats). The propositus was an isolated case of Duchenne muscular dystrophy with marked dystrophin deficiency in muscle biopsy. The patient was still ambulatory post age 16. Myotonic dystrophy could interfere to some extent with the progression of Duchenne dystrophy. However, other interpretations are possible. Twelve percent of dystrophin revertant fibers as observed by immunohistochemistry could be sufficient to ameliorate typical DMD clinical severity, or the patient may present a somatic mosaic. The pathophysiological interactions of these two unlinked disorders are discussed at the clinical and histopathological levels.. 7726234 0 27 Duchenne muscular dystrophy SpecificDisease D020388 7726234 32 50 myotonic dystrophy SpecificDisease D009223 7726234 119 137 myotonic dystrophy SpecificDisease D009223 7726234 142 169 Duchenne muscular dystrophy SpecificDisease D020388 7726234 171 174 DMD SpecificDisease D020388 7726234 230 248 myotonic dystrophy SpecificDisease D009223 7726234 521 548 Duchenne muscular dystrophy SpecificDisease D020388 7726234 561 582 dystrophin deficiency SpecificDisease OMIM:310200 7726234 647 665 Myotonic dystrophy SpecificDisease D009223 7726234 721 739 Duchenne dystrophy SpecificDisease D020388 7726234 910 913 DMD Modifier D020388 8500791|t|Autosomal recessive transmission of hemophilia A due to a von Willebrand factor mutation. 8500791|a|The differential diagnosis of the genetic bleeding disorders, hemophilia A and von Willebrand disease, is occasionally confounded by the close molecular relationship of coagulation factor VIII and von Willebrand factor (vWF). This report describes the autosomal inheritance of a hemophilia A phenotype due to a mutation of vWF that results in defective factor VIII binding. The proband was a female patient with low levels of factor VIII activity. Polymerase chain reaction (PCR) amplification and DNA sequencing were employed to examine exons encoding the putative factor VIII binding domain of vWF. The patient was found to be homozygous for a single point mutation causing a Thr-- > Met substitution at amino acid position 28 in the mature vWF subunit. The phenotypic expression of the mutation was determined to be recessive because heterozygous family members were clinically unaffected. Recombinant vWF containing the observed amino acid substitution was expressed in COS-1 cells. The mutant vWF was processed and secreted normally, and was functionally equivalent to wild-type vWF in its ability to bind to platelets. However, the mutant failed to bind factor VIII, demonstrating that the mutation was functionally related to the observed hemophilia phenotype. The family we describe demonstrates the recessive inheritance of a recently recognized class of genetic bleeding disorders, we call " autosomal hemophilia. " We conclude that vWF mutation may be an under recognized cause of hemophilia, especially in cases where the inheritance pattern is not consistent with X-linked transmission. 8500791 36 48 hemophilia A SpecificDisease D006467 8500791 58 72 von Willebrand Modifier D014842 8500791 124 150 genetic bleeding disorders DiseaseClass D025861 8500791 152 164 hemophilia A SpecificDisease D006467 8500791 169 191 von Willebrand disease SpecificDisease D014842 8500791 287 301 von Willebrand Modifier D014842 8500791 369 381 hemophilia A Modifier D006467 8500791 1336 1346 hemophilia Modifier D006467|D002836 8500791 1502 1512 hemophilia SpecificDisease D006467|D002836 8500791 1582 1592 hemophilia SpecificDisease D006467|D002836 10737980|t|Determination of 30 X-linked adrenoleukodystrophy mutations, including 15 not previously described. 10737980|a|X-linked Adrenoleukodystrophy (X-ALD) is the most frequent peroxisomal disease. It mainly involves the nervous system white matter, adrenal cortex and testes. Several distinct clinical phenotypes are known. The principal biochemical abnormality is the accumulation of saturated very-long-chain fatty acids (VLCFAs > C22 0, mainly C26 0), which is due to impaired capacity for beta-oxidation in peroxisomes. Diagnosis is usually based on the VLCFA levels in plasma or cultured skin fibroblasts in both patients and carriers. In 0. 1% of affected males, however, the plasma C26 0 level is borderline normal, and 15% of obligate female carriers have normal results. Effective mutation detection in these families is therefore fundamental to unambiguously determine the genetic status of each individual at risk. Of particular concern are female members of kindreds segregating X-ALD mutations, because normal VLCFA levels do not guarantee lack of carrier status. We describe a fast method for detection of X-ALD mutations. The method is based on SSCP analysis of nested PCR fragments followed by sequence-determination reactions. Using this methodology we have found X-ALD mutations in 30 kindreds, including 15 not previously reported. 10737980 20 49 X-linked adrenoleukodystrophy Modifier D000326 10737980 100 129 X-linked Adrenoleukodystrophy SpecificDisease D000326 10737980 131 136 X-ALD SpecificDisease D000326 10737980 159 178 peroxisomal disease DiseaseClass D018901 10737980 978 983 X-ALD Modifier D000326 10737980 1107 1112 X-ALD Modifier D000326 10737980 1268 1273 X-ALD Modifier D000326 7894491|t|Mutations in the BRCA1 gene in families with early-onset breast and ovarian cancer. 7894491|a|We analysed 50 probands with a family history of breast and/or ovarian cancer for germline mutations in the coding region of the BRCA1 candidate gene, using single-strand conformation polymorphism (SSCP) analysis on PCR-amplified genomic DNA. A total of eight putative disease-causing alterations were identified four of these are frameshifts and two are nonsense mutations. In addition, we found two missense mutations, one of which changes the final cysteine of the BRCA1 zinc finger motif to glycine. These data are consistent with a tumour suppressor model, and support the notion that this candidate gene is in fact BRCA1. The heterogeneity of mutations, coupled with the large size of the gene, indicates that clinical application of BRCA1 mutation testing will be technically challenging.. 7894491 57 82 breast and ovarian cancer CompositeMention D061325 7894491 133 161 breast and/or ovarian cancer CompositeMention D061325 7894491 622 628 tumour Modifier D009369 2355960|t|Screening for carriers of Tay-Sachs disease among Ashkenazi Jews. A comparison of DNA-based and enzyme-based tests. 2355960|a|BACKGROUND AND METHODS. The prevention of Tay-Sachs disease (GM2 gangliosidosis, type 1) depends on the identification of carriers of the gene for this autosomal recessive disorder. We compared the enzyme-based test widely used in screening for Tay-Sachs disease with a test based on analysis of DNA. We developed methods to detect the three mutations in the HEXA gene that occur with high frequency among Ashkenazi Jews two mutations cause infantile Tay-Sachs disease, and the third causes the adult-onset form of the disease. DNA segments containing these mutation sites were amplified with the polymerase chain reaction and analyzed for the presence of the mutations. RESULTS. Among 62 Ashkenazi obligate carriers of Tay-Sachs disease, the three specific mutations accounted for all but one of the mutant alleles (98 percent). In 216 Ashkenazi carriers identified by the enzyme test, DNA analysis showed that 177 (82 percent) had one of the identified mutations. Of the 177, 79 percent had the exon 11 insertion mutation, 18 percent had the intron 12 splice-junction mutation, and 3 percent had the less severe exon 7 mutation associated with adult-onset disease. The results of the enzyme tests in the 39 subjects (18 percent) who were defined as carriers but in whom DNA analysis did not identify a mutant allele were probably false positive (although there remains some possibility of unidentified mutations). In addition, of 152 persons defined as noncarriers by the enzyme-based test, 1 was identified as a carrier by DNA analysis (i. e e., a false negative enzyme-test result). CONCLUSIONS. The increased specificity and predictive value of the DNA-based test make it a useful adjunct to the diagnostic tests currently used to screen for carriers of Tay-Sachs disease. 2355960 26 43 Tay-Sachs disease SpecificDisease D013661 2355960 158 175 Tay-Sachs disease SpecificDisease D013661 2355960 177 203 GM2 gangliosidosis, type 1 SpecificDisease D013661 2355960 268 296 autosomal recessive disorder DiseaseClass D030342 2355960 361 378 Tay-Sachs disease SpecificDisease D013661 2355960 568 585 Tay-Sachs disease SpecificDisease D013661 2355960 837 854 Tay-Sachs disease SpecificDisease D013661 2355960 1876 1893 Tay-Sachs disease SpecificDisease D013661 8002973|t|Adrenoleukodystrophy gene encodes an 80 kDa membrane protein. 8002973|a|An antibody against the synthetic C-terminal peptides deduced from the cDNA of the gene responsible for X-linked adrenoleukodystrophy (ALD) was produced to characterize the product of the ALD gene. The antibody reacted with the 80 kDa band protein in control fibroblasts, while no bands were detected in the fibroblasts from a patient with ALD (# 163), in which mRNA of the ALD gene was undetectable based on Northern blot analysis. The 293T cells transfected with the full-coding cDNA inserted in the expression vector produced a new 80 kDa protein, as detected by Western blot. In an immunocytological study, the staining was in a punctate pattern, in the normal fibroblasts. However, there was no punctate staining in the # 163 cells. These data thus indicate that the ALD gene encodes an 80 kDa membrane protein.. 8002973 0 20 Adrenoleukodystrophy Modifier D000326 8002973 166 195 X-linked adrenoleukodystrophy SpecificDisease D000326 8002973 197 200 ALD SpecificDisease D000326 8002973 250 253 ALD Modifier D000326 8002973 402 405 ALD SpecificDisease D000326 8002973 436 439 ALD Modifier D000326 8002973 834 837 ALD Modifier D000326 2568588|t|Preferential germline mutation of the paternal allele in retinoblastoma. 2568588|a|The event triggering malignant proliferation in 70% of retinoblastoma tumours is loss of heterozygosity for chromosome 13q14, whereby the normal retinoblastoma gene (RB1) allele is lost and an already mutated RB1 allele remains in the tumour. The first allele suffers a mutational event--deletion, duplication or point mutation (manuscript in preparation) --either in the germ line (all bilateral patients) or in a somatic retinal cell (most unilateral patients). Most bilateral patients have no family history of retinoblastoma and are presumed to have new germline mutations which arose in the egg, sperm or early embryo. We have determined the parental origin of the retained allele in nine retinoblastoma tumours from eight unrelated non-familial cases by using RB1-linked genetic markers. Six tumours retained the paternal allele and three retained the maternal allele. Of the three unilateral tumours, only one retained the paternal RB1 allele. Thus, there is no evidence that the paternal RB1 allele is preferentially retained in retinoblastoma, as has been suggested to be the case in osteosarcoma. By contrast, tumours from four of the five bilateral patients retained the paternal RB1 allele. This suggests either that new germline RB1 mutations arise more frequently during spermatogenesis than during oogenesis, or that imprinting in the early embryo affects chromosomal susceptibility to mutation.. 2568588 57 71 retinoblastoma SpecificDisease D012175 2568588 128 150 retinoblastoma tumours SpecificDisease D012175 2568588 218 232 retinoblastoma Modifier D012175 2568588 308 314 tumour DiseaseClass D009369 2568588 587 601 retinoblastoma SpecificDisease D012175 2568588 767 789 retinoblastoma tumours SpecificDisease D012175 2568588 871 878 tumours DiseaseClass D009369 2568588 961 979 unilateral tumours DiseaseClass D009369 2568588 1110 1124 retinoblastoma SpecificDisease D012175 2568588 1166 1178 osteosarcoma SpecificDisease D012516 2568588 1193 1200 tumours DiseaseClass D009369 10817650|t|Mutations at the ataxia-telangiectasia locus and clinical phenotypes of A-T patients. 10817650|a|Mutations at the ataxia-telangiectasia (A-T) locus on chromosome band 11q22 cause a distinctive autosomal recessive syndrome in homozygotes and predispose heterozygotes to cancer, ischemic heart disease, and early mortality. PCR amplification from genomic DNA and automated sequencing of the entire coding region (66 exons) and splice junctions detected 77 mutations (85%) in 90 A-T chromosomes. Heteroduplex analysis detected another 42 mutations at the A-T locus. Out of a total of 71 unique mutations, 50 were found only in a single family, and 51 had not been reported previously. Most (58/71, 82%) mutations were frameshift and nonsense mutations that are predicted to cause truncation of the A-T protein; the less common mutation types were missense (9/71, 13%), splicing (3/71, 4%) and one in-frame deletion, 2546 3 (1/71, 1%). The mean survival and height distribution of 134 A-T patients correlated significantly with the specific mutations present in the patients. Patients homozygous for a single truncating mutation, typically near the N-terminal end of the gene, or heterozygous for the in-frame deletion 2546 3, were shorter and had significantly shorter survival than those heterozygous for a splice site or missense mutation, or heterozygous for two truncating mutations. Alterations of the length or amino acid composition of the A-T gene product affect the A-T clinical phenotype in different ways. Mutation analysis at the A-T locus may help estimate the prognosis of A-T patients.. 10817650 17 38 ataxia-telangiectasia Modifier D001260 10817650 72 75 A-T Modifier D001260 10817650 103 124 ataxia-telangiectasia Modifier D001260 10817650 126 129 A-T Modifier D001260 10817650 182 210 autosomal recessive syndrome DiseaseClass D030342 10817650 258 264 cancer DiseaseClass D009369 10817650 266 288 ischemic heart disease SpecificDisease D017202 10817650 465 468 A-T Modifier D001260 10817650 541 544 A-T Modifier D001260 10817650 784 787 A-T Modifier D001260 10817650 970 973 A-T Modifier D001260 10817650 1433 1436 A-T Modifier D001260 10817650 1461 1464 A-T Modifier D001260 10817650 1528 1531 A-T Modifier D001260 10817650 1573 1576 A-T Modifier D001260 7815415|t|Anticipation resulting in elimination of the myotonic dystrophy gene: a follow up study of one extended family. 7815415|a|We have re-examined an extended myotonic dystrophy (DM) family, previously described in 1955, in order to study the long term effects of anticipation in DM and in particular the implications for families affected by this disease. This follow up study provides data on 35 gene carriers and 46 asymptomatic at risk family members in five generations. Clinical anticipation, defined as the cascade of mild, adult, childhood, or congenital disease in subsequent generations, appeared to be a relentless process, occurring in all affected branches of the family. The cascade was found to proceed asynchronously in the different branches, mainly because of an unequal number of generations with mild disease. The transition from the mild to the adult type was associated with transmission through a male parent. Stable transmission of the asymptomatic/mild phenotype showed a female transmission bias. We further examined the extent and causes of gene loss in this pedigree. Gene loss in the patient group was complete, owing to infertility of the male patients with adult onset disease and the fact that mentally retarded patients did not procreate. Out of the 46 at risk subjects in the two youngest generations, only one was found to have a full mutation. This is the only subject who may transmit the gene to the sixth generation. No protomutation carriers were found in the fourth and fifth generations. Therefore it is highly probable that the DM gene will be eliminated from this pedigree within one generation. The high population frequency of DM can at present not be explained by the contribution of asymptomatic cases in the younger generations of known families, but is probably caused by the events in the ancestral generations.. 7815415 45 63 myotonic dystrophy Modifier D009223 7815415 144 162 myotonic dystrophy Modifier D009223 7815415 164 166 DM Modifier D009223 7815415 265 267 DM SpecificDisease D009223 7815415 537 555 congenital disease DiseaseClass D030342 7815415 1135 1146 infertility DiseaseClass D007246 7815415 1211 1228 mentally retarded DiseaseClass D008607 7815415 1556 1558 DM Modifier D009223 7815415 1658 1660 DM SpecificDisease D009223 10571943|t|Exon 9 mutations in the WT1 gene, without influencing KTS splice isoforms, are also responsible for Frasier syndrome. 10571943|a|We report new mutations in exon 9 of the WT1 gene that did not alter the ratio of +/- KTS splice isoforms in two unrelated patients with Frasier syndrome (FS). The mutation of intron 9 inducing defective alternative splicing was reported to be responsible for this syndrome. The mutations found in our cases occurred in the same exon of the WT1 gene as detected in Denys-Drash syndrome (DDS) and could not be explained by the previously proposed mechanism. The results suggest that the two syndromes originate from the same WT1 gene abnormality. From a molecular biological point of view, we concluded that the two diseases were not separable, and that FS should be included as an atypical form of DDS.. 10571943 100 116 Frasier syndrome SpecificDisease D052159 10571943 255 271 Frasier syndrome SpecificDisease D052159 10571943 273 275 FS SpecificDisease D052159 10571943 483 503 Denys-Drash syndrome SpecificDisease D030321 10571943 505 508 DDS SpecificDisease D030321 10571943 642 662 WT1 gene abnormality SpecificDisease D009396 10571943 771 773 FS SpecificDisease D052159 10571943 816 819 DDS SpecificDisease D030321 10500204|t|Deficit of in vivo mitochondrial ATP production in patients with Friedreich ataxia. 10500204|a|Friedreich ataxia (FRDA), the most common of the inherited ataxias, is an autosomal recessive degenerative disorder, characterized clinically by onset before the age of 25 of progressive gait and limb ataxia, absence of deep tendon reflexes, extensor plantar responses, and loss of position and vibration sense in the lower limbs. FRDA is caused by a GAA triplet expansion in the first intron of the FRDA gene on chromosome 9q13 in 97% of patients. The FRDA gene encodes a widely expressed 210-aa protein, frataxin, which is located in mitochondria and is severely reduced in FRDA patients. Frataxin function is still unknown but the knockout of the yeast frataxin homologue gene (YFH1) showed a severe defect of mitochondrial respiration and loss of mtDNA associated with elevated intramitochondrial iron. Here we report in vivo evidence of impaired mitochondrial respiration in skeletal muscle of FRDA patients. Using phosphorus magnetic resonance spectroscopy we demonstrated a maximum rate of muscle mitochondrial ATP production (V (max)) below the normal range in all 12 FRDA patients and a strong negative correlation between mitochondrial V (max) and the number of GAA repeats in the smaller allele. Our results show that FRDA is a nuclear-encoded mitochondrial disorder affecting oxidative phosphorylation and give a rationale for treatments aimed to improve mitochondrial function in this condition.. 10500204 65 82 Friedreich ataxia SpecificDisease D005621 10500204 84 101 Friedreich ataxia SpecificDisease D005621 10500204 103 107 FRDA SpecificDisease D005621 10500204 133 150 inherited ataxias DiseaseClass D013132 10500204 158 199 autosomal recessive degenerative disorder DiseaseClass D019636 10500204 259 291 progressive gait and limb ataxia CompositeMention D020234|D001259 10500204 293 324 absence of deep tendon reflexes DiseaseClass D012021 10500204 326 352 extensor plantar responses DiseaseClass D001405 10500204 358 394 loss of position and vibration sense CompositeMention D053421 10500204 415 419 FRDA SpecificDisease D005621 10500204 484 488 FRDA Modifier D005621 10500204 537 541 FRDA Modifier D005621 10500204 660 664 FRDA Modifier D005621 10500204 983 987 FRDA Modifier D005621 10500204 1160 1164 FRDA Modifier D005621 10500204 1313 1317 FRDA SpecificDisease D005621 10500204 1339 1361 mitochondrial disorder DiseaseClass D028361 2912886|t|Chronic nonspherocytic hemolytic anemia (CNSHA) and glucose 6 phosphate dehydrogenase (G6PD) deficiency in a patient with familial amyloidotic polyneuropathy (FAP). Molecular study of a new variant (G6PD Clinic) with markedly acidic pH optimum. 2912886|a|A new glucose-6-phosphate dehydrogenase (G6PD) variant with severe erythrocytic G6PD deficiency and a unique pH optimum is described in a young patient with chronic nonspherocytic hemolytic anemia (CNSHA) and familial amyloidotic polyneuropathy (FAP). Chronic hemolysis was present in the absence of infections, oxidant drugs or ingestion of faba beans. Residual enzyme activity was about 2. 6% and 63% of normal activity in erythrocytes and leucocytes, respectively. A molecular study using standard methods showed G6PD in the patient to have normal electrophoretic mobility (at pH 7. 0, 8. 0 and 8. 8), normal apparent affinity for substrates (Km, G6P and NADP) and a slightly abnormal utilization of substrate analogues (decreased deamino-NADP and increased 2-deoxyglucose-6-phosphate utilization). Heat stability was found to be markedly decreased (8% of residual activity after 20 min of incubation at 46 degrees C) and a particular characteristic of this enzyme was a biphasic pH curve with a greatly increased activity at low pH. Although molecular characteristics of this variant closely resemble those of G6PD Bangkok and G6PD Duarte, it can be distinguished from these and all other previously reported variants by virtue of its unusual pH curve. Therefore the present variant has been designated G6PD Clinic to distinguish it from other G6PD variants previously described 2912886 0 39 Chronic nonspherocytic hemolytic anemia SpecificDisease D000746 2912886 41 46 CNSHA SpecificDisease D000746 2912886 52 103 glucose 6 phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 2912886 122 157 familial amyloidotic polyneuropathy SpecificDisease D028227 2912886 159 162 FAP SpecificDisease D028227 2912886 312 340 erythrocytic G6PD deficiency SpecificDisease D005955 2912886 402 441 chronic nonspherocytic hemolytic anemia SpecificDisease D000746 2912886 443 448 CNSHA SpecificDisease D000746 2912886 454 489 familial amyloidotic polyneuropathy SpecificDisease D028227 2912886 491 494 FAP SpecificDisease D028227 2912886 497 514 Chronic hemolysis SpecificDisease D006461 3422216|t|Regional localization of polymorphic DNA loci on the proximal long arm of the X chromosome using deletions associated with choroideremia. 3422216|a|In two unrelated families, males have been identified who suffer from choroideremia and at the same time have an interstitial deletion on the proximal long arm of the X chromosome. By high-resolution banding we have characterized the deletion chromosomes as del (X) (q21. 1-q21 1-q21. 33) and del (X) (q21. 2-q21 2-q21. 31) respectively. By Southern blot analysis we have mapped ten different polymorphic DNA loci relative to the position of the deletion and the choroideremia locus TCD. One probe, p31, was shown to cover one of the breakpoints of the smallest deletion. The following order of the loci was suggested by deletion mapping cen-DXS106-DXS72-TCD- (DXYS1/DXYS23/DXYS5) - DXYS2- (DXYS12/DXS3) - (DXS17/DXS101) - Xqter. 3422216 123 136 choroideremia SpecificDisease D015794 3422216 208 221 choroideremia SpecificDisease D015794 3422216 601 614 choroideremia Modifier D015794 2016095|t|A de novo unbalanced reciprocal translocation identified as paternal in origin in the Prader-Willi syndrome. 2016095|a|Interstitial cytogenetic deletions involving the paternally derived chromosome 15q11-13 have been described in patients with the Prader-Willi syndrome (PWS). We report a child with PWS and a de novo unbalanced karyotype -45, XY, -9, -15, + der (9) t (9; 15) (q34; q13). Molecular studies with the DNA probe pML34 confirmed that only a single Prader Willi critical region (PWCR 15q11. 2-q12) copy was present. Hybridisation of patient and parental DNA with the multi-allelic probe CMW1, which maps to pter-15q13, showed that the chromosome involved in the translocation was paternal in origin. This is the first example of a paternally-derived PWCR allele loss caused by an unbalanced translocation that has arisen de novo. 2016095 86 107 Prader-Willi syndrome SpecificDisease D011218 2016095 238 259 Prader-Willi syndrome SpecificDisease D011218 2016095 261 264 PWS SpecificDisease D011218 2016095 290 293 PWS SpecificDisease D011218 2016095 451 463 Prader Willi Modifier D011218 10807385|t|Clinicopathologic features of BRCA-linked and sporadic ovarian cancer. 10807385|a|CONTEXT Most hereditary ovarian cancers are associated with germline mutations in BRCA1 or BRCA2. Attempts to define the clinical significance of BRCA mutation status in ovarian cancer have produced conflicting results, especially regarding survival. OBJECTIVE To determine whether hereditary ovarian cancers have distinct clinical and pathological features compared with sporadic (nonhereditary) ovarian cancers. DESIGN AND SETTING Retrospective cohort study of a consecutive series of 933 ovarian cancers diagnosed and treated at our institution, which is a comprehensive cancer center as designated by the National Cancer Institute, over a 12-year period (December 1986 to August 1998). PATIENTS The study was restricted to patients of Jewish origin because of the ease of BRCA1 and BRCA2 genotyping in this ethnic group. From the 189 patients who identified themselves as Jewish, 88 hereditary cases were identified with the presence of a germline founder mutation in BRCA1 or BRCA2. The remaining 101 cases from the same series not associated with a BRCA mutation and 2 additional groups (Gynecologic Oncology Group protocols 52 and 111) with ovarian cancer from clinical trials (for the survival analysis) were included for comparison. MAIN OUTCOME MEASURES Age at diagnosis, surgical stage, histologic cell type and grade, and surgical outcome; and response to chemotherapy and survival for advanced-stage (II and IV) cases. RESULTS Hereditary cancers were rarely diagnosed before age 40 years and were common after age 60 years, with mean age at diagnosis being significantly younger for BRCA1- vs BRCA2-linked patients (54 vs 62 years; P =. 04). Histology, grade, stage, and success of cytoreductive surgery were similar for hereditary and sporadic cases. The hereditary group had a longer disease-free interval following primary chemotherapy in comparison with the nonhereditary group, with a median time to recurrence of 14 months and 7 months, respectively (P <. 001). Those with hereditary cancers had improved survival compared with the nonhereditary group (P =. 004). For stage III cancers, BRCA mutation status was an independent prognostic variable (P =. 03). CONCLUSIONS Although BRCA-associated hereditary ovarian cancers in this population have surgical and pathological characteristics similar to those of sporadic cancers, advanced-stage hereditary cancer patients survive longer than nonhereditary cancer patients. Age penetrance is greater for BRCA1-linked than for BRCA2-linked cancers in this population. 10807385 30 69 BRCA-linked and sporadic ovarian cancer CompositeMention OMIM:604370|OMIM:612555|OMIM:613399|D010051 10807385 85 111 hereditary ovarian cancers SpecificDisease D061325 10807385 242 256 ovarian cancer SpecificDisease D010051 10807385 355 381 hereditary ovarian cancers SpecificDisease D061325 10807385 445 485 sporadic (nonhereditary) ovarian cancers SpecificDisease D010051 10807385 565 580 ovarian cancers SpecificDisease D010051 10807385 648 654 cancer Modifier D009369 10807385 692 698 Cancer Modifier D009369 10807385 1223 1237 ovarian cancer SpecificDisease D010051 10807385 1517 1535 Hereditary cancers DiseaseClass D009369 10807385 2069 2087 hereditary cancers DiseaseClass D009369 10807385 2164 2181 stage III cancers DiseaseClass D009369 10807385 2276 2318 BRCA-associated hereditary ovarian cancers SpecificDisease OMIM:604370|OMIM:612555|OMIM:613399 10807385 2405 2421 sporadic cancers DiseaseClass D009369 10807385 2423 2455 advanced-stage hereditary cancer Modifier D009386 10807385 2485 2505 nonhereditary cancer Modifier D009369 10807385 2546 2588 BRCA1-linked than for BRCA2-linked cancers CompositeMention OMIM:604370|OMIM:612555 1302008|t|Germline intronic and exonic mutations in the Wilms' tumour gene (WT1) affecting urogenital development. 1302008|a|Denys-Drash syndrome is a rare human developmental disorder affecting the urogenital system and leading to renal failure, intersex disorders and Wilms tumour. In this report, four individuals with this syndrome are described carrying germline point mutations in the Wilms tumour suppressor gene, WT1. Three of these mutations were in the zinc finger domains of WT1. The fourth occurred within intron 9, preventing splicing at one of the alternatively chosen splice donor sites of exon 9 when assayed in vitro. These results provide genetic evidence for distinct functional roles of the WT1 isoforms in urogenital development.. 1302008 46 59 Wilms' tumour Modifier D009396 1302008 105 125 Denys-Drash syndrome SpecificDisease D030321 1302008 142 164 developmental disorder DiseaseClass D002658 1302008 212 225 renal failure DiseaseClass D051437 1302008 227 245 intersex disorders DiseaseClass D012734 1302008 250 262 Wilms tumour SpecificDisease D009396 1302008 371 383 Wilms tumour Modifier D009396 3014348|t|Analysis of deletions in DNA from patients with Becker and Duchenne muscular dystrophy. 3014348|a|Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disorder for which the biochemical defect is as yet unknown. Recently, two cloned segments of human X-chromosome DNA have been described which detect structural alterations within or near the genetic locus responsible for the disorder. Both of these cloned segments were described as tightly linked to the locus and were capable of detecting deletions in the DNA of boys affected with DMD. In an attempt to determine more precisely the occurrence of these deletions within a large population of DMD patients and the accuracy of one of the segments, DXS164 (pERT87), in determining the inheritance of the DMD X chromosome, the subclones 1, 8 and 15 were made available to many investigators throughout the world. Here we describe the combined results of more than 20 research laboratories with respect to the occurrence of deletions at the DXS164 locus in DNA samples isolated from patients with DMD and Becker muscular dystrophy (BMD). The results indicate that the DXS164 locus apparently recombines with DMD 5% of the time, but is probably located between independent sites of mutation which yield DMD. The breakpoints of some deletions are delineated within the DXS164 locus, and it is evident that the deletions at the DMD locus are frequent and extremely large.. 3014348 48 86 Becker and Duchenne muscular dystrophy CompositeMention D020388|C537666 3014348 88 115 Duchenne muscular dystrophy SpecificDisease D020388 3014348 117 120 DMD SpecificDisease D020388 3014348 128 163 X-linked recessive genetic disorder DiseaseClass D040181 3014348 540 543 DMD SpecificDisease D020388 3014348 650 653 DMD Modifier D020388 3014348 759 762 DMD Modifier D020388 3014348 1050 1053 DMD SpecificDisease D020388 3014348 1058 1083 Becker muscular dystrophy SpecificDisease C537666 3014348 1085 1088 BMD SpecificDisease C537666 3014348 1161 1164 DMD SpecificDisease D020388 3014348 1255 1258 DMD SpecificDisease D020388 3014348 1378 1381 DMD Modifier D020388 7802009|t|Additional case of female monozygotic twins discordant for the clinical manifestations of Duchenne muscular dystrophy due to opposite X-chromosome inactivation. 7802009|a|A pair of female monozygotic (MZ) twins, heterozygous carriers for a deletion in the DMD gene and discordant for the clinical manifestations of Duchenne muscular dystrophy, were analyzed by molecular studies, in situ hybridization, and methylation pattern of X chromosomes to search for opposite X inactivation as an explanation of their clinical discordance. Results in lymphocytes and skin fibroblast cell lines suggest a partial mirror inactivation with the normal X chromosome preferentially active in the unaffected twin, and the maternal deleted X chromosome preferentially active in the affected twin. A review shows that MZ female twins discordant for X-linked diseases are not uncommon. Twinning and X inactivation may be interrelated and could explain the female twins discordant for X-linked traits.. 7802009 90 117 Duchenne muscular dystrophy SpecificDisease D020388 7802009 246 249 DMD Modifier D020388 7802009 305 332 Duchenne muscular dystrophy SpecificDisease D020388 7802009 821 838 X-linked diseases DiseaseClass D040181 1127526|t|Analbuminemia in a neonate. 1127526|a|A small-for-gestational-age infant, found to have analbuminemia in the neonatal period, is reported and the twelve cases recorded in the world literature are reviewed. Patients lacking this serum protein are essentially asymptomatic, apart from minimal ankle edema and ease of fatigue. Apparent compensatory mechanisms which come into play when serum albumin is low include prolonged half-life of albumin and transferrin, an increase in serum globulins, beta lipoprotein, and glycoproteins, arterial hypotension with reduced capillary hydrostatic pressure, and the ability to respond with rapid sodium and chloride diuresis in response to small volume changes. Examination of plasma amino acids, an investigation not previously reported, revealed an extremely low plasma tryptophan level, a finding which may be important in view of the role of tryptophan in albumin synthesis.. 1127526 0 13 Analbuminemia SpecificDisease OMIM:103600 1127526 78 91 analbuminemia SpecificDisease OMIM:103600 1127526 281 292 ankle edema DiseaseClass D016512 1127526 297 312 ease of fatigue DiseaseClass D005221 1127526 519 539 arterial hypotension DiseaseClass D007022 7857677|t|Homozygous presence of the crossover (fusion gene) mutation identified in a type II Gaucher disease fetus: is this analogous to the Gaucher knock-out mouse model? 7857677|a|Gaucher disease (GD) is an inherited deficiency of beta-glucocerebrosidase (EC 3. 1. 2. 45, gene symbol GBA). In type I GD, the CNS is not involved (nonneuronopathic), whereas in type II GD (acute neuronopathic) CNS involvement is early and rapidly progressive, while in type III GD (subacute neuronopathic) CNS involvement occurs later and is slowly progressive. The T6433C (L444P) substitution is prevalent in type GD II. It may occur alone as a single base-pair mutation but often is found as part of a complex allele containing additional GBA nucleotide substitutions, G6468C (A456P) and G6482C (V460V), without (recNciI) or with (recTL) G5957C (D409H). This complex allele is presumed to have formed by recombination (crossover, fusion) of the structural gene with the pseudogene, which contains the mutated sequences. Two complex alleles have never been demonstrated to coexist in any individual. We devised a selective PCR method for the specific amplification of the normal and/or fusion gene. Using this procedure we demonstrated the fusion gene in homozygous form for the first time, in a Macedonian/Ashkenazi Jewish GD type II fetus. Both parents were carriers of the recombination. This was confirmed by direct sequence analysis. A previous conceptus in this family was stillborn at 36 weeks, with features of severe type II GD. Neonates showing a severe clinical phenotype, analogous to the early neonatal lethal disease occurring in mice homozygous for a null allele produced by targeted disruption of GBA, have been described elsewhere, but the specific mutations in these cases have not yet been characterized. (ABSTRACT TRUNCATED AT 250 WORDS) 7857677 76 99 type II Gaucher disease Modifier D005776 7857677 163 178 Gaucher disease SpecificDisease D005776 7857677 180 182 GD SpecificDisease D005776 7857677 200 237 deficiency of beta-glucocerebrosidase SpecificDisease D005776 7857677 276 285 type I GD SpecificDisease D005776 7857677 342 352 type II GD SpecificDisease D005776 7857677 434 445 type III GD SpecificDisease D005776 7857677 575 585 type GD II SpecificDisease D005776 7857677 1290 1300 GD type II Modifier D005776 7857677 1445 1454 stillborn DiseaseClass D005313 7857677 1492 1502 type II GD SpecificDisease D005776 10403837|t|Decrease in GTP cyclohydrolase I gene expression caused by inactivation of one allele in hereditary progressive dystonia with marked diurnal fluctuation. 10403837|a|Hereditary progressive dystonia with marked diurnal fluctuation (HPD; dopa-responsive dystonia, DRD) have been recently found to be caused by a genetic defect in the GTP cyclohydrolase I (GCH1) gene. In this study, we quantified the mRNA level of GCH1 in phytohemagglutinin (PHA) -stimulated mononuclear blood cells from one Japanese family that do not have a mutation in the coding region or splice junctions of the gene. The results showed that the amounts of the GCH1 mRNA were decreased to about 40% of the normal level in both patients and carriers. In addition, we found that the GCH1 mRNA was transcribed from only one allele, indicating that the other allele was in an inactive state. These results suggest that some novel mutations should exist on one of the alleles in some unknown region of the GCH1 gene, and may decrease the GCH1 mRNA causing the HPD/DRD symptoms.. 10403837 89 120 hereditary progressive dystonia SpecificDisease D020821 10403837 154 185 Hereditary progressive dystonia SpecificDisease D020821 10403837 219 222 HPD SpecificDisease D020821 10403837 224 248 dopa-responsive dystonia SpecificDisease C538007 10403837 250 253 DRD SpecificDisease C538007 10403837 298 312 genetic defect DiseaseClass D030342 10403837 1014 1017 HPD Modifier D020821 10403837 1018 1021 DRD Modifier C538007 10447259|t|Novel mutations in the Wiskott-Aldrich syndrome protein gene and their effects on transcriptional, translational, and clinical phenotypes. 10447259|a|Wiskott-Aldrich syndrome (WAS) is an X-linked recessive immunodeficiency characterized by thrombocytopenia, eczema, and recurrent infections, and caused by mutations in the WAS protein (WASP) gene. WASP contains several functional domains through which it interacts with proteins involved in intracellular signaling and regulation of the actin cytoskeleton. In this report, 17 WASP gene mutations were identified, 12 of which are novel. DNA of affected males and obligate carriers was PCR amplified and analyzed by SSCA, heteroduplex analysis, and direct sequencing. The effects of the mutations at the mRNA and protein level were ascertained by RT-PCR and Western blot analyses. All missense mutations were located in exons 1-4. Most of the nonsense, frameshift and splice site mutations were found in exons 6-11. Mutations that alter splice sites led to the synthesis of several types of mRNAs, a fraction of which represented the normally spliced product. The presence of normally spliced transcripts was correlated with a milder phenotype. When one such case was studied by Western blotting, reduced amounts of normal-size WASP were present. In other cases as well, a correlation was found between the amount of normal or mutant WASP present and the phenotypes of the affected individuals. No protein was detected in two individuals with severe WAS. Reduced levels of a normal-size WASP with a missense mutation were seen in two individuals with XLT. It is concluded that mutation analysis at the DNA level is not sufficient for predicting clinical course. Studies at the transcript and protein level are needed for a better assessment.. 10447259 23 47 Wiskott-Aldrich syndrome Modifier D014923 10447259 139 163 Wiskott-Aldrich syndrome SpecificDisease D014923 10447259 165 168 WAS SpecificDisease D014923 10447259 176 211 X-linked recessive immunodeficiency DiseaseClass D053632 10447259 229 245 thrombocytopenia DiseaseClass D013921 10447259 247 253 eczema DiseaseClass D004485 10447259 312 315 WAS Modifier D014923 10447259 1488 1491 WAS SpecificDisease D014923 10447259 1589 1592 XLT SpecificDisease OMIM:313900 511159|t|Incidence and characteristics of glucose-6-phosphate dehydrogenase variants in Japan. 511159|a|A total of 3000 men living in Yamaguchi were screened for glucose-6-phosphate dehydrogenase (G6PD) deficiency using Beutlers spot test and three types of starch gel electrophoresis. These electrophoresis used a phosphate buffer system at pH 7. 0, a TRIS-EDTA-borate buffer system at pH 8. 6, and a TRIS-hydrochloride buffer system at pH 8.. Fifteen G6PD-deficient variants were found at the rate of 0. 5% and classified into four groups. As new variants, G6PD Konan, Kamiube, and Kiwa were identified. These three variants had a mild to moderate G6PD deficiency and were not associated with any clinical signs. G6PD Konan had fast electrophoretic mobility as compared with normal levels, G6PD Kiwa had slightly elevated electrophoretic mobility, and G6PD Kamiube had normal electrophoretic mobility. These three variants had normal levels of Km G6P, Km NADP, and Ki NADPH, normal utilizations of both 2-deoxy-G6P and deamino-NAPD, normal heat stability, and a normal pH curve. The other variant was G6PD Ube, which we had previously found in Yamaguchi (Nakashima et al., 1977). One boy with G6PD Ube was Korean 511159 144 195 glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 511159 435 449 G6PD-deficient Modifier D005955 511159 632 647 G6PD deficiency SpecificDisease D005955 8266996|t|Difference in methylation patterns within the D15S9 region of chromosome 15q11-13 in first cousins with Angelman syndrome and Prader-Willi syndrome. 8266996|a|Abnormalities of chromosome region 15q11-13 are associated with Angelman syndrome (AS) and Prader-Willi syndrome (PWS). Differences between the methylation patterns of the region of chromosome 15q11-13 which hybridizes to the highly conserved DNA, DN34, in normal individuals and in patients with AS and PWS have been described. We report on a family in which first cousins are affected by AS and PWS as a result of a familial paracentric inversion of 15q11-q13. The results of the studies on this family demonstrate the differences in the methylation patterns in the 2 conditions and the phenomenon of genomic imprinting, whereby genetic information is expressed differently dependent on the parent of origin.. 8266996 104 121 Angelman syndrome SpecificDisease D017204 8266996 126 147 Prader-Willi syndrome SpecificDisease D011218 8266996 213 230 Angelman syndrome SpecificDisease D017204 8266996 232 234 AS SpecificDisease D017204 8266996 240 261 Prader-Willi syndrome SpecificDisease D011218 8266996 263 266 PWS SpecificDisease D011218 8266996 446 448 AS SpecificDisease D017204 8266996 453 456 PWS SpecificDisease D011218 8266996 539 541 AS SpecificDisease D017204 8266996 546 549 PWS SpecificDisease D011218 1307245|t|Mutations in the candidate gene for Norrie disease. 1307245|a|Recently, we and others have isolated a candidate gene for X linked Norrie disease (ND) which was found to be deleted or disrupted in several patients. As a prerequisite for the identification of point mutations in the ND gene we have established the exon-intron structure of this gene. In 17 unrelated patients and 15 controls, PCR products derived from the promoter region, exons 1 and 2 as well as the coding part of exon 3 were analysed with the single strand conformation polymorphism (SSCP) technique. In 12 patients altered PCR fragments were detected which were studied in detail by direct sequencing. Eleven different mutations were found, and all but one are likely to give rise to significant structural changes in the predicted protein. These findings, and the absence of functionally relevant base changes in healthy controls, emphasize the causal role of this candidate gene in Norrie disease and pave the way for reliable diagnosis and carrier detection.. 1307245 36 50 Norrie disease SpecificDisease C537849 1307245 111 134 X linked Norrie disease SpecificDisease C537849 1307245 136 138 ND SpecificDisease C537849 1307245 271 273 ND Modifier C537849 1307245 944 958 Norrie disease SpecificDisease C537849 7874117|t|A single amino acid substitution (G103D) in the type II collagen triple helix produces Kniest dysplasia. 7874117|a|Kniest dysplasia is a moderately severe chondrodysplasia phenotype that results from mutations in the gene for type II collagen, COL2A1. Characteristics of the disorder include a short trunk and extremities, mid-face hypoplasia, cleft palate, myopia, retinal detachment, and hearing loss. Recently, deletions of all or part of exon 12 have been identified in individuals with Kniest dysplasia, suggesting that mutations within this region of the protein may primarily result in the Kniest dysplasia phenotype. We used SSCP to analyze an amplified genomic DNA fragment containing exon 12 from seven individuals with Kniest dysplasia. An abnormality was identified in one patient. DNA sequence analysis demonstrated that the patient was heterozygous for a G to A transition that implied substitution of glycine103 of the triple helical domain by aspartate. The mutation was not observed in DNA from either of the clinically unaffected parents of the proband. Protein microsequencing demonstrated expression of the abnormal allele in cartilage. These data demonstrate that point mutations which result in single amino acid substitutions can produce Kniest dysplasia and further support the hypothesis that alteration of a domain, which includes the region encoded by exon 12, in the type II collagen protein leads to this disorder.. 7874117 87 103 Kniest dysplasia SpecificDisease C537207 7874117 105 121 Kniest dysplasia SpecificDisease C537207 7874117 145 161 chondrodysplasia Modifier D010009 7874117 284 311 short trunk and extremities SpecificDisease D006130 7874117 313 332 mid-face hypoplasia DiseaseClass D019767 7874117 334 346 cleft palate SpecificDisease D002972 7874117 348 354 myopia DiseaseClass D009216 7874117 356 374 retinal detachment SpecificDisease D012163 7874117 380 392 hearing loss DiseaseClass D034381 7874117 481 497 Kniest dysplasia SpecificDisease C537207 7874117 587 603 Kniest dysplasia Modifier C537207 7874117 720 736 Kniest dysplasia SpecificDisease C537207 7874117 1251 1267 Kniest dysplasia SpecificDisease C537207 10434119|t|Linkage analysis of 5 novel van der Woude syndrome kindreds to 1q32-q41 markers further supports locus homogeneity of the disease trait. 10434119|a|van der Woude syndrome (vWS, MIM 119300) is a rare autosomal dominant clefting condition with cardinal features of mucous cysts (lower-lip pits) and clefts to the lip and/or palate. The vWS gene has been assigned to a locus in 1q32-q41 by linkage analysis and physical mapping. We have investigated 5 novel vWS families through probands attended for cleft lip and/or palate repair at the Department of Maxillofacial Surgery of Hopital Trousseau, Paris, in order to tentatively refine the genetic map of the vWS region in 1q32-q41 and possibly identify unlinked pedigrees. Linkage analysis was carried out to 6 microsatellite markers (D1S249, D1S425, D1S491, D1S205, D1S414, D1S425), yielding a maximum cumulative LOD score of Z = 3. 27 at theta = 0. 00 for D1S245. The innermost four markers were found to be tightly linked to one another, with no evidence for recombination. Our results support linkage of vWS within a region of tightly linked markers and do not favour locus heterogeneity of the disease trait. 10434119 28 50 van der Woude syndrome Modifier C536528 10434119 137 159 van der Woude syndrome SpecificDisease C536528 10434119 161 164 vWS SpecificDisease C536528 10434119 188 225 autosomal dominant clefting condition DiseaseClass C536528 10434119 252 264 mucous cysts SpecificDisease D009078 10434119 266 280 lower-lip pits SpecificDisease C536528 10434119 286 317 clefts to the lip and/or palate CompositeMention D002971|D002972 10434119 323 326 vWS Modifier C536528 10434119 444 447 vWS Modifier C536528 10434119 487 510 cleft lip and/or palate Modifier D002971|D002972 10434119 644 647 vWS Modifier C536528 10434119 1044 1047 vWS SpecificDisease C536528 3578281|t|Hypopigmentation in the Prader-Willi syndrome. 3578281|a|Cutaneous and ocular pigmentation were evaluated in 29 individuals with the Prader-Willi syndrome (PWS). Criteria for hypopigmentation included the presence of type I or II skin, the lightest skin type in the family by history, and iris translucency on globe transillumination. On the basis of these criteria, 48% of the PWS individuals were hypopigmented. The presence of hypopigmentation correlated with a small interstitial deletion on the proximal long arm of chromosome 15; however, this deletion was also found in individuals who did not meet the full criteria for hypopigmentation. Hairbulb tyrosinase activity and glutathione content, as well as urine cysteinyldopa excretion, were low in PWS individuals with and without hypopigmentation and did not separate these two groups. We conclude that hypopigmentation is found in a significant proportion of individuals with PWS and that the hypopigmentation may be associated with a deletion of the long arm of chromosome 15. The mechanism for the hypopigmentation is unknown.. 3578281 0 16 Hypopigmentation DiseaseClass D017496 3578281 24 45 Prader-Willi syndrome SpecificDisease D011218 3578281 47 80 Cutaneous and ocular pigmentation CompositeMention D010859 3578281 123 144 Prader-Willi syndrome SpecificDisease D011218 3578281 146 149 PWS SpecificDisease D011218 3578281 165 181 hypopigmentation DiseaseClass D017496 3578281 368 371 PWS Modifier D011218 3578281 389 402 hypopigmented Modifier D017496 3578281 420 436 hypopigmentation DiseaseClass D017496 3578281 618 634 hypopigmentation DiseaseClass D017496 3578281 744 747 PWS Modifier D011218 3578281 777 793 hypopigmentation DiseaseClass D017496 3578281 850 866 hypopigmentation DiseaseClass D017496 3578281 924 927 PWS SpecificDisease D011218 3578281 941 957 hypopigmentation DiseaseClass D017496 3578281 1048 1064 hypopigmentation DiseaseClass D017496 495634|t|Deficiency of the fifth component of complement in human subjects. Clinical, genetic and immunologic studies in a large kindred. 495634|a|The discovery of a large kindred with a heritable deficiency of the fifth component of complement (C5) has permitted the accumulation of new clinical, genetic and immunologic data concerning the role of C5 in human subjects. The proband, who has had nine episodes of disseminated gonococcal infection, has a hemolytic C5 level of approximately 0. 5 per cent of normal. No C5 protein was detectable, but low levels of functional C5 activity could be found using a sensitive bactericidal assay. The probands twin as well as another sister also had extremely low levels of hemolytic C5 (approximately 0. 5 per cent normal), but both these subjects have been healthy. Hemolytic complement and bacteriolytic activity could be restored by the addition of purified C5. No chemotactic activity for polymorphonuclear leukocytes could be generated in the C5-deficient serums upon activation of either the classic or alternative pathways, again demonstrating the importance of C5 in human subjects for the production of chemotactic factors. The chemotactic responsiveness of the patients polymorphonuclear leukocytes and monocytes to preformed chemotactic factors was not depressed. Twenty-two of 32 other family members from three generations had depressed whole hemolytic complement levels. In 19 of 30 family members, levels of hemolytic C5 ranged from 13 to 64 per cent of normal. No linkage for C5 deficiency and the A or B loci of the major histocompatibility complex could be found. These data suggest an autosomal codominant mode of inheritance of C5 deficiency. Deficiency of C5 is compatible with good health, but it can be associated with repeated disseminated gonococcal infection 495634 0 47 Deficiency of the fifth component of complement SpecificDisease OMIM:609536 495634 179 226 deficiency of the fifth component of complement SpecificDisease OMIM:609536 495634 409 429 gonococcal infection SpecificDisease D006069 495634 974 986 C5-deficient Modifier OMIM:609536 495634 1518 1531 C5 deficiency SpecificDisease OMIM:609536 495634 1674 1687 C5 deficiency SpecificDisease OMIM:609536 495634 1689 1705 Deficiency of C5 SpecificDisease OMIM:609536 495634 1790 1810 gonococcal infection SpecificDisease D006069 8533762|t|A new glucose-6-phosphate dehydrogenase variant, G6PD Orissa (44 Ala-->Gly), is the major polymorphic variant in tribal populations in India. 8533762|a|Deficiency of glucose-6-phosphate dehydrogenase (G6PD) is usually found at high frequencies in areas of the world where malaria has been endemic. The frequency and genetic basis of G6PD deficiency have been studied in Africa, around the Mediterranean, and in the Far East, but little such information is available about the situation in India. To determine the extent of heterogeneity of G6PD, we have studied several different Indian populations by screening for G6PD deficiency, followed by molecular analysis of deficient alleles. The frequency of G6PD deficiency varies between 3% and 15% in different tribal and urban groups. Remarkably, a previously unreported deficient variant, G6PD Orissa (44 Ala-- > Gly), is responsible for most of the G6PD deficiency in tribal Indian populations but is not found in urban populations, where most of the G6PD deficiency is due to the G6PD Mediterranean (188 Ser-- > Phe) variant. The KmNADP of G6PD Orissa is fivefold higher than that of the normal enzyme. This may be due to the fact that the alanine residue that is replaced by glycine is part of a putative coenzyme-binding site.. 8533762 142 189 Deficiency of glucose-6-phosphate dehydrogenase SpecificDisease D005955 8533762 262 269 malaria SpecificDisease D008288 8533762 323 338 G6PD deficiency SpecificDisease D005955 8533762 606 621 G6PD deficiency SpecificDisease D005955 8533762 693 708 G6PD deficiency SpecificDisease D005955 8533762 889 904 G6PD deficiency SpecificDisease D005955 8533762 991 1006 G6PD deficiency SpecificDisease D005955 1655284|t|Germline mutations in the Wilms' tumor suppressor gene are associated with abnormal urogenital development in Denys-Drash syndrome. 1655284|a|Denys-Drash syndrome is a rare human condition in which severe urogenital aberrations result in renal failure, pseudohermaphroditism, and Wilms tumor (nephroblastoma). To investigate its possible role, we have analyzed the coding exons of the Wilms tumor suppressor gene (WT1) for germline mutations. In ten independent cases of Denys-Drash syndrome, point mutations in the zinc finger domains of one WT1 gene copy were found. Nine of these mutations are found within exon 9 (zinc finger III); the remaining mutation is in exon 8 (zinc finger II). These mutations directly affect DNA sequence recognition. In two families analyzed, the mutations were shown to arise de novo. Wilms tumors from three individuals and one juvenile granulosa cell tumor demonstrate reduction to homozygosity for the mutated WT1 allele. Our results provide evidence of a direct role for WT1 in Denys-Drash syndrome and thus urogenital system development.. 1655284 26 38 Wilms' tumor Modifier D009396 1655284 110 130 Denys-Drash syndrome SpecificDisease D030321 1655284 132 152 Denys-Drash syndrome SpecificDisease D030321 1655284 195 217 urogenital aberrations DiseaseClass D014564 1655284 228 241 renal failure SpecificDisease D051437 1655284 243 264 pseudohermaphroditism SpecificDisease D012734 1655284 270 281 Wilms tumor SpecificDisease D009396 1655284 283 297 nephroblastoma SpecificDisease D009396 1655284 375 386 Wilms tumor Modifier D009396 1655284 461 481 Denys-Drash syndrome SpecificDisease D030321 1655284 807 819 Wilms tumors SpecificDisease D009396 1655284 875 880 tumor DiseaseClass D009369 1655284 1004 1024 Denys-Drash syndrome SpecificDisease D030321 10366443|t|Molecular basis of feline beta-glucuronidase deficiency: an animal model of mucopolysaccharidosis VII. 10366443|a|A family of domestic cats was found that exhibited clinical and biochemical abnormalities consistent with mucopolysaccharidosis VII, an autosomal recessive lysosomal storage disorder caused by beta-glucuronidase deficiency. beta-Glucuronidase activity was undetectable in affected cat fibroblasts and restored by retroviral gene transfer of rat beta-glucuronidase cDNA. beta-Glucuronidase mRNA was normal in affected cat testis by Northern blot analysis. Normal feline beta-glucuronidase cDNA was cloned and characterized, and amplified from affected cat fibroblasts by reverse transcription coupled polymerase chain reaction. There was a G-to-A transition in the affected cat cDNA that predicted an E351K substitution, destroyed a BssSI site, and eliminated GUSB enzymatic activity in expression studies. Multiple species comparison and the crystal structure of human beta-glucuronidase indicated that E351 is a highly conserved residue most likely essential in maintenance of the enzymes conformation. BssSI digestion of polymerase chain reaction products amplified from genomic DNA indicated that affected cats were homozygous and cats with half-normal beta-glucuronidase activity were heterozygous for the missense mutation. Carriers identified in this manner produced affected kittens in prospective breedings, and a feline MPS VII breeding colony has been established.. 10366443 26 55 beta-glucuronidase deficiency SpecificDisease D016538 10366443 76 101 mucopolysaccharidosis VII SpecificDisease D016538 10366443 209 234 mucopolysaccharidosis VII SpecificDisease D016538 10366443 239 285 autosomal recessive lysosomal storage disorder DiseaseClass D016464 10366443 296 325 beta-glucuronidase deficiency SpecificDisease D016538 10366443 1432 1439 MPS VII Modifier D016538 10581027|t|Loss-of-function mutations in the cathepsin C gene result in periodontal disease and palmoplantar keratosis. 10581027|a|Papillon-Lefevre syndrome, or keratosis palmoplantaris with periodontopathia (PLS, MIM 245000), is an autosomal recessive disorder that is mainly ascertained by dentists because of the severe periodontitis that afflicts patients. Both the deciduous and permanent dentitions are affected, resulting in premature tooth loss. Palmoplantar keratosis, varying from mild psoriasiform scaly skin to overt hyperkeratosis, typically develops within the first three years of life. Keratosis also affects other sites such as elbows and knees. Most PLS patients display both periodontitis and hyperkeratosis. Some patients have only palmoplantar keratosis or periodontitis, and in rare individuals the periodontitis is mild and of late onset. The PLS locus has been mapped to chromosome 11q14-q21 (refs 7, 8, 9). Using homozygosity mapping in eight small consanguineous families, we have narrowed the candidate region to a 1. 2-cM interval between D11S4082 and D11S931. The gene (CTSC) encoding the lysosomal protease cathepsin C (or dipeptidyl aminopeptidase I) lies within this interval. We defined the genomic structure of CTSC and found mutations in all eight families. In two of these families we used a functional assay to demonstrate an almost total loss of cathepsin C activity in PLS patients and reduced activity in obligate carriers. 10581027 61 80 periodontal disease SpecificDisease D010510 10581027 85 107 palmoplantar keratosis DiseaseClass D007645 10581027 109 134 Papillon-Lefevre syndrome SpecificDisease D010214 10581027 139 163 keratosis palmoplantaris DiseaseClass D007645 10581027 169 185 periodontopathia SpecificDisease D010510 10581027 187 190 PLS SpecificDisease D010214 10581027 211 239 autosomal recessive disorder DiseaseClass D030342 10581027 301 314 periodontitis SpecificDisease D010518 10581027 420 430 tooth loss SpecificDisease D016388 10581027 432 454 Palmoplantar keratosis DiseaseClass D007645 10581027 507 521 hyperkeratosis SpecificDisease D017488 10581027 580 589 Keratosis SpecificDisease D007642 10581027 646 649 PLS Modifier D010214 10581027 672 685 periodontitis SpecificDisease D010518 10581027 690 704 hyperkeratosis SpecificDisease D017488 10581027 730 752 palmoplantar keratosis DiseaseClass D007645 10581027 756 769 periodontitis SpecificDisease D010518 10581027 799 812 periodontitis SpecificDisease D010518 10581027 1386 1389 PLS Modifier D010214 8563759|t|Brca1 deficiency results in early embryonic lethality characterized by neuroepithelial abnormalities. 8563759|a|The breast and ovarian cancer susceptibility gene, BRCA1, has been cloned and shown to encode a zinc-finger protein of unknown function. Mutations in BRCA1 account for at least 80% of families with both breast and ovarian cancer, as well as some non-familial sporadic ovarian cancers. The loss of wild-type BRCA1 in tumours of individuals carrying one nonfunctional BRCA1 allele suggests that BRCA1 encodes a tumour suppressor that may inhibit the proliferation of mammary epithelial cells. To examine the role of BRCA1 in normal tissue growth and differentiation, and to generate a potential model for the cancer susceptibility associated with loss of BRCA1 function, we have created a mouse line carrying a mutation in one Brca1 allele. Analysis of mice homozygous for the mutant allele indicate that Brca1 is critical for normal development, as these mice died in utero between 10 and 13 days of gestation (E10-E13). Abnormalities in Brca1-deficient embryos were most evident in the neural tube, with 40% of the embryos presenting with varying degrees of spina bifida and anencephaly. In addition, the neuroepithelium in Brca1-deficient embryos appeared disorganized, with signs of both rapid proliferation and excessive cell death.. 8563759 0 16 Brca1 deficiency SpecificDisease OMIM:604370 8563759 34 53 embryonic lethality DiseaseClass D020964 8563759 71 100 neuroepithelial abnormalities DiseaseClass D018302 8563759 106 131 breast and ovarian cancer Modifier D061325 8563759 305 330 breast and ovarian cancer CompositeMention D061325 8563759 370 385 ovarian cancers SpecificDisease D010051 8563759 418 425 tumours DiseaseClass D009369 8563759 511 517 tumour Modifier D009369 8563759 709 715 cancer Modifier D009369 8563759 1039 1054 Brca1-deficient Modifier OMIM:604370 8563759 1160 1172 spina bifida SpecificDisease D016135 8563759 1177 1188 anencephaly SpecificDisease D000757 8563759 1226 1241 Brca1-deficient Modifier OMIM:604370 8301658|t|X linked recessive thrombocytopenia. 8301658|a|A Saudi Arab boy presented in early childhood with thrombocytopenia, morphologically large and normal sized platelets, increased mean platelet volume, and a hypermegakaryocytic bone marrow. There was no clinical and laboratory evidence of any significant immunological abnormalities. Similar findings in two other brothers suggested strongly that they were all suffering from an X linked recessive thrombocytopenic disorder. Results of DNA analysis with the probe M27 beta are consistent with X linkage and indicate also that the locus of the relevant gene lies close to or is identical to the locus of the gene for the Wiskott-Aldrich syndrome (WAS). Because of various features which include the presence of large and normal sized platelets (rather than small platelets) and freedom from significant immune deficiencies, it is likely that the X linked recessive thrombocytopenia in this family is an isolated entity quite distinct from the classical WAS phenotype. However, a modified expression of the WAS gene producing a mild phenotypic variant cannot be excluded entirely.. 8301658 0 35 X linked recessive thrombocytopenia SpecificDisease OMIM:313900 8301658 88 104 thrombocytopenia SpecificDisease D013921 8301658 292 319 immunological abnormalities DiseaseClass D007154 8301658 416 460 X linked recessive thrombocytopenic disorder SpecificDisease OMIM:313900 8301658 657 681 Wiskott-Aldrich syndrome SpecificDisease D014923 8301658 683 686 WAS SpecificDisease D014923 8301658 839 858 immune deficiencies DiseaseClass D007154 8301658 882 917 X linked recessive thrombocytopenia SpecificDisease OMIM:313900 8301658 989 992 WAS Modifier D014923 8301658 1042 1045 WAS Modifier D014923 7315872|t|Genetics of cerebrotendinous xanthomatosis (CTX): an autosomal recessive trait with high gene frequency in Sephardim of Moroccan origin. 7315872|a|We described 6 patients (from 3 families) affected with cerebrotendinous xanthomatosis (CTX). All are Sephardic Jews of Moroccan extraction. In view of the small number of CTX patients diagnosed in the world (a total of 50 including our 6 patients), we are probably dealing with an ethnic subgroup with a high CTX gene frequency, which we have estimated to be 1/108. Since there are differences in expression in this disease, we recommend cholestanol study in cases of undiagnosed cataract or tendinous xanthomas in childhood or early adolescence. The diagnosis in CTX is important not only for genetic counseling, but also in veiw of possible treatment.. 7315872 12 42 cerebrotendinous xanthomatosis SpecificDisease D019294 7315872 44 47 CTX SpecificDisease D019294 7315872 193 223 cerebrotendinous xanthomatosis SpecificDisease D019294 7315872 225 228 CTX SpecificDisease D019294 7315872 309 312 CTX Modifier D019294 7315872 618 626 cataract SpecificDisease D002386 7315872 630 649 tendinous xanthomas SpecificDisease D014973 7315872 702 705 CTX SpecificDisease D019294 8408659|t|A missense mutation in the cholesteryl ester transfer protein gene with possible dominant effects on plasma high density lipoproteins. 8408659|a|Plasma HDL are a negative risk factor for atherosclerosis. Cholesteryl ester transfer protein (CETP; 476 amino acids) transfers cholesteryl ester from HDL to other lipoproteins. Subjects with homozygous CETP deficiency caused by a gene splicing defect have markedly elevated HDL; however, heterozygotes have only mild increases in HDL. We describe two probands with a CETP missense mutation (442 D G). Although heterozygous, they have threefold increases in HDL concentration and markedly decreased plasma CETP mass and activity, suggesting that the mutation has dominant effects on CETP and HDL in vivo. Cellular expression of mutant cDNA results in secretion of only 30% of wild type CETP activity. Moreover, coexpression of wild type and mutant cDNAs leads to inhibition of wild type secretion and activity. The dominant effects of the CETP missense mutation during cellular expression probably explains why the probands have markedly increased HDL in the heterozygous state, and suggests that the active molecular species of CETP may be multimeric.. 8408659 177 192 atherosclerosis SpecificDisease D050197 8408659 338 353 CETP deficiency SpecificDisease OMIM:143470 2571579|t|Huntington disease: no evidence for locus heterogeneity. 2571579|a|A total of 63 families with Huntington disease (HD) were examined for linkage between HD and G8 (D4S10). The families included 57 Caucasian, four Black American, and two Japanese. The combined maximum lod score was 87. 69 at theta = 0. 04 (99% confidence interval 0. 018-0. 071). The maximum frequency of recombination was 0. 03 in males and 0. 05 in females. Fifty-seven families gave positive lod scores; five small families gave mildly negative lod scores. The maximum likelihood estimate of alpha, the proportion of linked loci, was 1. 0 with a lower 99% confidence interval of 0. 88. These data suggest that there is only one HD locus, although a second rare locus cannot be ruled out. 2571579 0 18 Huntington disease SpecificDisease D006816 2571579 85 103 Huntington disease SpecificDisease D006816 2571579 105 107 HD SpecificDisease D006816 2571579 143 145 HD SpecificDisease D006816 2571579 688 690 HD Modifier D006816 3258663|t|Homozygous and heterozygous deletions of the von Willebrand factor gene in patients and carriers of severe von Willebrand disease. 3258663|a|Severe von Willebrand disease is characterized by undetectable or trace quantities of von Willebrand factor in plasma and tissue stores. We have studied the genomic DNA of 10 affected individuals from six families with this disorder using probes from the 5 and 3 ends of the vWF cDNA and with a probe extending from the 5 end into the central region. Southern blots of restriction endonuclease digests and gene dosage analysis measurements carried out with quantitative slot blots of undigested genomic DNA separated these patients into three groups. The first group consisted of a family with complete homozygous deletions of the vWF gene in the four probands. Gene dosage analysis was consistent with heterozygous deletions in both of the asymptomatic parents and four asymptomatic siblings of this kindred (P less than 0. 01). The second group was comprised of a family in which there was a complete heterozygous deletion of the vWF gene in the proband and one asymptomatic parent, suggesting that a different type of genetic abnormality was inherited from the other parent. Thus, the patient appeared to be doubly heterozygous for interacting genetic abnormalities affecting vWF expression. In the third group, no gene deletions could be detected. Alloantibodies developed only in the kindred with homozygous deletions. These techniques should prove useful in identifying carriers of severe von Willebrand disease and also in defining patients predictably at risk of developing alloantibodies to vWF. 3258663 45 59 von Willebrand Modifier D014842 3258663 100 129 severe von Willebrand disease SpecificDisease D056729 3258663 131 160 Severe von Willebrand disease SpecificDisease D056729 3258663 217 231 von Willebrand Modifier D014842 3258663 1152 1171 genetic abnormality DiseaseClass D030342 3258663 1278 1299 genetic abnormalities DiseaseClass D030342 3258663 1519 1548 severe von Willebrand disease SpecificDisease D056729 7795591|t|Spectrum of germline mutations in the RB1 gene: a study of 232 patients with hereditary and non hereditary retinoblastoma. 7795591|a|Germline mutations in the RB1 gene confer hereditary predisposition to retinoblastoma. We have performed a mutation survey of the RB1 gene in 232 patients with hereditary or non hereditary retinoblastoma. We systematically explored all 27 exons and flanking sequences as well as the promotor. All types of point mutations are represented and are found unequally distributed along the RB1 gene sequence. In the population we studied, exons 3, 8, 18 and 19 are preferentially altered. The range of frequency of detection of germline mutations is about 20%, indicating that other mechanisms of inactivation of RB1 should be involved. The spectrum of mutations presented here should help to improve the clinical management of retinoblastoma and to understand the molecular mechanisms leading to tumorigenesis.. 7795591 77 121 hereditary and non hereditary retinoblastoma CompositeMention D012175 7795591 194 208 retinoblastoma SpecificDisease D012175 7795591 283 326 hereditary or non hereditary retinoblastoma CompositeMention D012175 7795591 845 859 retinoblastoma SpecificDisease D012175 2773936|t|Pelizaeus-Merzbacher disease: an X-linked neurologic disorder of myelin metabolism with a novel mutation in the gene encoding proteolipid protein. 2773936|a|The nosology of the inborn errors of myelin metabolism has been stymied by the lack of molecular genetic analysis. Historically, Pelizaeus-Merzbacher disease has encompassed a host of neurologic disorders that present with a deficit of myelin, the membrane elaborated by glial cells that encircles and successively enwraps axons. We describe here a Pelizaeus-Merzbacher pedigree of the classical type, with X-linked inheritance, a typical clinical progression, and a pathologic loss of myelinating cells and myelin in the central nervous system. To discriminate variants of Pelizaeus-Merzbacher disease, a set of oligonucleotide primers was constructed to polymerase-chain-reaction (PCR) amplify and sequence the gene encoding proteolipid protein (PLP), a structural protein that comprises half of the protein of the myelin sheath. The PLP gene in one of two affected males and the carrier mother of this family exhibited a single base difference in the more than 2 kb of the PLP gene sequenced, a C----T transition that would create a serine substitution for proline at the carboxy end of the protein. Our results delineate the clinical features of Pelizaeus-Merzbacher disease, define the possible molecular pathology of this dysmyelinating disorder, and address the molecular classification of inborn errors of myelin metabolism. Patients with the classical form (type I) and the more severely affected, connatal variant of Pelizaeus-Merzbacher disease (type II) would be predicted to display mutation at the PLP locus. The other variants (types III-VI), which have sometimes been categorized as Pelizaeus-Merzbacher disease, may represent mutations in genes encoding other structural myelin proteins or proteins critical to myelination.. 2773936 0 28 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 2773936 33 82 X-linked neurologic disorder of myelin metabolism DiseaseClass D020279 2773936 167 201 inborn errors of myelin metabolism DiseaseClass D020279 2773936 276 304 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 2773936 331 351 neurologic disorders DiseaseClass D009422 2773936 372 389 deficit of myelin DiseaseClass D003711 2773936 496 516 Pelizaeus-Merzbacher Modifier OMIM:312080 2773936 721 749 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 2773936 1297 1325 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 2773936 1375 1398 dysmyelinating disorder DiseaseClass D003711 2773936 1444 1478 inborn errors of myelin metabolism DiseaseClass D020279 2773936 1574 1612 Pelizaeus-Merzbacher disease (type II) SpecificDisease OMIM:312080 2773936 1746 1774 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 7874163|t|CAG expansions in a novel gene for Machado-Joseph disease at chromosome 14q32.1. 7874163|a|We have identified a novel gene containing CAG repeats and mapped it to chromosome 14q32. 1, the genetic locus for Machado-Joseph disease (MJD). In normal individuals the gene contains between 13 and 36 CAG repeats, whereas most of the clinically diagnosed patients and all of the affected members of a family with the clinical and pathological diagnosis of MJD show expansion of the repeat-number (from 68-79). Southern blot analyses and genomic cloning demonstrates the existence of related genes. These results raise the possibility that similar abnormalities in related genes may give rise to diseases similar to MJD. 7874163 35 57 Machado-Joseph disease SpecificDisease D017827 7874163 196 218 Machado-Joseph disease SpecificDisease D017827 7874163 220 223 MJD SpecificDisease D017827 7874163 439 442 MJD SpecificDisease D017827 7874163 698 701 MJD SpecificDisease D017827 3659917|t|Conservation of the Duchenne muscular dystrophy gene in mice and humans. 3659917|a|A portion of the Duchenne muscular dystrophy (DMD) gene transcript from human fetal skeletal muscle and mouse adult heart was sequenced, representing approximately 25 percent of the total, 14-kb DMD transcript. The nucleic acid and predicted amino acid sequences from the two species are nearly 90 percent homologous. The amino acid sequence that is predicted from this portion of the DMD gene indicates that the protein product might serve a structural role in muscle, but the abundance and tissue distribution of the messenger RNA suggests that the DMD protein is not nebulin.. 3659917 20 47 Duchenne muscular dystrophy Modifier D020388 3659917 90 117 Duchenne muscular dystrophy Modifier D020388 3659917 119 122 DMD Modifier D020388 3659917 268 271 DMD Modifier D020388 3659917 458 461 DMD Modifier D020388 3659917 624 627 DMD Modifier D020388 2303408|t|Deficiency of the murine fifth complement component (C5). A 2-base pair gene deletion in a 5'-exon. 2303408|a|To ascertain the molecular mechanism that causes murine C5 deficiency, genomic and cDNA libraries were constructed from mouse liver DNA and mRNA employing the congenic strains B10. D2/nSnJ and B10. D2/oSnJ that are sufficient and deficient for C5, respectively. Genomic fragments were isolated which correspond to PvuII and HindIII restriction fragment length polymorphisms associated with C5 deficiency. Sequence analyses demonstrated that each of these polymorphisms resulted from single base pair substitutions and that neither substitution would probably cause or contribute to the C5 deficiency. Sequence analyses of C5 sufficient and deficient cDNAs revealed a 2 base-pair deletion in the deficient cDNAs. The " TA " deletion was located near the 5 end of the cDNA. This deletion shifts the reading frame of the C5 mRNA so that the termination codon UGA is present 4 base pairs downstream from the deletion. Genomic DNA was amplified and sequenced corresponding to the area surrounding the 2-base pair deletion. Six C5-deficient strains, A/HeJ, AKR/J, DBA/2J, NZB/B1NJ, SWR/J, and B10. D2/oSnJ, and four C5-sufficient strains, Balb/CJ, C57Bl/6J, DBA/1J, and B10. D2/nSnJ, were analyzed. The sequencing data revealed that the 2 base pairs were deleted from the C5 gene of each deficient mouse tested but not from the C5 gene of any sufficient mouse. These data demonstrate that 1) there is an identical 2-base pair deletion in an exon of the C5 gene in several different C5-deficient mouse strains; 2) the mRNA transcribed from the C5D gene retains this deletion; and 3) this mutation should result in C5 protein deficiency. 2303408 0 56 Deficiency of the murine fifth complement component (C5) SpecificDisease OMIM:609536 2303408 156 169 C5 deficiency SpecificDisease OMIM:609536 2303408 330 346 deficient for C5 SpecificDisease OMIM:609536 2303408 490 503 C5 deficiency SpecificDisease OMIM:609536 2303408 686 699 C5 deficiency SpecificDisease OMIM:609536 2303408 1122 1134 C5-deficient Modifier OMIM:609536 2303408 1577 1589 C5-deficient Modifier OMIM:609536 2303408 1708 1729 C5 protein deficiency SpecificDisease OMIM:609536 2241452|t|Recurrent meningitis in a patient with congenital deficiency of the C9 component of complement. First case of C9 deficiency in Europe. 2241452|a|We describe the first cases, to our knowledge, of C9 deficiency in Europe that were detected in a Swiss family, of which two members--one with a complete deficiency and the other with approximately half-normal C9 levels--experienced bacterial meningitis. The index patient, a 56-year-old white man with a history of purulent meningitis at the age of 23 years, presented with an acute meningococcal meningitis. No impairment of cellular immunity or immunoglobulin deficiency could be found. Complement assays showed a complete deficiency of the C9 component, while the other individual component levels were normal and the hemolytic activity (measured using the CH50 assay) was only slightly reduced. A family study revealed complete C9 deficiency in the patients healthy brother and half-normal C9 concentrations in his sister, his son (who also had experienced an episode of bacterial meningitis), and his niece, consistent with an inherited C9 deficiency. This first case of recurrent meningitis in a white patient with complete C9 deficiency suggests that this complement defect may also be a risk factor for bacterial, especially neisserial, infections.. 2241452 10 20 meningitis SpecificDisease D008581 2241452 39 94 congenital deficiency of the C9 component of complement SpecificDisease OMIM:613825 2241452 110 123 C9 deficiency SpecificDisease OMIM:613825 2241452 185 198 C9 deficiency SpecificDisease OMIM:613825 2241452 368 388 bacterial meningitis SpecificDisease D016920 2241452 451 470 purulent meningitis SpecificDisease D008586 2241452 513 543 acute meningococcal meningitis SpecificDisease D008585 2241452 548 579 impairment of cellular immunity DiseaseClass D007153 2241452 583 608 immunoglobulin deficiency SpecificDisease D004406 2241452 652 691 complete deficiency of the C9 component SpecificDisease OMIM:613825 2241452 859 881 complete C9 deficiency SpecificDisease OMIM:613825 2241452 1011 1031 bacterial meningitis SpecificDisease D016920 2241452 1078 1091 C9 deficiency SpecificDisease OMIM:613825 2241452 1112 1132 recurrent meningitis SpecificDisease D008581+D012008 2241452 1157 1179 complete C9 deficiency SpecificDisease OMIM:613825 2241452 1199 1216 complement defect DiseaseClass D007153 2241452 1247 1291 bacterial, especially neisserial, infections DiseaseClass D016870 6387532|t|Bone marrow transplant in adrenoleukodystrophy. 6387532|a|An allogeneic bone marrow transplant (BMT) from a normal HLA identical sibling donor was performed in a 13-year-old boy with rapidly progressive adrenoleukodystrophy (ALD). Engraftment and complete hematologic recovery occurred within 4 weeks, but neurologic deterioration continued. The patient died of an adenovirus infection 141 days after BMT. ALD is characterized by abnormally high plasma levels of very long chain fatty acids (VLCFA) as a result of impaired capacity to degrade them. Ten days after BMT, the white blood cell VLCFA levels and enzyme activity became normal; after 3 months, there was progressive reduction of plasma VLCFA to levels only slightly above normal.. 6387532 26 46 adrenoleukodystrophy SpecificDisease D000326 6387532 193 213 adrenoleukodystrophy SpecificDisease D000326 6387532 215 218 ALD SpecificDisease D000326 6387532 296 320 neurologic deterioration DiseaseClass D019636 6387532 355 375 adenovirus infection SpecificDisease D000257 6387532 396 399 ALD SpecificDisease D000326 7458742|t|Increased incidence of cataracts in male subjects deficient in glucose-6-phosphate dehydrogenase. 7458742|a|Glucose-6-phosphate dehydrogenase (G6PD) deficiency in RBCs was found significantly more frequently in 210 male cataractous patients than in 672 control subjects of Sardinian origin. The frequency of the deficiency was increasingly higher in presenile cataracts. In the G6PD-deficient group, the incidence of cortical and total cataracts was also increased. It is suggested that decrease of the G6PD activity in the lens, which accompanies its deficiency in the erythrocyte, might play a role in the cataracto-genesis of these patients. Moreover, G6PD deficiency should be added to other conditions, such as the galactosemic states and riboflavin deficiency, where cataracts represent a sensitive indicator of metabolic abnormalities of the RBC.. 7458742 23 32 cataracts SpecificDisease D002386 7458742 50 96 deficient in glucose-6-phosphate dehydrogenase SpecificDisease D005955 7458742 98 149 Glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 7458742 210 221 cataractous Modifier D002386 7458742 350 359 cataracts SpecificDisease D002386 7458742 368 382 G6PD-deficient Modifier D005955 7458742 407 435 cortical and total cataracts CompositeMention D002386 7458742 645 660 G6PD deficiency SpecificDisease D005955 7458742 710 722 galactosemic Modifier OMIM:230400 7458742 734 755 riboflavin deficiency SpecificDisease D012257 7458742 763 772 cataracts SpecificDisease D002386 7458742 808 831 metabolic abnormalities DiseaseClass D008659 2894613|t|Von Hippel-Lindau disease maps to the region of chromosome 3 associated with renal cell carcinoma. 2894613|a|Von Hippel-Lindau disease (VHL) is an autosomal dominant disorder with inherited susceptibility to various forms of cancer, including hemangioblastomas of the central nervous system, phaeochromocytomas, pancreatic malignancies, and renal cell carcinomas. Renal cell carcinomas constitute a particularly frequent cause of death in this disorder, occurring as bilateral and multifocal tumours, and presenting at an earlier age than in sporadic, non-familial cases of this tumour type. We report here that the VHL gene is linked to the locus encoding the human homologoue of the RAF1 oncogene, which maps to chromosome 3p25 (ref. 4). Crossovers with the VHL locus suggest that the defect responsible for the VHL phenotype is not a mutation in the RAF1 gene itself. An alternative or prior event to oncogene activation in tumour formation may be the inactivation of a putative tumour suppressor which can be associated with both the inherited and sporadic forms of the cancer. Sporadic renal cell carcinomas have previously been associated with the loss of regions on chromosome 3p (refs 5, 6). Consequently, sporadic and VHL-associated forms of renal cell carcinoma might both result from alterations causing loss of function of the same tumour suppressor gene on this chromosome.. 2894613 0 25 Von Hippel-Lindau disease SpecificDisease D006623 2894613 77 97 renal cell carcinoma SpecificDisease D002292 2894613 99 124 Von Hippel-Lindau disease SpecificDisease D006623 2894613 126 129 VHL SpecificDisease D006623 2894613 137 164 autosomal dominant disorder DiseaseClass D030342 2894613 215 221 cancer DiseaseClass D009369 2894613 233 250 hemangioblastomas SpecificDisease D018325 2894613 282 300 phaeochromocytomas SpecificDisease D010673 2894613 302 325 pancreatic malignancies SpecificDisease OMIM:260350 2894613 331 352 renal cell carcinomas SpecificDisease D002292 2894613 354 375 Renal cell carcinomas SpecificDisease D002292 2894613 457 489 bilateral and multifocal tumours CompositeMention D009369 2894613 569 575 tumour Modifier D009369 2894613 606 609 VHL Modifier D006623 2894613 750 753 VHL Modifier D006623 2894613 804 807 VHL Modifier D006623 2894613 917 923 tumour Modifier D009369 2894613 972 978 tumour Modifier D009369 2894613 1028 1070 inherited and sporadic forms of the cancer CompositeMention D009369 2894613 1072 1102 Sporadic renal cell carcinomas SpecificDisease D002292 2894613 1241 1261 renal cell carcinoma SpecificDisease D002292 2894613 1334 1340 tumour Modifier D009369 2895982|t|Tightly linked flanking markers for the Lowe oculocerebrorenal syndrome, with application to carrier assessment. 2895982|a|The Lowe oculocerebrorenal syndrome (OCRL) is characterized by congenital cataract, mental retardation, and defective renal tubular function. A map assignment of OCRL to Xq24-q26 has been made previously by linkage analysis with DXS42 at Xq24-q26 (theta = 0, z = 5. 09) and with DXS10 at Xq26 (theta = 0, z = 6. 45). Two additional families were studied and three additional polymorphisms were identified at DXS42 by using a 35-kb sequence isolated with the probe detecting the original polymorphism at DXS42. With additional OCRL families made informative for DXS42, theta remained 0 with z = 6. 63; and for DXS10 theta = 0. 03 and z = 7. 07. Evidence for placing OCRL at Xq25 also comes from a female with Lowe syndrome and an X; 3 translocation. We have used the Xq25 breakpoint in this patient to determine the position of OCRL relative to the two linked markers. Each derivative chromosome was isolated away from its normal counterpart in somatic cell hybrids. DXS42 was mapped to the derivative chromosome X containing Xpterq25, and DXS10 was mapped to the derivative chromosome 3 containing Xq25-qter. The markers DXS10 and DXS42 therefore show tight linkage with OCRL in six families and flank the Xq25 breakpoint in a female patient with an X; 3 translocation. Linkage analysis with flanking markers was used to assess OCRL carrier status in women at risk. Results, when compared with carrier determination by ophthalmologic examination, indicated that the slit-lamp exam can be a sensitive and specific method of carrier determination in many cases 2895982 40 71 Lowe oculocerebrorenal syndrome SpecificDisease D009800 2895982 117 148 Lowe oculocerebrorenal syndrome SpecificDisease D009800 2895982 150 154 OCRL SpecificDisease D009800 2895982 187 195 cataract SpecificDisease D002386 2895982 197 215 mental retardation DiseaseClass D008607 2895982 221 253 defective renal tubular function DiseaseClass D015499 2895982 275 279 OCRL SpecificDisease D009800 2895982 639 643 OCRL Modifier D009800 2895982 821 834 Lowe syndrome SpecificDisease D009800 2895982 1284 1288 OCRL SpecificDisease D009800 2895982 1441 1445 OCRL Modifier D009800 10716718|t|Abnormal development of Purkinje cells and lymphocytes in Atm mutant mice. 10716718|a|Motor incoordination, immune deficiencies, and an increased risk of cancer are the characteristic features of the hereditary disease ataxia-telangiectasia (A-T), which is caused by mutations in the ATM gene. Through gene targeting, we have generated a line of Atm mutant mice, Atm (y/y) mice. In contrast to other Atm mutant mice, Atm (y/y) mice show a lower incidence of thymic lymphoma and survive beyond a few months of age. Atm (y/y) mice exhibit deficits in motor learning indicative of cerebellar dysfunction. Even though we found no gross cerebellar degeneration in older Atm (y/y) animals, ectopic and abnormally differentiated Purkinje cells were apparent in mutant mice of all ages. These findings establish that some neuropathological abnormalities seen in A-T patients also are present in Atm mutant mice. In addition, we report a previously unrecognized effect of Atm deficiency on development or maintenance of CD4 (+) 8 (+) thymocytes. We discuss these findings in the context of the hypothesis that abnormal development of Purkinje cells and lymphocytes contributes to the pathogenesis of A-T.. 10716718 75 95 Motor incoordination DiseaseClass D002524 10716718 97 116 immune deficiencies DiseaseClass D007154 10716718 143 149 cancer DiseaseClass D009369 10716718 189 207 hereditary disease DiseaseClass D030342 10716718 208 229 ataxia-telangiectasia SpecificDisease D001260 10716718 231 234 A-T SpecificDisease D001260 10716718 447 462 thymic lymphoma SpecificDisease D013953 10716718 567 589 cerebellar dysfunction DiseaseClass D002526 10716718 621 644 cerebellar degeneration DiseaseClass D013132 10716718 803 834 neuropathological abnormalities DiseaseClass D009422 10716718 843 846 A-T Modifier D001260 10716718 952 966 Atm deficiency SpecificDisease OMIM:208900 10716718 1180 1183 A-T SpecificDisease D001260 1303173|t|Both mutations in G6PD A- are necessary to produce the G6PD deficient phenotype. 1303173|a|The high prevalence of glucose 6-phosphate dehydrogenase (G6PD) deficiency in African populations is due almost entirely to the enzyme variant A-, which differs from the wild-type G6PD B by two amino acid replacements, 68 Val-- > Met and 126 Asn-- > Asp. The non-deficient polymorphic variant G6PD A contains only the mutation 126 Asn-- > Asp. The frequencies of the G6PD A and of the G6PD A- genes in parts of Africa are both about 0. 2. The 68 Val-- > Met mutation has not been found in a B background. This could be because the 68 Val-- > Met mutation happened to arise in an A gene in the first instance, or because the 68 Val-- > Met mutation alone is not sufficient to cause G6PD deficiency. We have approached this question by producing G6PD B, A, A-, and G6PD 68 Val-- > Met in a bacterial expression system and analysing their biochemical properties. With each single mutation we found a slight decrease in both the specific activity and the yield of enzyme when compared to G6PD B. When both mutations were introduced together, there was a roughly additive effect on specific activity, but a much more drastic effect on enzyme yield (4% of normal). This synergistic effect was also demonstrated on thermal stability, especially at low NADP concentrations. Comparable results were produced when the replacement 119 Gln-- > Glu was studied instead of 126 Asn-- > Asp. We infer that the coexistence of the two mutations is responsible for enzyme deficiency in G6PD A- because they act synergistically in causing instability of the enzyme. 1303173 55 69 G6PD deficient Modifier D005955 1303173 104 155 glucose 6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 1303173 762 777 G6PD deficiency SpecificDisease D005955 1303173 1527 1552 enzyme deficiency in G6PD SpecificDisease D005955 2253937|t|Detection of 98% of DMD/BMD gene deletions by polymerase chain reaction. 2253937|a|We describe oligonucleotide primer sequences that can be used to amplify eight exons plus the muscle promoter of the dystrophin gene in a single multiplex polymerase chain reaction (PCR). When used in conjunction with an existing primer set, these two multiplex reactions detect about 98% of deletions in patients with Duchenne or Becker muscular dystrophy (DMD, BMD). Furthermore, these primers amplify most of the exons in the deletion prone " hot spot " region around exons 44 to 53, allowing determination of deletion endpoints and prediction of mutational effects on the translational reading frame. Thus, use of these PCR-based assays will allow deletion detection and prenatal diagnosis for most DMD/BMD patients in a fraction of the time required for Southern blot analysis.. 2253937 20 23 DMD Modifier D020388 2253937 24 27 BMD Modifier C537666 2253937 392 429 Duchenne or Becker muscular dystrophy CompositeMention D020388|C537666 2253937 431 434 DMD SpecificDisease D020388 2253937 436 439 BMD SpecificDisease C537666 2253937 776 779 DMD Modifier D020388 2253937 780 783 BMD Modifier C537666 2591962|t|Recombination events that locate myotonic dystrophy distal to APOC2 on 19q. 2591962|a|We previously reported a recombination in an individual with myotonic dystrophy (DM) which placed the markers D19S19 and APOC2 on the same side of the DM locus. Haplotyping of this family with more recently characterized probes which are either tightly linked to DM or distal to the linkage group at q13. 2 shows that the DM locus is distal to APOC2. This is confirmed by other recombinants where DM segregates with distal probes. Additional marker to marker recombinations in unaffected individuals are reported and support the order and orientation of the DM linkage group as pter- (INSR, LDLR, S9) - (S19, BCL3, APOC2) - (CKMM, DM) - (S22, + + + PRKCG) -qter. The data presented here cannot determine whether DM is proximal or distal to CKMM. The consequences of this probe order for antenatal diagnosis and future research aiming to isolate the gene which is affected in DM are discussed. 2591962 33 51 myotonic dystrophy SpecificDisease D009223 2591962 137 155 myotonic dystrophy SpecificDisease D009223 2591962 157 159 DM SpecificDisease D009223 2591962 227 229 DM Modifier D009223 2591962 339 341 DM SpecificDisease D009223 2591962 398 400 DM Modifier D009223 2591962 473 475 DM SpecificDisease D009223 2591962 634 636 DM Modifier D009223 2591962 788 790 DM SpecificDisease D009223 2591962 951 953 DM SpecificDisease D009223 10425038|t|New mutations, polymorphisms, and rare variants in the ATM gene detected by a novel SSCP strategy. 10425038|a|The gene for ataxia-telangiectasia, ATM, spans about 150 kb of genomic DNA. ATM mutations are found along the entire gene, with no evidence of a mutational hot spot. Using DNA as the starting material, we screened the ATM gene in 92 A-T patients, using an optimized single-strand conformation polymorphism (SSCP) technique that detected all previously known mutations in the polymerase chain reaction (PCR) segments being analyzed. To expedite screening, we sequentially loaded the SSCP gels with three different sets of PCR products that were pretested to avoid overlapping patterns. Many of the DNA changes we detected were intragenic polymorphisms. Of an expected 177 unknown mutations, we detected approximately 70%, mostly protein truncating mutations (that would have been detectable by protein truncation testing if RNA starting material had been available). Mutations have now been defined for every exon of the ATM gene. Herein, we present 35 new mutations and 34 new intragenic polymorphisms or rare variants within the ATM gene. This is the most comprehensive compilation of ATM polymorphisms assembled to date. Defining polymorphic sites as well as mutations in the ATM gene will be of great importance in designing automated methods for detecting mutations.. 10425038 112 133 ataxia-telangiectasia SpecificDisease D001260 10425038 332 335 A-T Modifier D001260 10417280|t|Chromosome breakage in the Prader-Willi and Angelman syndromes involves recombination between large, transcribed repeats at proximal and distal breakpoints. 10417280|a|Prader-Willi syndrome (PWS) and Angelman syndrome (AS) are distinct neurobehavioral disorders that most often arise from a 4-Mb deletion of chromosome 15q11-q13 during paternal or maternal gametogenesis, respectively. At a de novo frequency of approximately. 67-1/10, 000 births, these deletions represent a common structural chromosome change in the human genome. To elucidate the mechanism underlying these events, we characterized the regions that contain two proximal breakpoint clusters and a distal cluster. Novel DNA sequences potentially associated with the breakpoints were positionally cloned from YACs within or near these regions. Analyses of rodent-human somatic-cell hybrids, YAC contigs, and FISH of normal or rearranged chromosomes 15 identified duplicated sequences (the END repeats) at or near the breakpoints. The END-repeat units are derived from large genomic duplications of a novel gene (HERC2), many copies of which are transcriptionally active in germline tissues. One of five PWS/AS patients analyzed to date has an identifiable, rearranged HERC2 transcript derived from the deletion event. We postulate that the END repeats flanking 15q11-q13 mediate homologous recombination resulting in deletion. Furthermore, we propose that active transcription of these repeats in male and female germ cells may facilitate the homologous recombination process. 10417280 27 62 Prader-Willi and Angelman syndromes CompositeMention D011218|D017204 10417280 157 178 Prader-Willi syndrome SpecificDisease D011218 10417280 180 183 PWS SpecificDisease D011218 10417280 189 206 Angelman syndrome SpecificDisease D017204 10417280 208 210 AS SpecificDisease D017204 10417280 225 250 neurobehavioral disorders DiseaseClass D019954 10417280 1159 1162 PWS Modifier D011218 10417280 1163 1165 AS Modifier D017204 8530105|t|A 4-megabase YAC contig that spans the Langer-Giedion syndrome region on human chromosome 8q24.1: use in refining the location of the trichorhinophalangeal syndrome and multiple exostoses genes (TRPS1 and EXT1). 8530105|a|We have constructed a physical map covering over 4 Mb of human chromosome 8q24. 1 and used this map to refine the locations of the genes responsible for Langer-Giedion syndrome. The map is composed of overlapping YAC clones that were identified and ordered in relation to sequence tagged sites mapped to the Langer-Giedion chromosomal region on somatic cell hybrids. The minimal region of overlap of Langer-Giedion syndrome deletions, previously identified by analysis of 15 patients, was placed on the map by analysis of 2 patients whose deletions define the endpoints. The chromosome 8 breakpoint of a balanced t (8; 9) (q24. 11; q33. 3) translocation from a patient with trichorhinophalangeal syndrome (TRPS I) was found to be located just within the proximal end of the minimal deletion region. A deletion of 8q24. 11-q24. 3 in a patient with multiple exostoses was found to overlap the distal end of the LGS deletion region, indicating that the EXT1 gene is distal to the TRPS1 gene and supporting the hypothesis that Langer-Giedion syndrome is due to loss of functional copies of both the TRPS1 and the EXT1 genes 8530105 39 62 Langer-Giedion syndrome Modifier D015826 8530105 134 164 trichorhinophalangeal syndrome SpecificDisease OMIM:190350 8530105 365 388 Langer-Giedion syndrome SpecificDisease D015826 8530105 612 635 Langer-Giedion syndrome Modifier D015826 8530105 886 916 trichorhinophalangeal syndrome SpecificDisease OMIM:190350 8530105 918 922 TRPS SpecificDisease OMIM:190350 8530105 1121 1124 LGS Modifier D015826 8530105 1235 1258 Langer-Giedion syndrome SpecificDisease D015826 8401501|t|Genetic mapping of the breast-ovarian cancer syndrome to a small interval on chromosome 17q12-21: exclusion of candidate genes EDH17B2 and RARA. 8401501|a|A susceptibility gene for hereditary breast-ovarian cancer, BRCA1, has been assigned by linkage analysis to chromosome 17q21. Candidate genes in this region include EDH17B2, which encodes estradiol 17 beta-hydroxysteroid dehydrogenase II (17 beta-HSD II), and RARA, the gene for retinoic acid receptor alpha. We have typed 22 breast and breast-ovarian cancer families with eight polymorphisms from the chromosome 17q12-21 region, including two in the EDH17B2 gene. Genetic recombination with the breast cancer trait excludes RARA from further consideration as a candidate gene for BRCA1. Both BRCA1 and EDH17B2 map to a 6 cM interval (between THRA1 and D17S579) and no recombination was observed between the two genes. However, direct sequencing of overlapping PCR products containing the entire EDH17B2 gene in four unrelated affected women did not uncover any sequence variation, other than previously described polymorphisms. Mutations in the EDH17B2 gene, therefore do not appear to be responsible for the hereditary breast-ovarian cancer syndrome. Single meiotic crossovers in affected women suggest that BRCA1 is flanked by the loci RARA and D17S78.. 8401501 23 53 breast-ovarian cancer syndrome DiseaseClass D061325 8401501 171 203 hereditary breast-ovarian cancer DiseaseClass D061325 8401501 471 503 breast and breast-ovarian cancer Modifier D001943|D061325 8401501 641 654 breast cancer Modifier D001943 8401501 1155 1196 hereditary breast-ovarian cancer syndrome DiseaseClass D061325 313733|t|Hereditary C2 deficiency associated with common variable immunodeficiency. 313733|a|Homozygous C2 deficiency in a 19-year-old boy was associated with variable immunodeficiency manifested by marked hypoimmunoglobulinemia and impaired antibody responses, normal circulating B lymphocytes, and subnormal T-cell functions. Neither antilymphocytic autoantibodies nor chromosomal abnormalities were found. Serum immunoglobulin levels were within normal limits in his parents and brother who were heterozygous for C2 deficiency. The patients lymphocytes were homozygous at the HLA-D locus but expressed an antigen different from DW2.. 313733 0 24 Hereditary C2 deficiency SpecificDisease OMIM:217000 313733 57 73 immunodeficiency DiseaseClass D007153 313733 86 99 C2 deficiency SpecificDisease OMIM:217000 313733 150 166 immunodeficiency DiseaseClass D007153 313733 188 210 hypoimmunoglobulinemia DiseaseClass D007153 313733 353 378 chromosomal abnormalities DiseaseClass D002869 313733 498 511 C2 deficiency SpecificDisease OMIM:217000 3012567|t|Isolation of molecular probes associated with the chromosome 15 instability in the Prader-Willi syndrome. 3012567|a|Flow cytometry and recombinant DNA techniques have been used to obtain reagents for a molecular analysis of the Prader-Willi syndrome (PWS). HindIII total-digest libraries were prepared in lambda phage Charon 21A from flow-sorted inverted duplicated no. 15 human chromosomes and propagated on recombination-proficient (LE392) and recBC-, sbcB- (DB1257) bacteria. Twelve distinct chromosome 15-specific probes have been isolated. Eight localized to the region 15q11----13. Four of these eight sublocalized to band 15q11. 2 and are shown to be deleted in DNA of one of two patients examined with the PWS. Heteroduplex analysis of two of these clones, which grew on DB1257 but not on LE392, revealed stem-loop structures in the inserts, indicative of inverted, repeated DNA elements. Such DNA repeats might account for some of the cloning instability of DNA segments from proximal 15q. Analysis of the genetic and physical instability associated with the repeated sequences we have isolated from band 15q11. 2 may elucidate the molecular basis for the instability of this chromosomal region in patients with the PWS or other diseases associated with chromosomal abnormalities in the proximal long arm of human chromosome 15 3012567 83 104 Prader-Willi syndrome SpecificDisease D011218 3012567 218 239 Prader-Willi syndrome SpecificDisease D011218 3012567 241 244 PWS SpecificDisease D011218 3012567 704 707 PWS SpecificDisease D011218 3012567 1215 1218 PWS SpecificDisease D011218 3012567 1253 1278 chromosomal abnormalities DiseaseClass D002869 7543316|t|Myotonic dystrophy: evidence for a possible dominant-negative RNA mutation. 7543316|a|The trinucleotide expansion mutation causing myotonic dystrophy is in the 3 untranslated region of a protein kinase gene. The molecular mechanisms by which the expanded repeat causes the clinically variable and multisystemic disease, myotonic dystrophy, are not understood. It has been particularly difficult to rationalize the dominant inheritance with the fact that the expansion mutation lies outside of the protein-encoding gene elements, and should not be translated into protein. Here we use muscle biopsies from classical adult-onset myotonic dystrophy patients to study the accumulation of transcripts from both the normal and expanded DM kinase genes in patient muscle, and compare the results to normal and myopathic controls. We found relatively small decreases of DM kinase RNA in the total RNA pool from muscle; however, these reductions were not disease specific. Analysis of poly (A) + RNA showed dramatic decreases of both the mutant and normal DM kinase RNAs, and these changes were disease-specific. Our findings are consistent with a novel molecular pathogenetic mechanism for myotonic dystrophy both the normal and expanded DM kinase genes are transcribed in patient muscle, but the abnormal expansion-containing RNA has a dominant effect on RNA metabolism by preventing the accumulation of poly (A) + RNA. The ability of the expansion mutation to alter accumulation of poly (A) + RNA in trans suggests that myotonic dystrophy may be the first example of a dominant-negative mutation manifested at the RNA level.. 7543316 0 18 Myotonic dystrophy SpecificDisease D009223 7543316 121 139 myotonic dystrophy SpecificDisease D009223 7543316 287 308 multisystemic disease DiseaseClass D004194 7543316 310 328 myotonic dystrophy SpecificDisease D009223 7543316 617 635 myotonic dystrophy Modifier D009223 7543316 720 722 DM Modifier D009223 7543316 793 802 myopathic Modifier D009135 7543316 852 854 DM Modifier D009223 7543316 1037 1039 DM Modifier D009223 7543316 1172 1190 myotonic dystrophy SpecificDisease D009223 7543316 1221 1223 DM Modifier D009223 7543316 1505 1523 myotonic dystrophy SpecificDisease D009223 8004674|t|Isolation of the gene for McLeod syndrome that encodes a novel membrane transport protein. 8004674|a|McLeod syndrome is an X-linked multisystem disorder characterized by abnormalities in the neuromuscular and hematopoietic systems. We have assembled a cosmid contig of 360 kb that encompasses the McLeod gene locus. A 50 kb deletion was detected by screening DNA from patients with radiolabeled whole cosmids, and two transcription units were identified within this deletion. The mRNA expression pattern of one of them, designated as XK, correlates closely to the McLeod phenotype. XK encodes a novel protein with structural characteristics of prokaryotic and eukaryotic membrane transport proteins. Nucleotide sequence analysis of XK from two unrelated McLeod patients has identified point mutations at conserved splice donor and acceptor sites. These findings provide direct evidence that XK is responsible for McLeod syndrome.. 8004674 26 41 McLeod syndrome SpecificDisease OMIM:300842 8004674 91 106 McLeod syndrome SpecificDisease OMIM:300842 8004674 113 142 X-linked multisystem disorder DiseaseClass D040181 8004674 287 293 McLeod Modifier OMIM:300842 8004674 554 560 McLeod Modifier OMIM:300842 8004674 744 750 McLeod Modifier OMIM:300842 8004674 903 918 McLeod syndrome SpecificDisease OMIM:300842 10790204|t|Molecular basis of very long chain acyl-CoA dehydrogenase deficiency in three Israeli patients: identification of a complex mutant allele with P65L and K247Q mutations, the former being an exonic mutation causing exon 3 skipping. 10790204|a|Very long chain acyl-CoA dehydrogenase (VLCAD) deficiency is a life-threatening disorder of mitochondrial fatty acid beta-oxidation. We identified four novel mutations in three unrelated patients. All patients had the severe childhood form of VLCAD deficiency with early onset and high mortality. Immunoblot analysis revealed that VLCAD protein was undetectable in patients 2 and 3, whereas normal-size VLCAD protein and an aberrant form of VLCAD (4kDa smaller) were detected in patient 1. As expected, null mutations were found in patients 2 and 3 patient 2 is homozygous for a frameshift mutation, del 4 bp at 798-801, and patient 3 is homozygous for a nonsense mutation 65C > A (S22X). Patient 1 was homozygous for a complex mutant allele containing two alterations, including a 194C > T transition (P65L) and 739A > C transversion (K247Q); in the case of P65L, the amino acid change does not reduce enzyme activity. However, the nucleotide change resulted in exon 3 skipping, whereas the latter K247Q mutation had a drastic effect on enzyme activity. We verified these events by in vivo splicing experiments and transient expression analysis of mutant cDNAs. The P65L mutation locates 11 bases upstream of a splice donor site of intron 3. This is an example of an exonic mutation which affects exon-splicing.. 10790204 19 68 very long chain acyl-CoA dehydrogenase deficiency SpecificDisease C536353 10790204 230 287 Very long chain acyl-CoA dehydrogenase (VLCAD) deficiency SpecificDisease C536353 10790204 473 489 VLCAD deficiency SpecificDisease C536353 10932179|t|Amino-terminal fragments of mutant huntingtin show selective accumulation in striatal neurons and synaptic toxicity. 10932179|a|Huntington disease (HD) is caused by expansion of a glutamine repeat in the amino-terminal region of huntingtin. Despite its widespread expression, mutant huntingtin induces selective neuronal loss in striatal neurons. Here we report that, in mutant mice expressing HD repeats, the production and aggregation of N-terminal huntingtin fragments preferentially occur in HD-affected neurons and their processes and axonal terminals. N-terminal fragments of mutant huntingtin form aggregates and induce neuritic degeneration in cultured striatal neurons. N-terminal mutant huntingtin also binds to synaptic vesicles and inhibits their glutamate uptake in vitro. The specific processing and accumulation of toxic fragments of N-terminal huntingtin in HD-affected striatal neurons, especially in their neuronal processes and axonal terminals, may contribute to the selective neuropathology of HD.. 10932179 117 135 Huntington disease SpecificDisease D006816 10932179 137 139 HD SpecificDisease D006816 10932179 383 385 HD Modifier D006816 10932179 485 487 HD Modifier D006816 10932179 616 637 neuritic degeneration DiseaseClass D009410 10932179 863 865 HD Modifier D006816 10932179 1004 1006 HD SpecificDisease D006816 8240110|t|Phenotypic variation including retinitis pigmentosa, pattern dystrophy, and fundus flavimaculatus in a single family with a deletion of codon 153 or 154 of the peripherin/RDS gene. 8240110|a|BACKGROUND AND OBJECTIVES Mutations of the peripherin/RDS gene have been reported in autosomal dominant retinitis pigmentosa, pattern macular dystrophy, and retinitis punctata albescens. We report herein the occurrence of three separate phenotypes within a single family with a novel 3-base pair deletion of codon 153 or 154 of the peripherin/RDS gene. DESIGN Case reports with clinical features, fluorescein angiography, kinetic perimetry, electrophysiological studies, and molecular genetics. SETTING University medical centers. PATIENTS A 75-year-old woman, her two daughters (aged 44 and 50 years), and her 49-year-old son were screened for peripherin/RDS mutations because of the presence of multiple phenotypes within the same family. RESULTS The mother presented at age 63 years with a profoundly abnormal electroretinogram (ERG) and adult-onset retinitis pigmentosa that progressed dramatically over 12 years, with marked loss of peripheral visual field. One daughter developed pattern macular dystrophy at age 31 years. At age 44 years, her ERG was moderately abnormal but her clinical disease was limited to the macula. Another daughter presented at age 42 years with macular degeneration and over 10 years developed the clinical picture of fundus flavimaculatus. Her peripheral visual field was preserved but her ERG was moderately abnormal. The son had onset of macular degeneration at age 44 years. Pericentral scotomas were present and the ERG was markedly abnormal. Fluorescein angiography revealed punctate pigment epithelial transmission defects. CONCLUSIONS A 3-base pair deletion of codon 153 or 154 of the peripherin/RDS gene can produce clinically disparate phenotypes even within the same family.. 8240110 31 51 retinitis pigmentosa SpecificDisease D012174 8240110 53 70 pattern dystrophy SpecificDisease D008268 8240110 76 97 fundus flavimaculatus SpecificDisease C535804 8240110 267 306 autosomal dominant retinitis pigmentosa SpecificDisease D012174 8240110 316 333 macular dystrophy SpecificDisease D008268 8240110 339 367 retinitis punctata albescens SpecificDisease OMIM:136880 8240110 1039 1059 retinitis pigmentosa SpecificDisease D012174 8240110 1172 1197 pattern macular dystrophy SpecificDisease D008268 8240110 1364 1384 macular degeneration SpecificDisease D008268 8240110 1437 1458 fundus flavimaculatus SpecificDisease C535804 8240110 1560 1580 macular degeneration SpecificDisease D008268 8240110 1598 1618 Pericentral scotomas SpecificDisease D012607 10441571|t|Missense mutation in the alternative splice region of the PAX6 gene in eye anomalies. 10441571|a|The PAX6 gene is involved in ocular morphogenesis, and PAX6 mutations have been detected in various types of ocular anomalies, including aniridia, Peters anomaly, corneal dystrophy, congenital cataract, and foveal hypoplasia. The gene encodes a transcriptional regulator that recognizes target genes through its paired-type DNA-binding domain. The paired domain is composed of two distinct DNA-binding subdomains, the N-terminal subdomain (NTS) and the C-terminal subdomain (CTS), which bind respective consensus DNA sequences. The human PAX6 gene produces two alternative splice isoforms that have the distinct structure of the paired domain. The insertion, into the NTS, of 14 additional amino acids encoded by exon 5a abolishes the DNA-binding activity of the NTS and unmasks the DNA-binding ability of the CTS. Thus, exon 5a appears to function as a molecular switch that specifies target genes. We ascertained a novel missense mutation in four pedigrees with Peters anomaly, congenital cataract, Axenfeldt anomaly, and/or foveal hypoplasia, which, to our knowledge, is the first mutation identified in the splice-variant region. A T-- > A transition at the 20th nucleotide position of exon 5a results in a Val-- > Asp (GTC-- > GAC) substitution at the 7th codon of the alternative splice region. Functional analyses demonstrated that the V54D mutation slightly increased NTS binding and decreased CTS transactivation activity to almost half.. 10441571 71 84 eye anomalies DiseaseClass D005124 10441571 195 211 ocular anomalies DiseaseClass D005124 10441571 223 231 aniridia SpecificDisease D015783 10441571 233 247 Peters anomaly SpecificDisease C537884 10441571 249 266 corneal dystrophy SpecificDisease D003317 10441571 268 287 congenital cataract SpecificDisease D002386 10441571 293 310 foveal hypoplasia SpecificDisease OMIM:136520 10441571 1050 1064 Peters anomaly SpecificDisease C537884 10441571 1066 1085 congenital cataract SpecificDisease D002386 10441571 1087 1104 Axenfeldt anomaly SpecificDisease C535679 10441571 1113 1130 foveal hypoplasia SpecificDisease OMIM:136520 1673289|t|A detailed multipoint map of human chromosome 4 provides evidence for linkage heterogeneity and position-specific recombination rates. 1673289|a|Utilizing the CEPH reference panel and genotypic data for 53 markers, we have constructed a 20-locus multipoint genetic map of human chromosome 4. New RFLPs are reported for four loci. The map integrates a high-resolution genetic map of 4p16 into a continuous map extending to 4q31 and an unlinked cluster of three loci at 4q35. The 20 linked markers form a continuous linkage group of 152 cM in males and 202 cM in females. Likely genetic locations are provided for 25 polymorphic anonymous sequences and 28 gene-specific RFLPs. The map was constructed employing the LINKAGE and CRIMAP computational methodologies to build the multipoint map via a stepwise algorithm. A detailed 10-point map of the 4p16 region constructed from the CEPH panel provides evidence for heterogeneity in the linkage maps constructed from families segregating for Huntington disease (HD). It additionally provides evidence for position-specific recombination frequencies in the telomeric region of 4p.. 1673289 977 995 Huntington disease SpecificDisease D006816 1673289 997 999 HD SpecificDisease D006816 8566965|t|The murine homolog of the human breast and ovarian cancer susceptibility gene Brca1 maps to mouse chromosome 11D. 8566965|a|The recently cloned human breast and ovarian cancer susceptibility gene, BRCA1, is located on human chromosome 17q21. We have isolated murine genomic clones containing Brca1 as a first step in generating a mouse model for the loss of BRCA1 function. A mouse genomic library was screened using probes corresponding to exon 11 of the human BRCA1 gene. Two overlapping mouse clones were identified that hybridized to human BRCA1 exons 9-12. Sequence analysis of 1. 4 kb of the region of these clones corresponding to part of human exon 11 revealed 72% nucleic acid identity but only 50% amino acid identity with the human gene. The longest of the mouse Brca1 genomic clones maps to chromosome 11D, as determined by two-color fluorescence in situ hybridization. The synteny to human chromosome 17 was confirmed by cohybridization with the mouse probe for the NF1-gene. This comparative study confirms that the relative location of the BRCA1 gene has been conserved between mice and humans. 8566965 32 57 breast and ovarian cancer Modifier D061325 8566965 140 165 breast and ovarian cancer Modifier D061325 8101038|t|High residual arylsulfatase A (ARSA) activity in a patient with late-infantile metachromatic leukodystrophy. 8101038|a|We identified a patient suffering from late-infantile metachromatic leukodystrophy (MLD) who has a residual arylsulfatase A (ARSA) activity of about 10%. Fibroblasts of the patient show significant sulfatide degradation activity exceeding that of adult MLD patients. Analysis of the ARSA gene in this patient revealed heterozygosity for two new mutant alleles in one allele, deletion of C 447 in exon 2 leads to a frameshift and to a premature stop codon at amino acid position 105; in the second allele, a G-- > A transition in exon 5 causes a Gly309-- > Ser substitution. Transient expression of the mutant Ser309-ARSA resulted in only 13% enzyme activity of that observed in cells expressing normal ARSA. The mutant ARSA is correctly targeted to the lysosomes but is unstable. These findings are in contrast to previous results showing that the late-infantile type of MLD is always associated with the complete absence of ARSA activity. The expression of the mutant ARSA protein may be influenced by particular features of oligodendrocytes, such that the level of mutant enzyme is lower in these cells than in others.. 8101038 64 107 late-infantile metachromatic leukodystrophy SpecificDisease D007966 8101038 148 191 late-infantile metachromatic leukodystrophy SpecificDisease D007966 8101038 193 196 MLD SpecificDisease D007966 8101038 356 365 adult MLD Modifier D007966 8101038 958 984 late-infantile type of MLD SpecificDisease D007966 10802660|t|De novo deletions of SNRPN exon 1 in early human and mouse embryos result in a paternal to maternal imprint switch. 10802660|a|Prader-Willi syndrome (PWS) is a neurogenetic disease characterized by infantile hypotonia, gonadal hypoplasia, obsessive behaviour and neonatal feeding difficulties followed by hyperphagia, leading to profound obesity. PWS is due to a lack of paternal genetic information at 15q11-q13 (ref. 2). Five imprinted, paternally expressed genes map to the PWS region, MKRN3 (ref. 3), NDN (ref. 4), NDNL1 (ref. 5), SNRPN (refs 6-8) and IPW (ref. 9), as well as two poorly characterized framents designated PAR-1 and PAR-5 (ref. 10). Imprinting of this region involves a bipartite imprinting centre (IC), which overlaps SNRPN (refs 10, 11). Deletion of the SNRPN promoter/exon 1 region (the PWS IC element) appears to impair the establishment of the paternal imprint in the male germ line and leads to PWS. Here we report a PWS family in which the father is mosaic for an IC deletion on his paternal chromosome. The deletion chromosome has acquired a maternal methylation imprint in his somatic cells. We have made identical findings in chimaeric mice generated from two independent embryonic stem (ES) cell lines harbouring a similar deletion. Our studies demonstrate that the PWS IC element is not only required for the establishment of the paternal imprint, but also for its postzygotic maintenance.. 10802660 116 137 Prader-Willi syndrome SpecificDisease D011218 10802660 139 142 PWS SpecificDisease D011218 10802660 149 169 neurogenetic disease DiseaseClass D020271 10802660 187 206 infantile hypotonia SpecificDisease D009123 10802660 208 226 gonadal hypoplasia SpecificDisease D006058 10802660 294 305 hyperphagia SpecificDisease D006963 10802660 327 334 obesity SpecificDisease D009765 10802660 336 339 PWS SpecificDisease D011218 10802660 466 469 PWS Modifier D011218 10802660 799 802 PWS Modifier D011218 10802660 910 913 PWS SpecificDisease D011218 10802660 932 935 PWS Modifier D011218 10802660 1286 1289 PWS Modifier D011218 1897530|t|Molecular characterization of two galactosemia mutations: correlation of mutations with highly conserved domains in galactose-1-phosphate uridyl transferase. 1897530|a|Galactosemia is an autosomal recessive disorder of human galactose metabolism caused by deficiency of the enzyme galactose-1-phosphate uridyl transferase (GALT). The molecular basis of this disorder is at present not well understood. We report here two missense mutations which result in low or undetectable enzymatic activity. First, we identified at nucleotide 591 a transition which substitutes glutamine 188 by arginine. The mutated glutamine is not only highly conserved in evolution (conserved also in Escherichia coli and Saccharomyces cerevisiae), but is also two amino acid residues downstream from the active site histidine-proline-histidine triad and results in about 10% of normal enzymatic activity. The arginine 188 mutation is the most common galactosemia mutation characterized to date. It accounts for one-fourth of the galactosemia alleles studied. Second, we report the substitution of arginine 333 by tryptophan, caused by a transition at nucleotide 1025. The area surrounding this missense mutation is the most highly conserved domain in the homologous enzymes from E. coli, yeast, and humans, and this mutation results in undetectable enzymatic activity, suggesting that this is a severe mutation. This second mutation appears to be rare, since it was found only in the patient we sequenced. Our data provide further evidence for the heterogeneity of galactosemia at the molecular level, heterogeneity which might be related to the variable clinical outcome observed in this disorder.. 1897530 34 46 galactosemia Modifier D005693 1897530 158 170 Galactosemia SpecificDisease D005693 1897530 177 235 autosomal recessive disorder of human galactose metabolism DiseaseClass D005693 1897530 246 311 deficiency of the enzyme galactose-1-phosphate uridyl transferase SpecificDisease D005693 1897530 916 928 galactosemia Modifier D005693 1897530 995 1007 galactosemia Modifier D005693 1897530 1531 1543 galactosemia SpecificDisease D005693 10078732|t|Neurophysiologic follow-up of long-term dietary treatment in adult-onset adrenoleukodystrophy. 10078732|a|OBJECTIVE To monitor the effects of dietary treatment in adult-onset adrenoleukodystrophy (ALD) by means of somatosensory evoked potentials (SEPs) and motor evoked potentials (MEPs). BACKGROUND SEPs and MEPs have proved useful in revealing signs of progressively severe, central dying-back axonopathy in early stages of adult-onset ALD. METHODS Eight patients with adult-onset ALD underwent clinical examination, brain and spine MRI, and SEP and MEP studies before and after 3 years of Lorenzos oil dietary therapy. RESULTS Before treatment, brain MRI was normal in five patients. Three of these patients had pure spinal SEP abnormalities and in the remaining two patients SEPs showed signs of involvement of both the spinal and cerebral somatosensory tracts. After treatment, the three patients with pure spinal abnormalities showed clinical and neurophysiologic worsening, whereas the two patients with a more advanced stage of disease (exhibited by SEPs) showed substantially unchanged clinical and neurophysiologic features. The patients with abnormal brain MRI at the onset of treatment showed clinical and neurophysiologic worsening. CONCLUSIONS Lorenzos oil therapy had no effect on patients with evidence of inflammatory brain lesions. Moreover, in patients without clear signs of inflammatory damage, this treatment does not modify significantly the natural course of the disease. However, because effective treatments should begin before the onset of severe neurologic symptoms, SEPs and MEPs should be considered to evaluate the effectiveness of other experimental treatments in the patient with a negative brain MRI. 10078732 73 93 adrenoleukodystrophy SpecificDisease D000326 10078732 165 185 adrenoleukodystrophy SpecificDisease D000326 10078732 187 190 ALD SpecificDisease D000326 10078732 376 397 dying-back axonopathy SpecificDisease D016472 10078732 429 432 ALD SpecificDisease D000326 10078732 475 478 ALD SpecificDisease D000326 10078732 713 737 spinal SEP abnormalities DiseaseClass D016472 10078732 905 925 spinal abnormalities DiseaseClass D016472 10078732 1316 1342 inflammatory brain lesions DiseaseClass D004660 10577908|t|The molecular basis of Sjogren-Larsson syndrome: mutation analysis of the fatty aldehyde dehydrogenase gene. 10577908|a|Sjogren-Larsson syndrome (SLS) is an autosomal recessive disorder characterized by ichthyosis, mental retardation, spasticity, and deficient activity of fatty aldehyde dehydrogenase (FALDH). To define the molecular defects causing SLS, we performed mutation analysis of the FALDH gene in probands from 63 kindreds with SLS. Among these patients, 49 different mutations-including 10 deletions, 2 insertions, 22 amino acid substitutions, 3 nonsense mutations, 9 splice-site defects, and 3 complex mutations-were found. All of the patients with SLS were found to carry mutations. Nineteen of the missense mutations resulted in a severe reduction of FALDH enzyme catalytic activity when expressed in mammalian cells, but one mutation (798G-- > C [K266N]) seemed to have a greater effect on mRNA stability. The splice-site mutations led to exon skipping or utilization of cryptic acceptor-splice sites. Thirty-seven mutations were private, and 12 mutations were seen in two or more probands of European or Middle Eastern descent. Four single-nucleotide polymorphisms (SNPs) were found in the FALDH gene. At least four of the common mutations (551C-- > T, 682C-- > T, 733G-- > A, and 798 + 1delG) were associated with multiple SNP haplotypes, suggesting that these mutations originated independently on more than one occasion or were ancient SLS genes that had undergone intragenic recombination. Our results demonstrate that SLS is caused by a strikingly heterogeneous group of mutations in the FALDH gene and provide a framework for understanding the genetic basis of SLS and the development of DNA-based diagnostic tests.. 10577908 23 47 Sjogren-Larsson syndrome SpecificDisease D016111 10577908 109 133 Sjogren-Larsson syndrome SpecificDisease D016111 10577908 135 138 SLS SpecificDisease D016111 10577908 146 174 autosomal recessive disorder DiseaseClass D030342 10577908 192 202 ichthyosis DiseaseClass D007057 10577908 204 222 mental retardation DiseaseClass D008607 10577908 224 234 spasticity DiseaseClass D009128 10577908 240 290 deficient activity of fatty aldehyde dehydrogenase SpecificDisease D016111 10577908 340 343 SLS SpecificDisease D016111 10577908 428 431 SLS SpecificDisease D016111 10577908 651 654 SLS SpecificDisease D016111 10577908 1445 1448 SLS Modifier D016111 10577908 1529 1532 SLS SpecificDisease D016111 10577908 1673 1676 SLS SpecificDisease D016111 7055648|t|Severe-glucose-6-phosphate dehydrogenase (G6PD) deficiency associated with chronic hemolytic anemia, granulocyte dysfunction, and increased susceptibility to infections: description of a new molecular variant (G6PD Barcelona). 7055648|a|Molecular, kinetic, and functional studies were carried out on erythrocytes and leukocytes in a Spanish male with G6PD deficiency, congenital nonspherocytic hemolytic anemia (CNSHA), and increased susceptibility to infections. G6PD activity was absent in patients red cells and was about 2% of normal in leukocytes. Molecular studies using standard methods (WHO, 1967) showed G6PD in the patient to have a slightly fast electrophoretic mobility at pH 8. 0 with otherwise normal properties (heat stability at 46 degrees C, apparent affinity for substrates, optimum pH, and utilization of substrate analogues). Other tests showed the patients granulocytes to engulf latex particles normally, but to have impaired reduction of nitroblue tetrazolium and ferricytochrome-c as well as reduced iodination. Chemotaxis and random migration of the patients granulocytes were normal as were myeloperoxidase, leukocyte alkaline phosphatase (LAP), and ultrastructural features. The molecular characteristics of G6PD in the patient differed from those of all previously reported variants associated with CNSHA, so the present variant was provisionally called G6PD Barcelona to distinguish it from other G6PD variants previously described. Possible mechanisms for the severe deficiency of G6PD in erythrocytes and granulocytes was investigated by studies on the immunologic specific activity of the mutant enzyme. 7055648 0 58 Severe-glucose-6-phosphate dehydrogenase (G6PD) deficiency SpecificDisease D005955 7055648 83 99 hemolytic anemia SpecificDisease D000743 7055648 101 124 granulocyte dysfunction DiseaseClass D007960 7055648 341 356 G6PD deficiency SpecificDisease D005955 7055648 358 400 congenital nonspherocytic hemolytic anemia SpecificDisease D000746 7055648 402 407 CNSHA SpecificDisease D000746 7055648 1317 1322 CNSHA SpecificDisease D000746 7055648 1487 1505 deficiency of G6PD SpecificDisease D005955 8162071|t|Mutations at the PAX6 locus are found in heterogeneous anterior segment malformations including Peters' anomaly. 8162071|a|Mutation or deletion of the PAX6 gene underlies many cases of aniridia. Three lines of evidence now converge to implicate PAX6 more widely in anterior segment malformations including Peters anomaly. First, a child with Peters anomaly is deleted for one copy of PAX6. Second, affected members of a family with dominantly inherited anterior segment malformations, including Peters anomaly are heterozygous for an R26G mutation in the PAX6 paired box. Third, a proportion of Sey/+ Smalleye mice, heterozygous for a nonsense mutation in murine Pax-6, have an ocular phenotype resembling Peters anomaly. We therefore propose that a variety of anterior segment anomalies may be associated with PAX6 mutations.. 8162071 55 85 anterior segment malformations DiseaseClass C537775 8162071 96 111 Peters' anomaly SpecificDisease C537884 8162071 175 183 aniridia SpecificDisease D015783 8162071 255 285 anterior segment malformations DiseaseClass C537775 8162071 296 310 Peters anomaly SpecificDisease C537884 8162071 332 346 Peters anomaly SpecificDisease C537884 8162071 443 473 anterior segment malformations DiseaseClass C537775 8162071 485 499 Peters anomaly SpecificDisease C537884 8162071 696 710 Peters anomaly SpecificDisease C537884 8162071 751 777 anterior segment anomalies DiseaseClass C537775 10364525|t|In Swedish families with hereditary prostate cancer, linkage to the HPC1 locus on chromosome 1q24-25 is restricted to families with early-onset prostate cancer. 10364525|a|Prostate cancer clusters in some families, and an estimated 5% -10% of all cases are estimated to result from inheritance of prostate cancer-susceptibility genes. We previously reported evidence of linkage to the 1q24-25 region (HPC1) in 91 North American and Swedish families each with multiple cases of prostate cancer (Smith et al. 1996). In the present report we analyze 40 (12 original and 28 newly identified) Swedish families with hereditary prostate cancer (HPC) that, on the basis of 40 markers spanning a 25-cM interval within 1q24-25, have evidence of linkage. In the complete set of families, a maximum two-point LOD score of 1. 10 was observed at D1S413 (at a recombination fraction [theta] of. 1), with a maximum NPL (nonparametric linkage) Z score of 1. 64 at D1S202 (P =. 05). The evidence of linkage to this region originated almost exclusively from the subset of 12 early-onset (age < 65 years) families, which yielded a maximum LOD score of 2. 38 at D1S413 (straight theta = 0) and an NPL Z score of 1. 95 at D1S422 (P =. 03). Estimates from heterogeneity tests suggest that, within Sweden, as many as 50% of early-onset families had evidence of linkage to the HPC1 region. These results are consistent with the hypothesis of linkage to HPC1 in a subset of families with prostate cancer, particularly those with an early age at diagnosis. 10364525 25 51 hereditary prostate cancer SpecificDisease C537243 10364525 144 159 prostate cancer SpecificDisease D011471 10364525 161 176 Prostate cancer SpecificDisease D011471 10364525 466 481 prostate cancer SpecificDisease D011471 10364525 599 625 hereditary prostate cancer SpecificDisease C537243 10364525 627 630 HPC SpecificDisease C537243 10364525 1451 1466 prostate cancer SpecificDisease D011471 2037285|t|Localisation of the myotonic dystrophy locus to 19q13.2-19q13.3 and its relationship to twelve polymorphic loci on 19q. 2037285|a|The order of fourteen polymorphic markers localised to the long arm of human chromosome 19 has been established by multipoint mapping in a set of 40 CEPH (Centre dEtude de Polymorphisme Humain, Paris) reference families. We report here the linkage relationship of the myotonic dystrophy (DM) locus to twelve of these markers as studied in 45 families with DM. The resulting genetic map is supported by the localisation of the DNA markers in a panel of somatic cell hybrids. Ten of the twelve markers have been shown to be proximal to the DM gene and two, PRKCG and D19S22, distal but at distances of approximately 25 cM and 15 cM, respectively. The closest proximal markers are APOC2 (apolipoprotein C-II) and CKM (creatine kinase, muscle) approximately 3 cM and 2 cM from the DM gene respectively, in the order APOC2-CKM-DM. The distance between APOC2, CKM and DM (of the order of 2 million base pairs) and their known orientation should permit directional chromosome walking and jumping. The data presented here should enable us to determine whether or not new markers are distal to APOC2/CKM and thus potentially flank the DM gene.. 2037285 20 38 myotonic dystrophy Modifier D009223 2037285 388 406 myotonic dystrophy Modifier D009223 2037285 408 410 DM Modifier D009223 2037285 476 478 DM SpecificDisease D009223 2037285 658 660 DM Modifier D009223 2037285 897 899 DM Modifier D009223 2037285 982 984 DM SpecificDisease D009223 2037285 1246 1248 DM Modifier D009223 10712225|t|A recurrent expansion of a maternal allele with 36 CAG repeats causes Huntington disease in two sisters. 10712225|a|Large intergenerational repeat expansions of the CAG trinucleotide repeat in the HD gene have been well documented for the male germline. We describe a recurrent large expansion of a maternal allele with 36 CAG repeats (to 66 and 57 repeats, respectively, in two daughters) associated with onset of Huntington disease (HD) in the second and third decade in a family without history of HD. Our findings give evidence of a gonadal mosaicism in the unaffected mother. We hypothesize that large expansions also occur in the female germline and that a negative selection of oocytes with long repeats might explain the different instability behavior of the male and the female germlines.. 10712225 70 88 Huntington disease SpecificDisease D006816 10712225 186 188 HD Modifier D006816 10712225 404 422 Huntington disease SpecificDisease D006816 10712225 424 426 HD SpecificDisease D006816 10712225 490 492 HD SpecificDisease D006816 10072428|t|Germline E-cadherin gene (CDH1) mutations predispose to familial gastric cancer and colorectal cancer. 10072428|a|Inherited mutations in the E-cadherin gene (CDH1) were described recently in three Maori kindreds with familial gastric cancer. Familial gastric cancer is genetically heterogeneous and it is not clear what proportion of gastric cancer susceptibility in non-Maori populations is due to germline CDH1 mutations. Therefore, we screened eight familial gastric cancer kindreds of British and Irish origin for germline CDH1 mutations, by SSCP analysis of all 16 exons and flanking sequences. Each family contained (i) two cases of gastric cancer in first degree relatives with one affected before age 50 years; or (ii) three or more cases of gastric cancer. Novel germline CDH1 mutations (a nonsense and a splice site) were detected in two families (25%). Both mutations were predicted to truncate the E-cadherin protein in the signal peptide domain. In one family there was evidence of non-penetrance and susceptibility to both gastric and colorectal cancer; thus, in addition to six cases of gastric cancer, a CDH1 mutation carrier developed colorectal cancer at age 30 years. We have confirmed that germline mutations in the CDH1 gene cause familial gastric cancer in non-Maori populations. However, only a minority of familial gastric cancers can be accounted for by CDH1 mutations. Loss of E-cadherin function has been implicated in the pathogenesis of sporadic colorectal and other cancers, and our findings provide evidence that germline CDH1 mutations predispose to early onset colorectal cancer. Thus, CDH1 should be investigated as a cause of inherited susceptibility to both gastric and colorectal cancers. 10072428 56 79 familial gastric cancer SpecificDisease D013274 10072428 84 101 colorectal cancer SpecificDisease D015179 10072428 206 229 familial gastric cancer SpecificDisease D013274 10072428 231 254 Familial gastric cancer SpecificDisease D013274 10072428 323 337 gastric cancer Modifier D013274 10072428 442 465 familial gastric cancer Modifier D013274 10072428 629 643 gastric cancer SpecificDisease D013274 10072428 740 754 gastric cancer SpecificDisease D013274 10072428 1027 1056 gastric and colorectal cancer CompositeMention D015179|D013274 10072428 1092 1106 gastric cancer SpecificDisease D013274 10072428 1142 1159 colorectal cancer SpecificDisease D015179 10072428 1242 1265 familial gastric cancer SpecificDisease D013274 10072428 1329 1344 gastric cancers SpecificDisease D013274 10072428 1465 1493 colorectal and other cancers DiseaseClass D015179|D009369 10072428 1584 1601 colorectal cancer SpecificDisease D015179 10072428 1684 1714 gastric and colorectal cancers CompositeMention D015179|D013274 3346017|t|Germinal mosaicism in Duchenne muscular dystrophy. 3346017|a|We have identified a Duchenne muscular dystrophy (DMD) pedigree where the disease is associated with a molecular deletion within the DMD locus. We have examined the meiotic segregation products of the common female ancestor using marker restriction fragment length polymorphisms (RFLPs) detected by probes that lie within this deletion. These studies show that this female has transmitted three distinct types of X chromosome to her offspring. This observation may be explained by postulating that the mutation arose as a postzygotic deletion within this common ancestor, who was consequently germinally mosaic.. 3346017 22 49 Duchenne muscular dystrophy SpecificDisease D020388 3346017 72 99 Duchenne muscular dystrophy Modifier D020388 3346017 101 104 DMD Modifier D020388 3346017 184 187 DMD Modifier D020388 3347839|t|Expression of the murine Duchenne muscular dystrophy gene in muscle and brain. 3347839|a|Complementary DNA clones were isolated that represent the 5 terminal 2. 5 kilobases of the murine Duchenne muscular dystrophy (Dmd) messenger RNA (mRNA). Mouse Dmd mRNA was detectable in skeletal and cardiac muscle and at a level approximately 90 percent lower in brain. Dmd mRNA is also present, but at much lower than normal levels, in both the muscle and brain of three different strains of dystrophic mdx mice. The identification of Dmd mRNA in brain raises the possibility of a relation between human Duchenne muscular dystrophy (DMD) gene expression and the mental retardation found in some DMD males. These results also provide evidence that the mdx mutations are allelic variants of mouse Dmd gene mutations. 3347839 25 52 Duchenne muscular dystrophy Modifier D020388 3347839 177 204 Duchenne muscular dystrophy Modifier D020388 3347839 473 483 dystrophic Modifier D020388 3347839 585 612 Duchenne muscular dystrophy SpecificDisease D020388 3347839 614 617 DMD SpecificDisease D020388 3347839 643 661 mental retardation DiseaseClass D008607 3347839 676 679 DMD Modifier D020388 7790377|t|Human peroxisomal targeting signal-1 receptor restores peroxisomal protein import in cells from patients with fatal peroxisomal disorders. 7790377|a|Two peroxisomal targeting signals, PTS1 and PTS2, are involved in the import of proteins into the peroxisome matrix. Human patients with fatal generalized peroxisomal deficiency disorders fall into at least nine genetic complementation groups. Cells from many of these patients are deficient in the import of PTS1-containing proteins, but the causes of the protein-import defect in these patients are unknown. We have cloned and sequenced the human cDNA homologue (PTS1R) of the Pichia pastoris PAS8 gene, the PTS1 receptor (McCollum, D., E. Monosov, and S. Subramani. 1993. J. Cell Biol. 121 761-774). The PTS1R mRNA is expressed in all human tissues examined. Antibodies to the human PTS1R recognize this protein in human, monkey, rat, and hamster cells. The protein is localized mainly in the cytosol but is also found to be associated with peroxisomes. Part of the peroxisomal PTS1R protein is tightly bound to the peroxisomal membrane. Antibodies to PTS1R inhibit peroxisomal protein-import of PTS1-containing proteins in a permeabilized CHO cell system. In vitro-translated PTS1R protein specifically binds a serine-lysine-leucine-peptide. A PAS8-PTS1R fusion protein complements the P. pastoris pas8 mutant. The PTS1R cDNA also complements the PTS1 protein-import defect in skin fibroblasts from patients--belonging to complementation group two--diagnosed as having neonatal adrenoleukodystrophy or Zellweger syndrome. The PTS1R gene has been localized to a chromosomal location where no other peroxisomal disorder genes are known to map. Our findings represent the only case in which the molecular basis of the protein-import deficiency in human peroxisomal disorders is understood. 7790377 116 137 peroxisomal disorders DiseaseClass D018901 7790377 294 326 peroxisomal deficiency disorders DiseaseClass D018901 7790377 1391 1417 PTS1 protein-import defect SpecificDisease OMIM:202370|OMIM:214100 7790377 1513 1542 neonatal adrenoleukodystrophy SpecificDisease OMIM:202370 7790377 1546 1564 Zellweger syndrome SpecificDisease D015211 7790377 1641 1661 peroxisomal disorder Modifier D018901 7790377 1759 1784 protein-import deficiency DiseaseClass D008661 7790377 1794 1815 peroxisomal disorders DiseaseClass D018901 8012387|t|X-linked spastic paraplegia and Pelizaeus-Merzbacher disease are allelic disorders at the proteolipid protein locus. 8012387|a|Three forms of X-linked spastic paraplegia (SPG) have been defined. One locus (SPG 1) maps to Xq28 while two clinically distinct forms map to Xq22 (SPG2). A rare X-linked dysmyelinating disorder of the central nervous system, Pelizaeus-Merzbacher disease (PMD), has also been mapped to Xq21-q22, and is caused by mutations in the proteolipid protein gene (PLP) which encodes two myelin proteins, PLP and DM20. While narrowing the genetic interval containing SPG2 in a large pedigree, we found that PLP was the closest marker to the disease locus, implicating PLP as a possible candidate gene. We have found that a point mutation (His139Tyr) in exon 3B of an affected male produces a mutant PLP but a normal DM20, and segregates with the disease (Zmax = 6. 63, theta = 0. 00). It appears, therefore, that SPG2 and PMD are allelic disorders 8012387 0 27 X-linked spastic paraplegia SpecificDisease OMIM:312920 8012387 32 60 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 8012387 65 82 allelic disorders DiseaseClass D030342 8012387 132 159 X-linked spastic paraplegia SpecificDisease OMIM:312920 8012387 161 164 SPG SpecificDisease OMIM:312920 8012387 279 311 X-linked dysmyelinating disorder DiseaseClass D020279 8012387 343 371 Pelizaeus-Merzbacher disease SpecificDisease OMIM:312080 8012387 373 376 PMD SpecificDisease OMIM:312080 8012387 930 933 PMD SpecificDisease OMIM:312080 8012387 938 955 allelic disorders DiseaseClass D030342