pmid
stringlengths
5
8
title
stringlengths
17
342
abstract
stringlengths
1
4.53k
all_entity_list
listlengths
2
73
label
sequencelengths
1
80
intra-sentence
sequencelengths
1
80
head_gene_entity
listlengths
1
80
tail_diease_entity
listlengths
1
80
head_name
sequencelengths
1
80
tail_name
sequencelengths
1
80
9394104
Association analysis of a regulatory variation of the serotonin transporter gene with severe alcohol dependence.
The present study tested the hypothesis that the short, low activity variant of a biallelic polymorphism in the 5' regulatory region of the human serotonin transporter (5-HTT) gene confers susceptibility to severe alcohol dependence marked by severe withdrawal symptoms. Applying a phenotype-genotype strategy, our population-based association analysis included 216 German controls and an extreme sample of 103 severely affected alcoholics who were selected from 315 German alcohol-dependent subjects by a history of alcohol withdrawal seizure or delirium. The frequency of the short allele (S) was significantly increased in the severely affected alcoholics, compared with that in the controls (X2 = 3.87, df = 1, nominal p = 0.049). The post-hoc exploration indicated that this allelic association resulted exclusively from a significant excess of the S/S genotype in the severely affected alcoholics (p = 0.035), suggesting a recessively acting effect. Consistently, we found a weak but significant correlation (p = 0.013) between the frequency of the S/S genotype and severity of withdrawal symptoms (WDS): no WDS [18.3%, odds ratio (OR) = 1.16], vegetative WDS only (21.8%, OR = 1.44), and severe WDS with either withdrawal seizure only or delirium only (25.0%, OR = 1.69), and both withdrawal seizure and delirium (30.8%, OR = 2.30). Further studies are required to test whether the tentative genotype-phenotype relationship occurred by chance or reflects a real genotypic association between a recessively modifying effect of the short variant of the functional 5-HTT promoter polymorphism and alcohol withdrawal vulnerability.
[ { "begin_idx": "93", "end_idx": "111", "entity_id": "D000437", "entity_type": "Disease", "text_name": "alcohol dependence" }, { "begin_idx": "327", "end_idx": "345", "entity_id": "D000437", "entity_type": "Disease", "text_name": "alcohol dependence" }, { "begin_idx": "660", "end_idx": "668", "entity_id": "D003693", "entity_type": "Disease", "text_name": "delirium" }, { "begin_idx": "1358", "end_idx": "1366", "entity_id": "D003693", "entity_type": "Disease", "text_name": "delirium" }, { "begin_idx": "1424", "end_idx": "1432", "entity_id": "D003693", "entity_type": "Disease", "text_name": "delirium" }, { "begin_idx": "649", "end_idx": "656", "entity_id": "D012640", "entity_type": "Disease", "text_name": "seizure" }, { "begin_idx": "1342", "end_idx": "1349", "entity_id": "D012640", "entity_type": "Disease", "text_name": "seizure" }, { "begin_idx": "1412", "end_idx": "1419", "entity_id": "D012640", "entity_type": "Disease", "text_name": "seizure" }, { "begin_idx": "363", "end_idx": "382", "entity_id": "D013375", "entity_type": "Disease", "text_name": "withdrawal symptoms" }, { "begin_idx": "1197", "end_idx": "1216", "entity_id": "D013375", "entity_type": "Disease", "text_name": "withdrawal symptoms" }, { "begin_idx": "1218", "end_idx": "1221", "entity_id": "D013375", "entity_type": "Disease", "text_name": "WDS" }, { "begin_idx": "1227", "end_idx": "1230", "entity_id": "D013375", "entity_type": "Disease", "text_name": "WDS" }, { "begin_idx": "1275", "end_idx": "1278", "entity_id": "D013375", "entity_type": "Disease", "text_name": "WDS" }, { "begin_idx": "1315", "end_idx": "1318", "entity_id": "D013375", "entity_type": "Disease", "text_name": "WDS" }, { "begin_idx": "54", "end_idx": "75", "entity_id": "6532", "entity_type": "Gene", "text_name": "serotonin transporter" }, { "begin_idx": "259", "end_idx": "280", "entity_id": "6532", "entity_type": "Gene", "text_name": "serotonin transporter" }, { "begin_idx": "282", "end_idx": "287", "entity_id": "6532", "entity_type": "Gene", "text_name": "5-HTT" }, { "begin_idx": "1682", "end_idx": "1687", "entity_id": "6532", "entity_type": "Gene", "text_name": "5-HTT" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "54", "end_idx": "75", "entity_id": "6532", "entity_type": "Gene", "text_name": "serotonin transporter" }, { "begin_idx": "282", "end_idx": "287", "entity_id": "6532", "entity_type": "Gene", "text_name": "5-HTT" } ]
[ { "begin_idx": "93", "end_idx": "111", "entity_id": "D000437", "entity_type": "Disease", "text_name": "alcohol dependence" }, { "begin_idx": "1275", "end_idx": "1278", "entity_id": "D013375", "entity_type": "Disease", "text_name": "WDS" } ]
[ "serotonin transporter", "5-HTT" ]
[ "alcohol dependence", "WDS" ]
9398842
Pendred syndrome is caused by mutations in a putative sulphate transporter gene (PDS).
Pendred syndrome is a recessively inherited disorder with the hallmark features of congenital deafness and thyroid goitre. By some estimates, the disorder may account for upwards of 10% of hereditary deafness. Previous genetic linkage studies localized the gene to a broad interval on human chromosome 7q22-31.1. Using a positional cloning strategy, we have identified the gene (PDS) mutated in Pendred syndrome and found three apparently deleterious mutations, each segregating with the disease in the respective families in which they occur. PDS produces a transcript of approximately 5 kb that was found to be expressed at significant levels only in the thyroid. The predicted protein, pendrin, is closely related to a number of known sulphate transporters. These studies provide compelling evidence that defects in pendrin cause Pendred syndrome thereby launching a new area of investigation into thyroid physiology, the pathogenesis of congenital deafness and the role of altered sulphate transport in human disease.
[ { "begin_idx": "0", "end_idx": "16", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred syndrome" }, { "begin_idx": "87", "end_idx": "103", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred syndrome" }, { "begin_idx": "482", "end_idx": "498", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred syndrome" }, { "begin_idx": "920", "end_idx": "936", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred syndrome" }, { "begin_idx": "631", "end_idx": "634", "entity_id": "C563184", "entity_type": "Disease", "text_name": "PDS" }, { "begin_idx": "170", "end_idx": "189", "entity_id": "D003638", "entity_type": "Disease", "text_name": "congenital deafness" }, { "begin_idx": "1028", "end_idx": "1047", "entity_id": "D003638", "entity_type": "Disease", "text_name": "congenital deafness" }, { "begin_idx": "194", "end_idx": "208", "entity_id": "D013959", "entity_type": "Disease", "text_name": "thyroid goitre" }, { "begin_idx": "109", "end_idx": "139", "entity_id": "D030342", "entity_type": "Disease", "text_name": "recessively inherited disorder" }, { "begin_idx": "276", "end_idx": "295", "entity_id": "D030342", "entity_type": "Disease", "text_name": "hereditary deafness" }, { "begin_idx": "81", "end_idx": "85", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS)" }, { "begin_idx": "466", "end_idx": "470", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS)" }, { "begin_idx": "631", "end_idx": "634", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "81", "end_idx": "85", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS)" }, { "begin_idx": "631", "end_idx": "634", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" } ]
[ { "begin_idx": "0", "end_idx": "16", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred syndrome" }, { "begin_idx": "170", "end_idx": "189", "entity_id": "D003638", "entity_type": "Disease", "text_name": "congenital deafness" } ]
[ "PDS)", "PDS" ]
[ "Pendred syndrome", "congenital deafness" ]
9407957
An association between the allele coding for a low activity variant of catechol-O-methyltransferase and the risk for breast cancer.
Mounting evidence suggests that catechol metabolites of estradiol may contribute to the development of estrogen-induced cancers. O-Methylation, catalyzed by catechol-O-methyltransferase (COMT), inactivates catechol estrogens. COMT is polymorphic in the human population, with 25% of Caucasians being homozygous for a low activity allele of the enzyme (COMT(LL)). We hypothesized that low activity COMT may be a risk factor for human breast cancer and designed a PCR-based RFLP assay to determine COMT genotype in a cohort of 112 matched, nested case-control samples. In the total study population, the odds ratios for the association of breast cancer risk with COMT(HL) and COMT(LL) genotypes were 1.30 [confidence interval (CI), 0.66-2.58] and 1.45 (CI, 0.69-3.07), respectively. Postmenopausal COMT(LL) women had a greater than 2-fold increased risk of developing breast cancer [odds ratio (OR), 2.18; CI, 0.93-5.11]. The association of COMT(LL) with the development of postmenopausal breast cancer was stronger and statistically significant in those women with a body mass index >24.47 kg/m2 (OR, 3.58; CI, 1.07-11.98). When COMT(LL) was combined with either glutathione S-transferase (GST) M1 null or with GSTP1 Ile-105-Val/Val-105-Val (intermediate/low activity, respectively) genotypes, the risk for developing postmenopausal breast cancer was also significantly increased. Our findings suggest that the allele encoding low activity COMT may be an important contributor to the postmenopausal development of breast cancer in certain women.
[ { "begin_idx": "798", "end_idx": "800", "entity_id": "C538324", "entity_type": "Disease", "text_name": "HL" }, { "begin_idx": "117", "end_idx": "130", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "565", "end_idx": "578", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "769", "end_idx": "782", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "998", "end_idx": "1011", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "1104", "end_idx": "1132", "entity_id": "D001943", "entity_type": "Disease", "text_name": "postmenopausal breast cancer" }, { "begin_idx": "1449", "end_idx": "1477", "entity_id": "D001943", "entity_type": "Disease", "text_name": "postmenopausal breast cancer" }, { "begin_idx": "1645", "end_idx": "1658", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "252", "end_idx": "259", "entity_id": "D009369", "entity_type": "Disease", "text_name": "cancers" }, { "begin_idx": "71", "end_idx": "99", "entity_id": "1312", "entity_type": "Gene", "text_name": "catechol-O-methyltransferase" }, { "begin_idx": "289", "end_idx": "317", "entity_id": "1312", "entity_type": "Gene", "text_name": "catechol-O-methyltransferase" }, { "begin_idx": "319", "end_idx": "323", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "358", "end_idx": "362", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "484", "end_idx": "488", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "529", "end_idx": "533", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "628", "end_idx": "632", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "793", "end_idx": "797", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "806", "end_idx": "810", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "928", "end_idx": "932", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "1071", "end_idx": "1075", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "1260", "end_idx": "1264", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "1571", "end_idx": "1575", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "1342", "end_idx": "1347", "entity_id": "2950", "entity_type": "Gene", "text_name": "GSTP1" }, { "begin_idx": "1294", "end_idx": "1319", "entity_id": "373156", "entity_type": "Gene", "text_name": "glutathione S-transferase" }, { "begin_idx": "1321", "end_idx": "1324", "entity_id": "373156", "entity_type": "Gene", "text_name": "GST" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "71", "end_idx": "99", "entity_id": "1312", "entity_type": "Gene", "text_name": "catechol-O-methyltransferase" }, { "begin_idx": "1321", "end_idx": "1324", "entity_id": "373156", "entity_type": "Gene", "text_name": "GST" } ]
[ { "begin_idx": "1104", "end_idx": "1132", "entity_id": "D001943", "entity_type": "Disease", "text_name": "postmenopausal breast cancer" }, { "begin_idx": "117", "end_idx": "130", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" } ]
[ "catechol-O-methyltransferase", "GST" ]
[ "postmenopausal breast cancer", "breast cancer" ]
9409270
The apolipoprotein E and beta-fibrinogen G/A-455 gene polymorphisms are associated with ischemic stroke involving large-vessel disease.
The relationship between the apolipoprotein E (apoE) and beta-fibrinogen G/A-455 polymorphisms and cerebrovascular disease (CVD) was examined in the present study. We compared 227 patients with the subtypes of CVD (large-vessel disease, lacunar stroke, cardiac embolism, or undetermined pathomechanisms) with 225 control subjects. The occurrence of apoE isoforms (E2, E3, and E4) and the beta-fibrinogen G/A-455 genotype was determined in these individuals. No differences in apoE polymorphisms or allele frequencies between the CVD patients and control subjects were found. However, analysis of apoE genotypes as a function of stroke subtype revealed that the apoE4 allele was significantly more common in those patients with macroangiopathy-associated CVD. The only CVD risk factor that distinguished patients with the E4 allele from those with other apoE genotypes was elevated cholesterol. No association between the beta-fibrinogen G/A-455 polymorphism and CVD was found. However, homozygosity for the A allele was more common in patients with CVD resulting from large-vessel disease. These data demonstrate that the apoE4 allele and the AA genotype of the beta-fibrinogen G/A-455 polymorphism occur significantly more frequently in patients with CVD resulting from stenosis of large, brain-supplying vessels. Such genetic analyses may further our understanding of the etiology of cerebrovascular disease.
[ { "begin_idx": "88", "end_idx": "103", "entity_id": "D002544", "entity_type": "Disease", "text_name": "ischemic stroke" }, { "begin_idx": "114", "end_idx": "134", "entity_id": "D004194", "entity_type": "Disease", "text_name": "large-vessel disease" }, { "begin_idx": "235", "end_idx": "258", "entity_id": "D004194", "entity_type": "Disease", "text_name": "cerebrovascular disease" }, { "begin_idx": "260", "end_idx": "263", "entity_id": "D004194", "entity_type": "Disease", "text_name": "CVD" }, { "begin_idx": "346", "end_idx": "349", "entity_id": "D004194", "entity_type": "Disease", "text_name": "CVD" }, { "begin_idx": "351", "end_idx": "371", "entity_id": "D004194", "entity_type": "Disease", "text_name": "large-vessel disease" }, { "begin_idx": "665", "end_idx": "668", "entity_id": "D004194", "entity_type": "Disease", "text_name": "CVD" }, { "begin_idx": "890", "end_idx": "893", "entity_id": "D004194", "entity_type": "Disease", "text_name": "CVD" }, { "begin_idx": "904", "end_idx": "907", "entity_id": "D004194", "entity_type": "Disease", "text_name": "CVD" }, { "begin_idx": "1098", "end_idx": "1101", "entity_id": "D004194", "entity_type": "Disease", "text_name": "CVD" }, { "begin_idx": "1185", "end_idx": "1188", "entity_id": "D004194", "entity_type": "Disease", "text_name": "CVD" }, { "begin_idx": "1204", "end_idx": "1224", "entity_id": "D004194", "entity_type": "Disease", "text_name": "large-vessel disease" }, { "begin_idx": "1388", "end_idx": "1391", "entity_id": "D004194", "entity_type": "Disease", "text_name": "CVD" }, { "begin_idx": "1522", "end_idx": "1545", "entity_id": "D004194", "entity_type": "Disease", "text_name": "cerebrovascular disease" }, { "begin_idx": "389", "end_idx": "405", "entity_id": "D006331", "entity_type": "Disease", "text_name": "cardiac embolism" }, { "begin_idx": "373", "end_idx": "387", "entity_id": "D020521", "entity_type": "Disease", "text_name": "lacunar stroke" }, { "begin_idx": "764", "end_idx": "770", "entity_id": "D020521", "entity_type": "Disease", "text_name": "stroke" }, { "begin_idx": "4", "end_idx": "20", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "165", "end_idx": "181", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "183", "end_idx": "187", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE" }, { "begin_idx": "485", "end_idx": "489", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE" }, { "begin_idx": "612", "end_idx": "616", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE" }, { "begin_idx": "732", "end_idx": "736", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE" }, { "begin_idx": "797", "end_idx": "802", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE4" }, { "begin_idx": "989", "end_idx": "993", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE" }, { "begin_idx": "1258", "end_idx": "1263", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE4" }, { "begin_idx": "499", "end_idx": "514", "entity_id": "6043", "entity_type": "Gene", "text_name": "(E2, E3, and E4" }, { "begin_idx": "499", "end_idx": "514", "entity_id": "6044", "entity_type": "Gene", "text_name": "(E2, E3, and E4" }, { "begin_idx": "499", "end_idx": "514", "entity_id": "9354", "entity_type": "Gene", "text_name": "(E2, E3, and E4" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "4", "end_idx": "20", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "499", "end_idx": "514", "entity_id": "9354", "entity_type": "Gene", "text_name": "(E2, E3, and E4" } ]
[ { "begin_idx": "373", "end_idx": "387", "entity_id": "D020521", "entity_type": "Disease", "text_name": "lacunar stroke" }, { "begin_idx": "351", "end_idx": "371", "entity_id": "D004194", "entity_type": "Disease", "text_name": "large-vessel disease" } ]
[ "apolipoprotein E", "(E2, E3, and E4" ]
[ "lacunar stroke", "large-vessel disease" ]
9416838
TP53 mutational pattern in Spanish and Polish non-small cell lung cancer patients: null mutations are associated with poor prognosis.
Inactivation of TP53 tumor suppressor gene is the most frequent molecular alteration in NSCLC, involving up to 60% of cases. Furthermore, TP53 mutational spectrum is related to the type of mutagen exposure, as well as racial and/or diet differences. Nearly 95% of TP53 perturbations affect codons included within exons 5-8 which encode for almost the entire DNA-binding domain. In this study we addressed the possible prognostic value of the molecular alterations identified in exons 5-8 of the TP53 gene in DNAs from 151 paraffin-embedded NSCLC sections corresponding to 59 Spanish and 92 Polish stage I-IIIA resected patients. PCR/single-strand conformation polymorphism (SSCP) analysis revealed that the occurrence of TP53 exon 5-8 mutations was 17/59 (29%) in the Spanish cohort and 17/92 (18%) in the Polish group. However, when DNA sequencing analysis was performed, these frequencies were reduced because of the presence of SSCP-false positive, intronic and silent mutations and polymorphisms. Fifteen of the 59 Spanish NSCLC tumors (25%) harbored TP53 mutations affecting exons 5-8 coding sequences, whereas only 12 of 92 Polish neoplasms (13%) contained alterations in the central hydrophobic region of p53. Our results indicate that the occurrence of TP53 mutations affecting exon 5-8 coding sequences in some European NSCLC populations may be lower than previously reported, and that the TP53 mutational patterns of these cohorts differ somewhat. The Spanish NSCLC patients contained missense mutations (9/59, 15%) and a relatively high percentage of null mutations (5/59, 8%) while the Polish patients mostly harbored missense mutations (9/92, 10%) and only one tumor contained a null type (1/92, 1%). Moreover, most TP53 missense mutations in the Spanish group were located outside the conserved regions, whereas the same mutations in the Polish group affected conserved amino acids. Furthermore, the Polish patients harbored a high percentage of G-->A transitions (most of them at non-CpG sites), while G-->T transversions were predominant in the Spanish group. Our findings suggest that there may be different racial or exogenous factors in these two populations which may help to explain both the distinct TP53 mutational pattern and the lower frequency obtained in the Polish group. The presence of missense mutations did not confer a worse clinical outcome in these subsets of NSCLC patients. However, patients whose tumors contained null TP53 gene mutations had a 5 month median disease-free survival time in contrast with 42 months in those patients without mutations (P=0.008). These findings suggest that loss of p53 function may enhance tumor progression in NSCLC patients independently of whether dominant negative TP53 missense mutations are present.
[ { "begin_idx": "46", "end_idx": "72", "entity_id": "D002289", "entity_type": "Disease", "text_name": "non-small cell lung cancer" }, { "begin_idx": "155", "end_idx": "160", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" }, { "begin_idx": "1161", "end_idx": "1173", "entity_id": "D009369", "entity_type": "Disease", "text_name": "NSCLC tumors" }, { "begin_idx": "1271", "end_idx": "1280", "entity_id": "D009369", "entity_type": "Disease", "text_name": "neoplasms" }, { "begin_idx": "1808", "end_idx": "1813", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" }, { "begin_idx": "2569", "end_idx": "2575", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumors" }, { "begin_idx": "2794", "end_idx": "2799", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" }, { "begin_idx": "0", "end_idx": "4", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "150", "end_idx": "154", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "272", "end_idx": "276", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "398", "end_idx": "402", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "629", "end_idx": "633", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "855", "end_idx": "859", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "1189", "end_idx": "1193", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "1346", "end_idx": "1349", "entity_id": "7157", "entity_type": "Gene", "text_name": "p53" }, { "begin_idx": "1395", "end_idx": "1399", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "1533", "end_idx": "1537", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "1863", "end_idx": "1867", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "2356", "end_idx": "2360", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "2591", "end_idx": "2595", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "2769", "end_idx": "2772", "entity_id": "7157", "entity_type": "Gene", "text_name": "p53" }, { "begin_idx": "2873", "end_idx": "2877", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "0", "end_idx": "4", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" }, { "begin_idx": "1533", "end_idx": "1537", "entity_id": "7157", "entity_type": "Gene", "text_name": "TP53" } ]
[ { "begin_idx": "46", "end_idx": "72", "entity_id": "D002289", "entity_type": "Disease", "text_name": "non-small cell lung cancer" }, { "begin_idx": "2794", "end_idx": "2799", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" } ]
[ "TP53", "TP53" ]
[ "non-small cell lung cancer", "tumor" ]
9425904
A polymorphism in the regulatory region of APOE associated with risk for Alzheimer's dementia.
The epsilon4 allele of the apolipoprotein E gene (APOE) has been associated with an increased risk of developing Alzheimer's disease (AD; refs 1,2). However, it is apparent that the APOEepsilon4 allele alone is neither necessary nor sufficient to cause the disease. We have recently found three new polymorphisms within the APOE transcriptional regulatory region (M.J.A. et al., manuscript submitted) and now establish an association between one of these polymorphisms (-491A/T) and dementia as observed in Alzheimer's disease, in two independent clinical populations. The results suggest that homozygosity of a common variant (-491A) is associated with increased risk for AD, and that this association is independent of APOEepsilon4 status. In vitro studies suggest that the -491A/T polymorphism may increase risk for AD by altering the level of ApoE protein expression.
[ { "begin_idx": "73", "end_idx": "93", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's dementia" }, { "begin_idx": "208", "end_idx": "227", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "229", "end_idx": "231", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "602", "end_idx": "621", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "768", "end_idx": "770", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "914", "end_idx": "916", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "578", "end_idx": "586", "entity_id": "D003704", "entity_type": "Disease", "text_name": "dementia" }, { "begin_idx": "43", "end_idx": "47", "entity_id": "348", "entity_type": "Gene", "text_name": "APOE" }, { "begin_idx": "122", "end_idx": "138", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "145", "end_idx": "149", "entity_id": "348", "entity_type": "Gene", "text_name": "APOE" }, { "begin_idx": "419", "end_idx": "423", "entity_id": "348", "entity_type": "Gene", "text_name": "APOE" }, { "begin_idx": "942", "end_idx": "946", "entity_id": "348", "entity_type": "Gene", "text_name": "ApoE" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "122", "end_idx": "138", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "419", "end_idx": "423", "entity_id": "348", "entity_type": "Gene", "text_name": "APOE" } ]
[ { "begin_idx": "73", "end_idx": "93", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's dementia" }, { "begin_idx": "578", "end_idx": "586", "entity_id": "D003704", "entity_type": "Disease", "text_name": "dementia" } ]
[ "apolipoprotein E", "APOE" ]
[ "Alzheimer's dementia", "dementia" ]
9428520
Molecular basis of sulfite oxidase deficiency from the structure of sulfite oxidase.
The molybdenum-containing enzyme sulfite oxidase catalyzes the conversion of sulfite to sulfate, the terminal step in the oxidative degradation of cysteine and methionine. Deficiency of this enzyme in humans usually leads to major neurological abnormalities and early death. The crystal structure of chicken liver sulfite oxidase at 1.9 A resolution reveals that each monomer of the dimeric enzyme consists of three domains. At the active site, the Mo is penta-coordinated by three sulfur ligands, one oxo group, and one water/hydroxo. A sulfate molecule adjacent to the Mo identifies the substrate binding pocket. Four variants associated with sulfite oxidase deficiency have been identified: two mutations are near the sulfate binding site, while the other mutations occur within the domain mediating dimerization.
[ { "begin_idx": "19", "end_idx": "45", "entity_id": "C538141", "entity_type": "Disease", "text_name": "sulfite oxidase deficiency" }, { "begin_idx": "730", "end_idx": "756", "entity_id": "C538141", "entity_type": "Disease", "text_name": "sulfite oxidase deficiency" }, { "begin_idx": "347", "end_idx": "358", "entity_id": "D003643", "entity_type": "Disease", "text_name": "early death" }, { "begin_idx": "316", "end_idx": "342", "entity_id": "D009422", "entity_type": "Disease", "text_name": "neurological abnormalities" }, { "begin_idx": "68", "end_idx": "83", "entity_id": "6821", "entity_type": "Gene", "text_name": "sulfite oxidase" }, { "begin_idx": "118", "end_idx": "133", "entity_id": "6821", "entity_type": "Gene", "text_name": "sulfite oxidase" }, { "begin_idx": "399", "end_idx": "414", "entity_id": "6821", "entity_type": "Gene", "text_name": "sulfite oxidase" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "68", "end_idx": "83", "entity_id": "6821", "entity_type": "Gene", "text_name": "sulfite oxidase" }, { "begin_idx": "68", "end_idx": "83", "entity_id": "6821", "entity_type": "Gene", "text_name": "sulfite oxidase" } ]
[ { "begin_idx": "19", "end_idx": "45", "entity_id": "C538141", "entity_type": "Disease", "text_name": "sulfite oxidase deficiency" }, { "begin_idx": "316", "end_idx": "342", "entity_id": "D009422", "entity_type": "Disease", "text_name": "neurological abnormalities" } ]
[ "sulfite oxidase", "sulfite oxidase" ]
[ "sulfite oxidase deficiency", "neurological abnormalities" ]
9438854
Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors.
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors in the human digestive tract, but their molecular etiology and cellular origin are unknown. Sequencing of c-kit complementary DNA, which encodes a proto-oncogenic receptor tyrosine kinase (KIT), from five GISTs revealed mutations in the region between the transmembrane and tyrosine kinase domains. All of the corresponding mutant KIT proteins were constitutively activated without the KIT ligand, stem cell factor (SCF). Stable transfection of the mutant c-kit complementary DNAs induced malignant transformation of Ba/F3 murine lymphoid cells, suggesting that the mutations contribute to tumor development. GISTs may originate from the interstitial cells of Cajal (ICCs) because the development of ICCs is dependent on the SCF-KIT interaction and because, like GISTs, these cells express both KIT and CD34.
[ { "begin_idx": "138", "end_idx": "156", "entity_id": "C535700", "entity_type": "Disease", "text_name": "mesenchymal tumors" }, { "begin_idx": "555", "end_idx": "571", "entity_id": "D002292", "entity_type": "Disease", "text_name": "stem cell factor" }, { "begin_idx": "573", "end_idx": "576", "entity_id": "D002292", "entity_type": "Disease", "text_name": "SCF" }, { "begin_idx": "882", "end_idx": "885", "entity_id": "D002292", "entity_type": "Disease", "text_name": "SCF" }, { "begin_idx": "747", "end_idx": "752", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" }, { "begin_idx": "45", "end_idx": "76", "entity_id": "D046152", "entity_type": "Disease", "text_name": "gastrointestinal stromal tumors" }, { "begin_idx": "78", "end_idx": "109", "entity_id": "D046152", "entity_type": "Disease", "text_name": "Gastrointestinal stromal tumors" }, { "begin_idx": "111", "end_idx": "116", "entity_id": "D046152", "entity_type": "Disease", "text_name": "GISTs" }, { "begin_idx": "362", "end_idx": "367", "entity_id": "D046152", "entity_type": "Disease", "text_name": "GISTs" }, { "begin_idx": "766", "end_idx": "771", "entity_id": "D046152", "entity_type": "Disease", "text_name": "GISTs" }, { "begin_idx": "920", "end_idx": "925", "entity_id": "D046152", "entity_type": "Disease", "text_name": "GISTs" }, { "begin_idx": "30", "end_idx": "35", "entity_id": "3815", "entity_type": "Gene", "text_name": "c-kit" }, { "begin_idx": "263", "end_idx": "268", "entity_id": "3815", "entity_type": "Gene", "text_name": "c-kit" }, { "begin_idx": "346", "end_idx": "349", "entity_id": "3815", "entity_type": "Gene", "text_name": "KIT" }, { "begin_idx": "488", "end_idx": "491", "entity_id": "3815", "entity_type": "Gene", "text_name": "KIT" }, { "begin_idx": "613", "end_idx": "618", "entity_id": "3815", "entity_type": "Gene", "text_name": "c-kit" }, { "begin_idx": "886", "end_idx": "889", "entity_id": "3815", "entity_type": "Gene", "text_name": "KIT" }, { "begin_idx": "952", "end_idx": "955", "entity_id": "3815", "entity_type": "Gene", "text_name": "KIT" }, { "begin_idx": "543", "end_idx": "553", "entity_id": "4254", "entity_type": "Gene", "text_name": "KIT ligand" }, { "begin_idx": "555", "end_idx": "571", "entity_id": "4254", "entity_type": "Gene", "text_name": "stem cell factor" }, { "begin_idx": "573", "end_idx": "576", "entity_id": "4254", "entity_type": "Gene", "text_name": "SCF" }, { "begin_idx": "882", "end_idx": "885", "entity_id": "4254", "entity_type": "Gene", "text_name": "SCF" }, { "begin_idx": "960", "end_idx": "964", "entity_id": "947", "entity_type": "Gene", "text_name": "CD34" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "30", "end_idx": "35", "entity_id": "3815", "entity_type": "Gene", "text_name": "c-kit" }, { "begin_idx": "886", "end_idx": "889", "entity_id": "3815", "entity_type": "Gene", "text_name": "KIT" } ]
[ { "begin_idx": "45", "end_idx": "76", "entity_id": "D046152", "entity_type": "Disease", "text_name": "gastrointestinal stromal tumors" }, { "begin_idx": "882", "end_idx": "885", "entity_id": "D002292", "entity_type": "Disease", "text_name": "SCF" } ]
[ "c-kit", "KIT" ]
[ "gastrointestinal stromal tumors", "SCF" ]
9443878
NAGLU mutations underlying Sanfilippo syndrome type B.
Sanfilippo syndrome type B (mucopolysaccharidosis III B) is a rare autosomal recessive disease caused by deficiency of alpha-N-acetylglucosaminidase, one of the enzymes required for the lysosomal degradation of heparan sulfate. The gene for this enzyme, NAGLU, recently was isolated, and several mutations were characterized. We have identified, in amplified exons from nine fibroblast cell lines derived from Sanfilippo syndrome type B patients, 10 additional mutations: Y92H, P115S, Y140C, E153K, R203X, 650insC, 901delAA, P358L, A664V, and L682R. Four of these mutations were found in homozygosity, and only two were seen in more than one cell line. Thus, Sanfilippo syndrome type B shows extensive molecular heterogeneity. Stable transfection of Chinese hamster ovary cells, by cDNA mutagenized to correspond to the NAGLU missense mutations, did not yield active enzyme, demonstrating the deleterious nature of the mutations. Nine of the 10 amino acid substitutions identified to date are clustered near the amino or the carboxyl end of alpha-N-acetylglucosaminidase, suggesting a role for these regions in the transport or function of the enzyme.
[ { "begin_idx": "27", "end_idx": "53", "entity_id": "D009084", "entity_type": "Disease", "text_name": "Sanfilippo syndrome type B" }, { "begin_idx": "55", "end_idx": "81", "entity_id": "D009084", "entity_type": "Disease", "text_name": "Sanfilippo syndrome type B" }, { "begin_idx": "83", "end_idx": "110", "entity_id": "D009084", "entity_type": "Disease", "text_name": "mucopolysaccharidosis III B" }, { "begin_idx": "465", "end_idx": "491", "entity_id": "D009084", "entity_type": "Disease", "text_name": "Sanfilippo syndrome type B" }, { "begin_idx": "714", "end_idx": "740", "entity_id": "D009084", "entity_type": "Disease", "text_name": "Sanfilippo syndrome type B" }, { "begin_idx": "122", "end_idx": "149", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive disease" }, { "begin_idx": "875", "end_idx": "880", "entity_id": "4669", "entity_type": "Gene", "text_name": "NAGLU" }, { "begin_idx": "0", "end_idx": "5", "entity_id": "4669", "entity_type": "Gene", "text_name": "NAGLU" }, { "begin_idx": "309", "end_idx": "314", "entity_id": "4669", "entity_type": "Gene", "text_name": "NAGLU" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "875", "end_idx": "880", "entity_id": "4669", "entity_type": "Gene", "text_name": "NAGLU" }, { "begin_idx": "875", "end_idx": "880", "entity_id": "4669", "entity_type": "Gene", "text_name": "NAGLU" } ]
[ { "begin_idx": "83", "end_idx": "110", "entity_id": "D009084", "entity_type": "Disease", "text_name": "mucopolysaccharidosis III B" }, { "begin_idx": "122", "end_idx": "149", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive disease" } ]
[ "NAGLU", "NAGLU" ]
[ "mucopolysaccharidosis III B", "autosomal recessive disease" ]
9446652
Antibodies from patients with heparin-induced thrombocytopenia/thrombosis recognize different epitopes on heparin: platelet factor 4.
Antibodies associated with heparin-induced thrombocytopenia/ thrombosis (HITT) are now thought to be specific for complexes formed between heparin and platelet factor 4 (PF4), a basic protein found normally in platelet alpha granules. How these antibodies cause thrombocytopenia and, in some patients, thrombosis, is not fully understood, in part because purified antibodies that could be labeled and used as probes to characterize target epitopes have not been available. We developed a novel method for antibody purification involving binding to and elution from PF4 complexed to heparin immobilized by end-linkage (EL) to a solid phase. Isolated antibodies were functional and after biotinylation, reacted with heparin: PF4 complexes in the same manner as unlabeled antibodies. Using these probes, we found that antibodies from 11 patients with HITT recognized two, and probably three, distinct sites on heparin: PF4 complexes. The antibodies did not bind to PF4 complexed with heparin immobilized by multiple chemical cross-linkages, suggesting that the heparin molecule must be in a flexible, relatively unconstrained state to react with PF4 in such a way as to create sites for HITT antibody binding.
[ { "begin_idx": "46", "end_idx": "62", "entity_id": "D013921", "entity_type": "Disease", "text_name": "thrombocytopenia" }, { "begin_idx": "161", "end_idx": "205", "entity_id": "D013921", "entity_type": "Disease", "text_name": "heparin-induced thrombocytopenia/ thrombosis" }, { "begin_idx": "207", "end_idx": "211", "entity_id": "D013921", "entity_type": "Disease", "text_name": "HITT" }, { "begin_idx": "396", "end_idx": "412", "entity_id": "D013921", "entity_type": "Disease", "text_name": "thrombocytopenia" }, { "begin_idx": "982", "end_idx": "986", "entity_id": "D013921", "entity_type": "Disease", "text_name": "HITT" }, { "begin_idx": "1318", "end_idx": "1322", "entity_id": "D013921", "entity_type": "Disease", "text_name": "HITT" }, { "begin_idx": "63", "end_idx": "73", "entity_id": "D013927", "entity_type": "Disease", "text_name": "thrombosis" }, { "begin_idx": "436", "end_idx": "446", "entity_id": "D013927", "entity_type": "Disease", "text_name": "thrombosis" }, { "begin_idx": "115", "end_idx": "132", "entity_id": "5196", "entity_type": "Gene", "text_name": "platelet factor 4" }, { "begin_idx": "285", "end_idx": "302", "entity_id": "5196", "entity_type": "Gene", "text_name": "platelet factor 4" }, { "begin_idx": "304", "end_idx": "307", "entity_id": "5196", "entity_type": "Gene", "text_name": "PF4" }, { "begin_idx": "699", "end_idx": "702", "entity_id": "5196", "entity_type": "Gene", "text_name": "PF4" }, { "begin_idx": "857", "end_idx": "860", "entity_id": "5196", "entity_type": "Gene", "text_name": "PF4" }, { "begin_idx": "1050", "end_idx": "1053", "entity_id": "5196", "entity_type": "Gene", "text_name": "PF4" }, { "begin_idx": "1096", "end_idx": "1099", "entity_id": "5196", "entity_type": "Gene", "text_name": "PF4" }, { "begin_idx": "1277", "end_idx": "1280", "entity_id": "5196", "entity_type": "Gene", "text_name": "PF4" } ]
[ "Yes", "Yes" ]
[ true, true ]
[ { "begin_idx": "115", "end_idx": "132", "entity_id": "5196", "entity_type": "Gene", "text_name": "platelet factor 4" }, { "begin_idx": "115", "end_idx": "132", "entity_id": "5196", "entity_type": "Gene", "text_name": "platelet factor 4" } ]
[ { "begin_idx": "63", "end_idx": "73", "entity_id": "D013927", "entity_type": "Disease", "text_name": "thrombosis" }, { "begin_idx": "161", "end_idx": "205", "entity_id": "D013921", "entity_type": "Disease", "text_name": "heparin-induced thrombocytopenia/ thrombosis" } ]
[ "platelet factor 4", "platelet factor 4" ]
[ "thrombosis", "heparin-induced thrombocytopenia/ thrombosis" ]
9452109
Mutational analyses of AVPR2 gene in three Japanese families with X-linked nephrogenic diabetes insipidus: two recurrent mutations, R137H and deltaV278, caused by the hypermutability at CpG dinucleotides.
[ { "begin_idx": "66", "end_idx": "105", "entity_id": "D018500", "entity_type": "Disease", "text_name": "X-linked nephrogenic diabetes insipidus" }, { "begin_idx": "23", "end_idx": "28", "entity_id": "554", "entity_type": "Gene", "text_name": "AVPR2" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "23", "end_idx": "28", "entity_id": "554", "entity_type": "Gene", "text_name": "AVPR2" } ]
[ { "begin_idx": "66", "end_idx": "105", "entity_id": "D018500", "entity_type": "Disease", "text_name": "X-linked nephrogenic diabetes insipidus" } ]
[ "AVPR2" ]
[ "X-linked nephrogenic diabetes insipidus" ]
9455977
Familial ALS is associated with mutations in all exons of SOD1: a novel mutation in exon 3 (Gly72Ser).
Mutations of the SOD1 gene, which encodes the enzyme copper/zinc superoxide dismutase, are associated with familial amyotrophic lateral sclerosis (ALS). SOD1 consists of five exons, and over 50 different mutations have been described involving exons 1,2,4 and 5. The absence of mutations in exon 3 has been attributed to a critical function of this exon, its integrity being necessary for the toxic effect of mutant SOD1, and it has been suggested that such mutations may be lethal rather than leading to adult onset disease. We identified the heterozygote mutation Gly72Ser (exon 3) in a family with two individuals affected by ALS. SOD enzyme activity was reduced by 45% when measured in erythrocytes indicating reduced enzyme activity, or reduced stability of the mutant protein. These findings indicate that exon 3 is not a privileged region from mutation; that all five exons should be investigated when seeking SOD1 mutations in human disease; and may help in a better understanding of the pathogenicity of these mutations in ALS.
[ { "begin_idx": "0", "end_idx": "12", "entity_id": "C531617", "entity_type": "Disease", "text_name": "Familial ALS" }, { "begin_idx": "210", "end_idx": "248", "entity_id": "C531617", "entity_type": "Disease", "text_name": "familial amyotrophic lateral sclerosis" }, { "begin_idx": "250", "end_idx": "253", "entity_id": "D000690", "entity_type": "Disease", "text_name": "ALS" }, { "begin_idx": "732", "end_idx": "735", "entity_id": "D000690", "entity_type": "Disease", "text_name": "ALS" }, { "begin_idx": "1135", "end_idx": "1138", "entity_id": "D000690", "entity_type": "Disease", "text_name": "ALS" }, { "begin_idx": "58", "end_idx": "62", "entity_id": "6647", "entity_type": "Gene", "text_name": "SOD1" }, { "begin_idx": "120", "end_idx": "124", "entity_id": "6647", "entity_type": "Gene", "text_name": "SOD1" }, { "begin_idx": "256", "end_idx": "260", "entity_id": "6647", "entity_type": "Gene", "text_name": "SOD1" }, { "begin_idx": "519", "end_idx": "523", "entity_id": "6647", "entity_type": "Gene", "text_name": "SOD1" }, { "begin_idx": "1020", "end_idx": "1024", "entity_id": "6647", "entity_type": "Gene", "text_name": "SOD1" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "58", "end_idx": "62", "entity_id": "6647", "entity_type": "Gene", "text_name": "SOD1" }, { "begin_idx": "58", "end_idx": "62", "entity_id": "6647", "entity_type": "Gene", "text_name": "SOD1" } ]
[ { "begin_idx": "210", "end_idx": "248", "entity_id": "C531617", "entity_type": "Disease", "text_name": "familial amyotrophic lateral sclerosis" }, { "begin_idx": "1135", "end_idx": "1138", "entity_id": "D000690", "entity_type": "Disease", "text_name": "ALS" } ]
[ "SOD1", "SOD1" ]
[ "familial amyotrophic lateral sclerosis", "ALS" ]
9455993
Molecular characteristics of cocaine-induced cardiomyopathy in rats.
Cocaine abuse induces severe cardiomyopathy. To investigate the molecular effects of acute and prolonged administration of cocaine, mRNAs encoding markers of either mechanical overload, as atrial natriuretic factor (ANF) and alpha- and beta-myosin heavy chains, or fibrosis as type I and III procollagens, were quantitated in the left ventricle of rats 4 h after one injection of cocaine (40 mg/kg, n = 7), or 14 (n = 15) and 28 days (n = 10) after chronic infusion of cocaine (40 mg/kg per day). Plasma cocaine and benzylecgonine concentrations were both significantly augmented during the infusion while plasma levels of triiodothyronine and thyroxine were lowered. Acute injection of cocaine induced ANF gene expression. Cocaine treatment during 28 days resulted in left ventricular hypertrophy (+ 20% after 24 days, P < 0.05) with normal blood pressure, associated with an accumulation of mRNAs encoding ANF and type I and III collagens (+66% and +55%, P < 0.05). Such a chronic treatment also induced a shift from the alpha- to the beta-myosin heavy chain gene expression (-40% and +50%, P < 0.05). In conclusion, cocaine activates markers of both hemodynamic overload and fibrosis. Such an activation may result from direct and/or indirect effects of the drug such as myocardial ischemia, mechanical overload and/or hypothyroidism.
[ { "begin_idx": "334", "end_idx": "342", "entity_id": "D005355", "entity_type": "Disease", "text_name": "fibrosis" }, { "begin_idx": "1247", "end_idx": "1255", "entity_id": "D005355", "entity_type": "Disease", "text_name": "fibrosis" }, { "begin_idx": "1391", "end_idx": "1405", "entity_id": "D007037", "entity_type": "Disease", "text_name": "hypothyroidism" }, { "begin_idx": "45", "end_idx": "59", "entity_id": "D009202", "entity_type": "Disease", "text_name": "cardiomyopathy" }, { "begin_idx": "98", "end_idx": "112", "entity_id": "D009202", "entity_type": "Disease", "text_name": "cardiomyopathy" }, { "begin_idx": "1343", "end_idx": "1362", "entity_id": "D017202", "entity_type": "Disease", "text_name": "myocardial ischemia" }, { "begin_idx": "838", "end_idx": "866", "entity_id": "D017379", "entity_type": "Disease", "text_name": "left ventricular hypertrophy" }, { "begin_idx": "69", "end_idx": "82", "entity_id": "D019970", "entity_type": "Disease", "text_name": "Cocaine abuse" }, { "begin_idx": "346", "end_idx": "352", "entity_id": "D020754", "entity_type": "Disease", "text_name": "type I" }, { "begin_idx": "985", "end_idx": "991", "entity_id": "D020754", "entity_type": "Disease", "text_name": "type I" }, { "begin_idx": "294", "end_idx": "329", "entity_id": "4625", "entity_type": "Gene", "text_name": "alpha- and beta-myosin heavy chains" }, { "begin_idx": "1106", "end_idx": "1129", "entity_id": "4625", "entity_type": "Gene", "text_name": "beta-myosin heavy chain" }, { "begin_idx": "258", "end_idx": "283", "entity_id": "4878", "entity_type": "Gene", "text_name": "atrial natriuretic factor" }, { "begin_idx": "285", "end_idx": "288", "entity_id": "4878", "entity_type": "Gene", "text_name": "ANF" }, { "begin_idx": "772", "end_idx": "775", "entity_id": "4878", "entity_type": "Gene", "text_name": "ANF" }, { "begin_idx": "977", "end_idx": "980", "entity_id": "4878", "entity_type": "Gene", "text_name": "ANF" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "258", "end_idx": "283", "entity_id": "4878", "entity_type": "Gene", "text_name": "atrial natriuretic factor" }, { "begin_idx": "1106", "end_idx": "1129", "entity_id": "4625", "entity_type": "Gene", "text_name": "beta-myosin heavy chain" } ]
[ { "begin_idx": "838", "end_idx": "866", "entity_id": "D017379", "entity_type": "Disease", "text_name": "left ventricular hypertrophy" }, { "begin_idx": "985", "end_idx": "991", "entity_id": "D020754", "entity_type": "Disease", "text_name": "type I" } ]
[ "atrial natriuretic factor", "beta-myosin heavy chain" ]
[ "left ventricular hypertrophy", "type I" ]
9457942
A new molecular variant of luteinizing hormone associated with female infertility.
OBJECTIVE: To investigate whether the newly described G1502 to A1502 mutation in exon 3 of the LH beta-subunit gene, causing the amino acid substitution of Ser102 for Gly102, is related to female infertility. DESIGN: Screening of fertile and infertile women for the G1502 to A1502 mutation in the LH beta-subunit gene. SETTING: Clinics and laboratories of the National University Hospital obstetrics and gynecology department, Singapore. PATIENT(S): Two hundred twelve healthy fertile women; 40 infertile women with menstrual disorders, polycystic ovary syndrome, and endometriosis; and 12 women with idiopathic infertility. INTERVENTION(S): Exon 3 of the LH beta-subunit gene was analyzed using polymerase chain reaction (PCR), restriction fragment length polymorphism (RFLP), and PCR-mediated direct DNA sequencing. MAIN OUTCOME MEASURE(S): The PCR products of patients were analyzed by RFLP, and the results were compared with those of fertile controls. DNA sequencing radiographs were compared between two mutation-bearing patients and four controls. RESULT(S): The mutation was identified in only two infertile women with endometriosis; other women studied were found to be negative for this mutation. CONCLUSION(S): The missense mutation in the LH beta-subunit gene may be implicated in female infertility, possibly endometriosis-associated infertility in some women.
[ { "begin_idx": "651", "end_idx": "664", "entity_id": "D004715", "entity_type": "Disease", "text_name": "endometriosis" }, { "begin_idx": "1210", "end_idx": "1223", "entity_id": "D004715", "entity_type": "Disease", "text_name": "endometriosis" }, { "begin_idx": "1405", "end_idx": "1418", "entity_id": "D004715", "entity_type": "Disease", "text_name": "endometriosis" }, { "begin_idx": "695", "end_idx": "706", "entity_id": "D007246", "entity_type": "Disease", "text_name": "infertility" }, { "begin_idx": "1430", "end_idx": "1441", "entity_id": "D007246", "entity_type": "Disease", "text_name": "infertility" }, { "begin_idx": "63", "end_idx": "81", "entity_id": "D007247", "entity_type": "Disease", "text_name": "female infertility" }, { "begin_idx": "272", "end_idx": "290", "entity_id": "D007247", "entity_type": "Disease", "text_name": "female infertility" }, { "begin_idx": "1376", "end_idx": "1394", "entity_id": "D007247", "entity_type": "Disease", "text_name": "female infertility" }, { "begin_idx": "599", "end_idx": "618", "entity_id": "D008599", "entity_type": "Disease", "text_name": "menstrual disorders" }, { "begin_idx": "620", "end_idx": "645", "entity_id": "D011085", "entity_type": "Disease", "text_name": "polycystic ovary syndrome" }, { "begin_idx": "178", "end_idx": "185", "entity_id": "3972", "entity_type": "Gene", "text_name": "LH beta" }, { "begin_idx": "380", "end_idx": "387", "entity_id": "3972", "entity_type": "Gene", "text_name": "LH beta" }, { "begin_idx": "739", "end_idx": "746", "entity_id": "3972", "entity_type": "Gene", "text_name": "LH beta" }, { "begin_idx": "1334", "end_idx": "1341", "entity_id": "3972", "entity_type": "Gene", "text_name": "LH beta" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "178", "end_idx": "185", "entity_id": "3972", "entity_type": "Gene", "text_name": "LH beta" }, { "begin_idx": "178", "end_idx": "185", "entity_id": "3972", "entity_type": "Gene", "text_name": "LH beta" } ]
[ { "begin_idx": "63", "end_idx": "81", "entity_id": "D007247", "entity_type": "Disease", "text_name": "female infertility" }, { "begin_idx": "1405", "end_idx": "1418", "entity_id": "D004715", "entity_type": "Disease", "text_name": "endometriosis" } ]
[ "LH beta", "LH beta" ]
[ "female infertility", "endometriosis" ]
9458110
Screening of the TAP1 gene by denaturing gradient gel electrophoresis in insulin-dependent diabetes mellitus: detection and comparison of new polymorphisms between patients and controls.
New protective or disease-associated polymorphisms in the TAP1 gene were sought in insulin-dependent diabetes mellitus (IDDM) patients with the use of denaturing gradient gel electrophoresis (DGGE) screening of genomic DNA. The TAP1 gene is located in the human leukocyte antigen (HLA) class II region of the genome and encodes components of a peptide transporter essential for antigen presentation by HLA class I molecules. Fragments of TAP1 corresponding to the 5' promoter, each of the 11 exons (with portions of adjacent intronic regions) and the 3' flanking region were amplified by the polymerase chain reaction and then subjected to DGGE. DNA fragments of TAP1 yielded DGGE bands with patterns whose frequencies differed between IDDM patients and controls. Specific DGGE band patterns with fragments corresponding to the promoter, exons or introns 3, 6, 7, 8, 9 or 10 of TAP1 were detected exclusively in either patients or controls. Sequencing of TAP1 fragments encompassing exon 7 gave rise to a DGGE band pattern exclusively observed in an IDDM patient and sequencing revealed a previously unidentified polymorphisms at codon 518 (GTC-->ATC, Val-->Ile). Another unique polymorphism uncovered by DGGE revealed by sequencing a polymorphism in intron 2 in a diabetic patient. The genotypes of additional HLA class II matched patients and controls were determined with regard to five exonic and one intronic TAP1 polymorphism. A 10 base pair intronic insertion in intron 9 was exclusively identified in controls and missing from patients (P = 0.017). Further large population-based studies may reveal whether these newly identified at risk or protective TAP1 variants confer markers of statistical risk in diverse population groups.
[ { "begin_idx": "1452", "end_idx": "1460", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "73", "end_idx": "108", "entity_id": "D003922", "entity_type": "Disease", "text_name": "insulin-dependent diabetes mellitus" }, { "begin_idx": "270", "end_idx": "305", "entity_id": "D003922", "entity_type": "Disease", "text_name": "insulin-dependent diabetes mellitus" }, { "begin_idx": "307", "end_idx": "311", "entity_id": "D003922", "entity_type": "Disease", "text_name": "IDDM" }, { "begin_idx": "923", "end_idx": "927", "entity_id": "D003922", "entity_type": "Disease", "text_name": "IDDM" }, { "begin_idx": "1237", "end_idx": "1241", "entity_id": "D003922", "entity_type": "Disease", "text_name": "IDDM" }, { "begin_idx": "17", "end_idx": "21", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "245", "end_idx": "249", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "415", "end_idx": "419", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "625", "end_idx": "629", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "850", "end_idx": "854", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "1065", "end_idx": "1069", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "1142", "end_idx": "1146", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "1601", "end_idx": "1605", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "1847", "end_idx": "1851", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "17", "end_idx": "21", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" }, { "begin_idx": "850", "end_idx": "854", "entity_id": "6890", "entity_type": "Gene", "text_name": "TAP1" } ]
[ { "begin_idx": "73", "end_idx": "108", "entity_id": "D003922", "entity_type": "Disease", "text_name": "insulin-dependent diabetes mellitus" }, { "begin_idx": "1452", "end_idx": "1460", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" } ]
[ "TAP1", "TAP1" ]
[ "insulin-dependent diabetes mellitus", "diabetic" ]
9466979
Overexpression of leukotriene C4 synthase in bronchial biopsies from patients with aspirin-intolerant asthma.
Aspirin causes bronchoconstriction in aspirin-intolerant asthma (AIA) patients by triggering cysteinyl-leukotriene (cys-LT) production, probably by removing PGE2-dependent inhibition. To investigate why aspirin does not cause bronchoconstriction in all individuals, we immunostained enzymes of the leukotriene and prostanoid pathways in bronchial biopsies from AIA patients, aspirin-tolerant asthma (ATA) patients, and normal (N) subjects. Counts of cells expressing the terminal enzyme for cys-LT synthesis, LTC4 synthase, were fivefold higher in AIA biopsies (11.5+/-2.2 cells/mm2, n = 10) than in ATA biopsies (2.2+/-0.7, n = 10; P = 0. 0006) and 18-fold higher than in N biopsies (0.6+/-0.4, n = 9; P = 0. 0002). Immunostaining for 5-lipoxygenase, its activating protein (FLAP), LTA4 hydrolase, cyclooxygenase (COX)-1, and COX-2 did not differ. Enhanced baseline cys-LT levels in bronchoalveolar lavage (BAL) fluid of AIA patients correlated uniquely with bronchial counts of LTC4 synthase+ cells (rho = 0.83, P = 0.01). Lysine-aspirin challenge released additional cys-LTs into BAL fluid in AIA patients (200+/-120 pg/ml, n = 8) but not in ATA patients (0. 7+/-5.1, n = 5; P = 0.007). Bronchial responsiveness to lysine-aspirin correlated exclusively with LTC4 synthase+ cell counts (rho = -0.63, P = 0.049, n = 10). Aspirin may remove PGE2-dependent suppression in all subjects, but only in AIA patients does increased bronchial expression of LTC4 synthase allow marked overproduction of cys-LTs leading to bronchoconstriction.
[ { "begin_idx": "83", "end_idx": "108", "entity_id": "C565935", "entity_type": "Disease", "text_name": "aspirin-intolerant asthma" }, { "begin_idx": "148", "end_idx": "173", "entity_id": "C565935", "entity_type": "Disease", "text_name": "aspirin-intolerant asthma" }, { "begin_idx": "175", "end_idx": "178", "entity_id": "C565935", "entity_type": "Disease", "text_name": "AIA" }, { "begin_idx": "471", "end_idx": "474", "entity_id": "C565935", "entity_type": "Disease", "text_name": "AIA" }, { "begin_idx": "658", "end_idx": "661", "entity_id": "C565935", "entity_type": "Disease", "text_name": "AIA" }, { "begin_idx": "1032", "end_idx": "1035", "entity_id": "C565935", "entity_type": "Disease", "text_name": "AIA" }, { "begin_idx": "1206", "end_idx": "1209", "entity_id": "C565935", "entity_type": "Disease", "text_name": "AIA" }, { "begin_idx": "1507", "end_idx": "1510", "entity_id": "C565935", "entity_type": "Disease", "text_name": "AIA" }, { "begin_idx": "485", "end_idx": "508", "entity_id": "D055963", "entity_type": "Disease", "text_name": "aspirin-tolerant asthma" }, { "begin_idx": "510", "end_idx": "513", "entity_id": "D055963", "entity_type": "Disease", "text_name": "ATA" }, { "begin_idx": "710", "end_idx": "713", "entity_id": "D055963", "entity_type": "Disease", "text_name": "ATA" }, { "begin_idx": "1255", "end_idx": "1258", "entity_id": "D055963", "entity_type": "Disease", "text_name": "ATA" }, { "begin_idx": "893", "end_idx": "907", "entity_id": "4048", "entity_type": "Gene", "text_name": "LTA4 hydrolase" }, { "begin_idx": "18", "end_idx": "41", "entity_id": "4056", "entity_type": "Gene", "text_name": "leukotriene C4 synthase" }, { "begin_idx": "619", "end_idx": "632", "entity_id": "4056", "entity_type": "Gene", "text_name": "LTC4 synthase" }, { "begin_idx": "1090", "end_idx": "1103", "entity_id": "4056", "entity_type": "Gene", "text_name": "LTC4 synthase" }, { "begin_idx": "1371", "end_idx": "1384", "entity_id": "4056", "entity_type": "Gene", "text_name": "LTC4 synthase" }, { "begin_idx": "1559", "end_idx": "1572", "entity_id": "4056", "entity_type": "Gene", "text_name": "LTC4 synthase" }, { "begin_idx": "909", "end_idx": "931", "entity_id": "4512", "entity_type": "Gene", "text_name": "cyclooxygenase (COX)-1" }, { "begin_idx": "937", "end_idx": "942", "entity_id": "4513", "entity_type": "Gene", "text_name": "COX-2" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "18", "end_idx": "41", "entity_id": "4056", "entity_type": "Gene", "text_name": "leukotriene C4 synthase" }, { "begin_idx": "937", "end_idx": "942", "entity_id": "4513", "entity_type": "Gene", "text_name": "COX-2" } ]
[ { "begin_idx": "485", "end_idx": "508", "entity_id": "D055963", "entity_type": "Disease", "text_name": "aspirin-tolerant asthma" }, { "begin_idx": "1507", "end_idx": "1510", "entity_id": "C565935", "entity_type": "Disease", "text_name": "AIA" } ]
[ "leukotriene C4 synthase", "COX-2" ]
[ "aspirin-tolerant asthma", "AIA" ]
9482579
Detection of five novel mutations of the cystic fibrosis transmembrane regulator (CFTR) gene in Pakistani patients with cystic fibrosis: Y569D, Q98X, 296+12(T>C), 1161delC and 621+2(T>C).
We analysed DNA samples from 26 Pakistani patients with cystic fibrosis (CF) living in the United Kingdom (14 from patients residing in the north west of England, who were referred directly to the North West Regional Molecular Genetics Laboratory, and 12 from other regional molecular genetics laboratories). Of 56 mutations seen in native U.K. CF patients, only DeltaF508, R709X, and 2184insA were detected in the Pakistani patients. Combined SSCP/Heteroduplex analysis, DGGE, and direct DNA cycle sequencing revealed five novel mutations: Y569D, Q98X, 296+12(T>C), 1161delC, and 621+2(T>C), which appear to be specific to Pakistani CF families. In addition, a novel polymorphism, 297-67(A/C), and three previously described rare mutations, 1525-1(G>A), R560S, and 1898+1(G>T), were detected. In the 14 Pakistani CF patients from the north west of England, DeltaF508 accounted for approximately 32% (9/28 chromosomes) and the overall detection rate of CF mutations in this group was approximately 86% (24/28 chromosomes).
[ { "begin_idx": "120", "end_idx": "135", "entity_id": "D003550", "entity_type": "Disease", "text_name": "cystic fibrosis" }, { "begin_idx": "244", "end_idx": "259", "entity_id": "D003550", "entity_type": "Disease", "text_name": "cystic fibrosis" }, { "begin_idx": "261", "end_idx": "263", "entity_id": "D003550", "entity_type": "Disease", "text_name": "CF" }, { "begin_idx": "533", "end_idx": "535", "entity_id": "D003550", "entity_type": "Disease", "text_name": "CF" }, { "begin_idx": "822", "end_idx": "824", "entity_id": "D003550", "entity_type": "Disease", "text_name": "CF" }, { "begin_idx": "1002", "end_idx": "1004", "entity_id": "D003550", "entity_type": "Disease", "text_name": "CF" }, { "begin_idx": "1141", "end_idx": "1143", "entity_id": "D003550", "entity_type": "Disease", "text_name": "CF" }, { "begin_idx": "41", "end_idx": "80", "entity_id": "1080", "entity_type": "Gene", "text_name": "cystic fibrosis transmembrane regulator" }, { "begin_idx": "82", "end_idx": "86", "entity_id": "1080", "entity_type": "Gene", "text_name": "CFTR" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "41", "end_idx": "80", "entity_id": "1080", "entity_type": "Gene", "text_name": "cystic fibrosis transmembrane regulator" } ]
[ { "begin_idx": "120", "end_idx": "135", "entity_id": "D003550", "entity_type": "Disease", "text_name": "cystic fibrosis" } ]
[ "cystic fibrosis transmembrane regulator" ]
[ "cystic fibrosis" ]
9489796
X-linked spastic paraplegia due to a mutation (C506T; Ser169Phe) in exon 4 of the proteolipid protein gene (PLP).
A transition C506T was found in exon 4 of the proteolipid protein gene of a boy with spastic paraplegia. This mutation resulted in the substitution of phenylalanine for serine 169, which is in the third transmembrane domain of the proteolipid protein molecule. The mutation apparently arose de novo, as it was absent from his mother.
[ { "begin_idx": "0", "end_idx": "27", "entity_id": "C536857", "entity_type": "Disease", "text_name": "X-linked spastic paraplegia" }, { "begin_idx": "199", "end_idx": "217", "entity_id": "C536857", "entity_type": "Disease", "text_name": "spastic paraplegia" }, { "begin_idx": "82", "end_idx": "106", "entity_id": "5354", "entity_type": "Gene", "text_name": "proteolipid protein gene" }, { "begin_idx": "108", "end_idx": "111", "entity_id": "5354", "entity_type": "Gene", "text_name": "PLP" }, { "begin_idx": "160", "end_idx": "184", "entity_id": "5354", "entity_type": "Gene", "text_name": "proteolipid protein gene" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "82", "end_idx": "106", "entity_id": "5354", "entity_type": "Gene", "text_name": "proteolipid protein gene" } ]
[ { "begin_idx": "0", "end_idx": "27", "entity_id": "C536857", "entity_type": "Disease", "text_name": "X-linked spastic paraplegia" } ]
[ "proteolipid protein gene" ]
[ "X-linked spastic paraplegia" ]
9491812
A family based association study of T102C polymorphism in 5HT2A and schizophrenia plus identification of new polymorphisms in the promoter.
Several studies have shown an association between schizophrenia and the C allele of a T-C polymorphism at nucleotide 102 and the 5HT2A receptor gene. In the present study we observed this association in a sample of 63 parent/offspring trios where the proband received a diagnosis of DSM-III-R schizophrenia using TDT analysis (chi2 = 6.26, P= 0.006, chi2 = 9.00, P=0.001 when one affected offspring was selected at random from each family, suggesting that the results are due to association rather than linkage). There was no significant difference between the transmission of C102 from heterozygous fathers and mothers, which fails to support a role for genomic imprinting in this effect. T102C does not result in an alteration of the amino acid sequence of the protein. We therefore screened the promoter of 5HT2A for polymorphisms using single-strand confirmation polymorphism analysis. An A-G polymorphism at -1438 that creates an HpaII restriction site was identified. This was found to be in complete linkage disequilibrium with T102C and is hence a candidate for the pathogenic variant in schizophrenia. Functional analysis of A-1438G using luciferase assay demonstrated significant basal promoter activity in 5HT2A expressing HeLa cells by both the A and G variants. However, comparison of the A and G variants showed no significant differences in basal activity nor when promoter activity was induced by cAMP and protein kinase C-dependent mechanisms.
[ { "begin_idx": "68", "end_idx": "81", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "190", "end_idx": "203", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "433", "end_idx": "446", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "1236", "end_idx": "1249", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "59", "end_idx": "63", "entity_id": "3356", "entity_type": "Gene", "text_name": "HT2A" }, { "begin_idx": "269", "end_idx": "283", "entity_id": "3356", "entity_type": "Gene", "text_name": "5HT2A receptor" }, { "begin_idx": "950", "end_idx": "955", "entity_id": "3356", "entity_type": "Gene", "text_name": "5HT2A" }, { "begin_idx": "1358", "end_idx": "1362", "entity_id": "3356", "entity_type": "Gene", "text_name": "HT2A" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "269", "end_idx": "283", "entity_id": "3356", "entity_type": "Gene", "text_name": "5HT2A receptor" } ]
[ { "begin_idx": "68", "end_idx": "81", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" } ]
[ "5HT2A receptor" ]
[ "schizophrenia" ]
9498106
Association of angiotensin II type 1 receptor polymorphism with resistant essential hypertension.
Angiotensin II type 1 receptor (AT1) mediates the vasoconstrictive and growth-promoting effect of angiotensin II in humans. It has been reported that a polymorphism of the AT1 gene (an A/C transversion at position 1166: A-C1166) occurs more frequently in resistant hypertensives taking two or more antihypertensive drugs. On the contrary, a recent study of the influence of the A-C1166 polymorphism on aortic stiffness demonstrated that the distribution of the genotypes did not differ between normotensive and hypertensive subjects. In addition, a recent population-based survey of Caucasian hypertensives reported lower blood pressure values in CC homozygotes than in heterozygotes and AA homozygotes. Because of these controversial results and the lack of a sufficient amount of data the present study was designed to assess the contribution of the AT, gene A-C1166 polymorphism to resistant essential hypertension. Forty-eight subjects with resistant essential hypertension (HT) and 48 normotensive (NT), age and sex-adjusted controls (from a population of 300 healthy blood donors) were selected. All subjects were genotyped for the A-C1166 polymorphism in the 3'-UTR of the AT1 gene using PCR-based techniques. The influence of genotype on blood pressure (BP) was investigated using ANOVA Randomized Complete Block (ANOVA RCB) design according to sex, age and BMI. There were no significant differences in allele or genotype frequencies between HT and NT subjects (X2 = 0.61; P = NS). In HT subjects higher values of systolic blood pressure were associated with the C allele of the AT1 gene only in older and overweight patients (P < 0.001 and P < 0.001, respectively). Also in HT patients an association between the presence of the C allele of the AT1 gene and higher values of diastolic blood pressure was present in overweight patients (P = 0.001). These results suggest that in resistant hypertensive subjects the AT1 A-C1166 polymorphism is potentially involved in the regulation of blood pressure. As the effects of genotypes on blood pressure are pronounced in older and overweight subjects this polymorphism may amplify the effects of age and BMI on resistant essential hypertension.
[ { "begin_idx": "950", "end_idx": "952", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" }, { "begin_idx": "84", "end_idx": "96", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "363", "end_idx": "376", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertensives" }, { "begin_idx": "609", "end_idx": "621", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertensive" }, { "begin_idx": "691", "end_idx": "704", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertensives" }, { "begin_idx": "1003", "end_idx": "1015", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "1063", "end_idx": "1075", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "1077", "end_idx": "1079", "entity_id": "D006973", "entity_type": "Disease", "text_name": "HT" }, { "begin_idx": "1549", "end_idx": "1551", "entity_id": "D006973", "entity_type": "Disease", "text_name": "HT" }, { "begin_idx": "1592", "end_idx": "1594", "entity_id": "D006973", "entity_type": "Disease", "text_name": "HT" }, { "begin_idx": "1782", "end_idx": "1784", "entity_id": "D006973", "entity_type": "Disease", "text_name": "HT" }, { "begin_idx": "1996", "end_idx": "2008", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertensive" }, { "begin_idx": "2282", "end_idx": "2294", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "196", "end_idx": "210", "entity_id": "183", "entity_type": "Gene", "text_name": "angiotensin II" }, { "begin_idx": "15", "end_idx": "45", "entity_id": "185", "entity_type": "Gene", "text_name": "angiotensin II type 1 receptor" }, { "begin_idx": "98", "end_idx": "128", "entity_id": "185", "entity_type": "Gene", "text_name": "Angiotensin II type 1 receptor" }, { "begin_idx": "130", "end_idx": "133", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" }, { "begin_idx": "270", "end_idx": "273", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" }, { "begin_idx": "1278", "end_idx": "1281", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" }, { "begin_idx": "1686", "end_idx": "1689", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" }, { "begin_idx": "1853", "end_idx": "1856", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" }, { "begin_idx": "2022", "end_idx": "2025", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "15", "end_idx": "45", "entity_id": "185", "entity_type": "Gene", "text_name": "angiotensin II type 1 receptor" }, { "begin_idx": "1686", "end_idx": "1689", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" } ]
[ { "begin_idx": "363", "end_idx": "376", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertensives" }, { "begin_idx": "950", "end_idx": "952", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" } ]
[ "angiotensin II type 1 receptor", "AT1" ]
[ "hypertensives", "AT" ]
9501220
Mutations in the RPE65 gene in patients with autosomal recessive retinitis pigmentosa or leber congenital amaurosis.
RPE65 is a protein of unknown function expressed specifically by the retinal pigment epithelium. We examined all 14 exons of this gene in 147 unrelated patients with autosomal recessive retinitis pigmentosa (RP), in 15 patients with isolate RP, and in 45 patients with Leber congenital amaurosis (LCA). Sequence anomalies that were likely to be pathogenic were found in two patients with recessive RP, in one patient with isolate RP recategorized as recessive, and in seven patients with LCA. Cosegregation analysis in each available family showed that all affected individuals were either homozygotes or compound heterozygotes and that all unaffected individuals were either heterozygote carriers or homozygous wild type. In one family, there was one instance of a new mutation not present in either parent of the affected individual. In another family, affected members with recessive RP in three branches (i.e., three distinct pairs of parents) were compound heterozygotes for the same two mutations or homozygous for one of them. Based on our results, mutations in the RPE65 gene appear to account for approximately 2% of cases of recessive RP and approximately 16% of cases of LCA.
[ { "begin_idx": "45", "end_idx": "85", "entity_id": "D012174", "entity_type": "Disease", "text_name": "autosomal recessive retinitis pigmentosa" }, { "begin_idx": "283", "end_idx": "323", "entity_id": "D012174", "entity_type": "Disease", "text_name": "autosomal recessive retinitis pigmentosa" }, { "begin_idx": "325", "end_idx": "327", "entity_id": "D012174", "entity_type": "Disease", "text_name": "RP" }, { "begin_idx": "358", "end_idx": "360", "entity_id": "D012174", "entity_type": "Disease", "text_name": "RP" }, { "begin_idx": "505", "end_idx": "517", "entity_id": "D012174", "entity_type": "Disease", "text_name": "recessive RP" }, { "begin_idx": "547", "end_idx": "549", "entity_id": "D012174", "entity_type": "Disease", "text_name": "RP" }, { "begin_idx": "994", "end_idx": "1006", "entity_id": "D012174", "entity_type": "Disease", "text_name": "recessive RP" }, { "begin_idx": "1252", "end_idx": "1264", "entity_id": "D012174", "entity_type": "Disease", "text_name": "recessive RP" }, { "begin_idx": "89", "end_idx": "115", "entity_id": "D057130", "entity_type": "Disease", "text_name": "leber congenital amaurosis" }, { "begin_idx": "386", "end_idx": "412", "entity_id": "D057130", "entity_type": "Disease", "text_name": "Leber congenital amaurosis" }, { "begin_idx": "414", "end_idx": "417", "entity_id": "D057130", "entity_type": "Disease", "text_name": "LCA" }, { "begin_idx": "605", "end_idx": "608", "entity_id": "D057130", "entity_type": "Disease", "text_name": "LCA" }, { "begin_idx": "1299", "end_idx": "1302", "entity_id": "D057130", "entity_type": "Disease", "text_name": "LCA" }, { "begin_idx": "17", "end_idx": "22", "entity_id": "6121", "entity_type": "Gene", "text_name": "RPE65" }, { "begin_idx": "117", "end_idx": "122", "entity_id": "6121", "entity_type": "Gene", "text_name": "RPE65" }, { "begin_idx": "1190", "end_idx": "1195", "entity_id": "6121", "entity_type": "Gene", "text_name": "RPE65" } ]
[ "Yes", "Yes" ]
[ true, true ]
[ { "begin_idx": "17", "end_idx": "22", "entity_id": "6121", "entity_type": "Gene", "text_name": "RPE65" }, { "begin_idx": "17", "end_idx": "22", "entity_id": "6121", "entity_type": "Gene", "text_name": "RPE65" } ]
[ { "begin_idx": "45", "end_idx": "85", "entity_id": "D012174", "entity_type": "Disease", "text_name": "autosomal recessive retinitis pigmentosa" }, { "begin_idx": "89", "end_idx": "115", "entity_id": "D057130", "entity_type": "Disease", "text_name": "leber congenital amaurosis" } ]
[ "RPE65", "RPE65" ]
[ "autosomal recessive retinitis pigmentosa", "leber congenital amaurosis" ]
9502217
Possible association of missense mutation (Gly[-63]Glu) of the neurotrophin-3 gene with Alzheimer's disease in Japanese.
Several lines of evidence have suggested a possible involvement of neurotrophic factors in the pathogenesis of neurodegenerative disease. We examined whether a missense mutation (Gly[-63]Glu) of the neurotrophin-3 (NT-3) gene is associated with Alzheimer's disease (AD) in a Japanese sample of 123 patients and 215 controls. We found that homozygotes or heterozygotes for the mutated type (Glu[-63]) were significantly more common among the patients than the controls (P = 0.013, odds ratio 1.77, 95% CI 1.12-2.79). The mutated type was more frequent among the patients than the controls (P = 0.011, odds ratio 1.63, 95%CI 1.11-2.38). This association between NT-3 and AD was more prominent among those who did not carry the epsilon4 allele of the apolipoprotein E gene than those who carried the epsilon4 allele. Our results suggest that the Glu(-63) allele of the NT-3 gene by itself or another mutation nearby which would be in linkage disequilibrium to the mutation is a risk factor for AD.
[ { "begin_idx": "88", "end_idx": "107", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "366", "end_idx": "385", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "387", "end_idx": "389", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "790", "end_idx": "792", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "1112", "end_idx": "1114", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "232", "end_idx": "257", "entity_id": "D019636", "entity_type": "Disease", "text_name": "neurodegenerative disease" }, { "begin_idx": "869", "end_idx": "885", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "63", "end_idx": "77", "entity_id": "4908", "entity_type": "Gene", "text_name": "neurotrophin-3" }, { "begin_idx": "320", "end_idx": "334", "entity_id": "4908", "entity_type": "Gene", "text_name": "neurotrophin-3" }, { "begin_idx": "336", "end_idx": "340", "entity_id": "4908", "entity_type": "Gene", "text_name": "NT-3" }, { "begin_idx": "781", "end_idx": "785", "entity_id": "4908", "entity_type": "Gene", "text_name": "NT-3" }, { "begin_idx": "987", "end_idx": "991", "entity_id": "4908", "entity_type": "Gene", "text_name": "NT-3" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "63", "end_idx": "77", "entity_id": "4908", "entity_type": "Gene", "text_name": "neurotrophin-3" }, { "begin_idx": "869", "end_idx": "885", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" } ]
[ { "begin_idx": "88", "end_idx": "107", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "1112", "end_idx": "1114", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" } ]
[ "neurotrophin-3", "apolipoprotein E" ]
[ "Alzheimer's disease", "AD" ]
9515794
High frequency of beta-catenin (ctnnb1) mutations in the colon tumors induced by two heterocyclic amines in the F344 rat.
Activating mutations in the beta-catenin (CTNNB1) gene corresponding to N-terminal phosphorylation sites in the protein have been implicated in the development of human colon cancer. To determine the possible involvement of such mutations during chemically induced colon carcinogenesis, we examined the corresponding region of Ctnnb1 in colon tumors induced in the F344 rat by two cooked meat heterocyclic amines, 2-amino-3-methylimidazo[4,5-f]quinoline and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP). All of the colon tumors induced by 2-amino-3-methylimidazo[4,5-f]quinoline that were examined (5 of 5) and 4 of 7 PhIP-induced colon tumors had mutations within or flanking codons corresponding to important phosphorylation sites in beta-catenin. None of the colon tumors bearing Ctnnb1 mutations had genetic changes in the Apc gene, and those that contained wild-type Ctnnb1 were known from our previous work to contain Apc mutations. The results provide evidence for a major role of the beta-catenin/Apc pathway in the development of heterocyclic amine-induced colon tumors and give further weight to the view that regulation of beta-catenin is critical to the tumor suppressive effects of Apc during colon carcinogenesis. In contrast, Ctnnb1 mutations were completely absent in 23 PhIP-induced mammary tumors, in accordance with recent work showing that human breast carcinomas lack mutations in CTNNB1.
[ { "begin_idx": "1498", "end_idx": "1515", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast carcinomas" }, { "begin_idx": "57", "end_idx": "69", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon tumors" }, { "begin_idx": "291", "end_idx": "303", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon cancer" }, { "begin_idx": "387", "end_idx": "407", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon carcinogenesis" }, { "begin_idx": "459", "end_idx": "471", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon tumors" }, { "begin_idx": "647", "end_idx": "659", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon tumors" }, { "begin_idx": "763", "end_idx": "775", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon tumors" }, { "begin_idx": "894", "end_idx": "906", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon tumors" }, { "begin_idx": "1198", "end_idx": "1210", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon tumors" }, { "begin_idx": "1338", "end_idx": "1358", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon carcinogenesis" }, { "begin_idx": "1298", "end_idx": "1303", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" }, { "begin_idx": "1432", "end_idx": "1446", "entity_id": "D009369", "entity_type": "Disease", "text_name": "mammary tumors" }, { "begin_idx": "18", "end_idx": "30", "entity_id": "1499", "entity_type": "Gene", "text_name": "beta-catenin" }, { "begin_idx": "32", "end_idx": "38", "entity_id": "1499", "entity_type": "Gene", "text_name": "ctnnb1" }, { "begin_idx": "150", "end_idx": "162", "entity_id": "1499", "entity_type": "Gene", "text_name": "beta-catenin" }, { "begin_idx": "164", "end_idx": "170", "entity_id": "1499", "entity_type": "Gene", "text_name": "CTNNB1" }, { "begin_idx": "449", "end_idx": "455", "entity_id": "1499", "entity_type": "Gene", "text_name": "Ctnnb1" }, { "begin_idx": "868", "end_idx": "880", "entity_id": "1499", "entity_type": "Gene", "text_name": "beta-catenin" }, { "begin_idx": "915", "end_idx": "921", "entity_id": "1499", "entity_type": "Gene", "text_name": "Ctnnb1" }, { "begin_idx": "1004", "end_idx": "1010", "entity_id": "1499", "entity_type": "Gene", "text_name": "Ctnnb1" }, { "begin_idx": "1124", "end_idx": "1136", "entity_id": "1499", "entity_type": "Gene", "text_name": "beta-catenin" }, { "begin_idx": "1266", "end_idx": "1278", "entity_id": "1499", "entity_type": "Gene", "text_name": "beta-catenin" }, { "begin_idx": "1373", "end_idx": "1379", "entity_id": "1499", "entity_type": "Gene", "text_name": "Ctnnb1" }, { "begin_idx": "1534", "end_idx": "1540", "entity_id": "1499", "entity_type": "Gene", "text_name": "CTNNB1" }, { "begin_idx": "959", "end_idx": "962", "entity_id": "324", "entity_type": "Gene", "text_name": "Apc" }, { "begin_idx": "1056", "end_idx": "1059", "entity_id": "324", "entity_type": "Gene", "text_name": "Apc" }, { "begin_idx": "1137", "end_idx": "1140", "entity_id": "324", "entity_type": "Gene", "text_name": "Apc" }, { "begin_idx": "1327", "end_idx": "1330", "entity_id": "324", "entity_type": "Gene", "text_name": "Apc" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "18", "end_idx": "30", "entity_id": "1499", "entity_type": "Gene", "text_name": "beta-catenin" }, { "begin_idx": "18", "end_idx": "30", "entity_id": "1499", "entity_type": "Gene", "text_name": "beta-catenin" } ]
[ { "begin_idx": "387", "end_idx": "407", "entity_id": "D003110", "entity_type": "Disease", "text_name": "colon carcinogenesis" }, { "begin_idx": "1498", "end_idx": "1515", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast carcinomas" } ]
[ "beta-catenin", "beta-catenin" ]
[ "colon carcinogenesis", "breast carcinomas" ]
9529363
Mutation and polymorphism analysis of the human homogentisate 1, 2-dioxygenase gene in alkaptonuria patients.
Alkaptonuria (AKU), a rare hereditary disorder of phenylalanine and tyrosine catabolism, was the first disease to be interpreted as an inborn error of metabolism. AKU patients are deficient for homogentisate 1,2 dioxygenase (HGO); this deficiency causes homogentisic aciduria, ochronosis, and arthritis. We cloned the human HGO gene and characterized two loss-of-function mutations, P230S and V300G, in the HGO gene in AKU patients. Here we report haplotype and mutational analysis of the HGO gene in 29 novel AKU chromosomes. We identified 12 novel mutations: 8 (E42A, W97G, D153G, S189I, I216T, R225H, F227S, and M368V) missense mutations that result in amino acid substitutions at positions conserved in HGO in different species, 1 (F10fs) frameshift mutation, 2 intronic mutations (IVS9-56G-->A, IVS9-17G-->A), and 1 splice-site mutation (IVS5+1G-->T). We also report characterization of five polymorphic sites in HGO and describe the haplotypic associations of alleles at these sites in normal and AKU chromosomes. One of these sites, HGO-3, is a variable dinucleotide repeat; IVS2+35T/A, IVS5+25T/C, and IVS6+46C/A are intronic sites at which single nucleotide substitutions (dimorphisms) have been detected; and c407T/A is a relatively frequent nucleotide substitution in the coding sequence, exon 4, resulting in an amino acid change (H80Q). These data provide insight into the origin and evolution of the various AKU alleles.
[ { "begin_idx": "377", "end_idx": "385", "entity_id": "C537358", "entity_type": "Disease", "text_name": "aciduria" }, { "begin_idx": "87", "end_idx": "99", "entity_id": "D000474", "entity_type": "Disease", "text_name": "alkaptonuria" }, { "begin_idx": "110", "end_idx": "122", "entity_id": "D000474", "entity_type": "Disease", "text_name": "Alkaptonuria" }, { "begin_idx": "124", "end_idx": "127", "entity_id": "D000474", "entity_type": "Disease", "text_name": "AKU" }, { "begin_idx": "273", "end_idx": "276", "entity_id": "D000474", "entity_type": "Disease", "text_name": "AKU" }, { "begin_idx": "529", "end_idx": "532", "entity_id": "D000474", "entity_type": "Disease", "text_name": "AKU" }, { "begin_idx": "620", "end_idx": "623", "entity_id": "D000474", "entity_type": "Disease", "text_name": "AKU" }, { "begin_idx": "1113", "end_idx": "1116", "entity_id": "D000474", "entity_type": "Disease", "text_name": "AKU" }, { "begin_idx": "1532", "end_idx": "1535", "entity_id": "D000474", "entity_type": "Disease", "text_name": "AKU" }, { "begin_idx": "403", "end_idx": "412", "entity_id": "D001168", "entity_type": "Disease", "text_name": "arthritis" }, { "begin_idx": "245", "end_idx": "271", "entity_id": "D008661", "entity_type": "Disease", "text_name": "inborn error of metabolism" }, { "begin_idx": "387", "end_idx": "397", "entity_id": "D009794", "entity_type": "Disease", "text_name": "ochronosis" }, { "begin_idx": "137", "end_idx": "197", "entity_id": "D030342", "entity_type": "Disease", "text_name": "hereditary disorder of phenylalanine and tyrosine catabolism" }, { "begin_idx": "48", "end_idx": "78", "entity_id": "3081", "entity_type": "Gene", "text_name": "homogentisate 1, 2-dioxygenase" }, { "begin_idx": "304", "end_idx": "333", "entity_id": "3081", "entity_type": "Gene", "text_name": "homogentisate 1,2 dioxygenase" }, { "begin_idx": "335", "end_idx": "338", "entity_id": "3081", "entity_type": "Gene", "text_name": "HGO" }, { "begin_idx": "434", "end_idx": "437", "entity_id": "3081", "entity_type": "Gene", "text_name": "HGO" }, { "begin_idx": "517", "end_idx": "520", "entity_id": "3081", "entity_type": "Gene", "text_name": "HGO" }, { "begin_idx": "599", "end_idx": "602", "entity_id": "3081", "entity_type": "Gene", "text_name": "HGO" }, { "begin_idx": "817", "end_idx": "820", "entity_id": "3081", "entity_type": "Gene", "text_name": "HGO" }, { "begin_idx": "1028", "end_idx": "1031", "entity_id": "3081", "entity_type": "Gene", "text_name": "HGO" }, { "begin_idx": "1150", "end_idx": "1153", "entity_id": "3081", "entity_type": "Gene", "text_name": "HGO" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "48", "end_idx": "78", "entity_id": "3081", "entity_type": "Gene", "text_name": "homogentisate 1, 2-dioxygenase" }, { "begin_idx": "1150", "end_idx": "1153", "entity_id": "3081", "entity_type": "Gene", "text_name": "HGO" } ]
[ { "begin_idx": "87", "end_idx": "99", "entity_id": "D000474", "entity_type": "Disease", "text_name": "alkaptonuria" }, { "begin_idx": "137", "end_idx": "197", "entity_id": "D030342", "entity_type": "Disease", "text_name": "hereditary disorder of phenylalanine and tyrosine catabolism" } ]
[ "homogentisate 1, 2-dioxygenase", "HGO" ]
[ "alkaptonuria", "hereditary disorder of phenylalanine and tyrosine catabolism" ]
9529364
Identification of constitutional WT1 mutations, in patients with isolated diffuse mesangial sclerosis, and analysis of genotype/phenotype correlations by use of a computerized mutation database.
Constitutional mutations of the WT1 gene, encoding a zinc-finger transcription factor involved in renal and gonadal development, are found in most patients with Denys-Drash syndrome (DDS), or diffuse mesangial sclerosis (DMS) associated with pseudohermaphroditism and/or Wilms tumor (WT). Most mutations in DDS patients lie in exon 8 or exon 9, encoding zinc finger 2 or zinc finger 3, respectively, with a hot spot (R394W) in exon 9. We analyzed a series of 24 patients, 10 with isolated DMS (IDMS), 10 with DDS, and 4 with urogenital abnormalities and/or WT. We report WT1 heterozygous mutations in 16 patients, 4 of whom presented with IDMS. One male and two female IDMS patients with WT1 mutations underwent normal puberty. Two mutations associated with IDMS are different from those described in DDS patients. No WT1 mutations were detected in the six other IDMS patients, suggesting genetic heterogeneity of this disease. We analyzed genotype/phenotype correlations, on the basis of the constitution of a WT1 mutation database of 84 germ-line mutations, to compare the distribution and type of mutations, according to the different symptoms. This demonstrated (1) the association between mutations in exons 8 and 9 and DMS; (2) among patients with DMS, a higher frequency of exon 8 mutations among 46, XY patients with female phenotype than among 46,XY patients with sexual ambiguity or male phenotype; and (3) statistically significant evidence that mutations in exons 8 and 9 preferentially affect amino acids with different functions.
[ { "begin_idx": "74", "end_idx": "101", "entity_id": "C537346", "entity_type": "Disease", "text_name": "diffuse mesangial sclerosis" }, { "begin_idx": "387", "end_idx": "414", "entity_id": "C537346", "entity_type": "Disease", "text_name": "diffuse mesangial sclerosis" }, { "begin_idx": "416", "end_idx": "419", "entity_id": "C537346", "entity_type": "Disease", "text_name": "DMS" }, { "begin_idx": "675", "end_idx": "687", "entity_id": "C537346", "entity_type": "Disease", "text_name": "isolated DMS" }, { "begin_idx": "689", "end_idx": "693", "entity_id": "C537346", "entity_type": "Disease", "text_name": "IDMS" }, { "begin_idx": "834", "end_idx": "838", "entity_id": "C537346", "entity_type": "Disease", "text_name": "IDMS" }, { "begin_idx": "864", "end_idx": "868", "entity_id": "C537346", "entity_type": "Disease", "text_name": "IDMS" }, { "begin_idx": "953", "end_idx": "957", "entity_id": "C537346", "entity_type": "Disease", "text_name": "IDMS" }, { "begin_idx": "1058", "end_idx": "1062", "entity_id": "C537346", "entity_type": "Disease", "text_name": "IDMS" }, { "begin_idx": "1420", "end_idx": "1423", "entity_id": "C537346", "entity_type": "Disease", "text_name": "DMS" }, { "begin_idx": "1449", "end_idx": "1452", "entity_id": "C537346", "entity_type": "Disease", "text_name": "DMS" }, { "begin_idx": "466", "end_idx": "477", "entity_id": "D009396", "entity_type": "Disease", "text_name": "Wilms tumor" }, { "begin_idx": "479", "end_idx": "481", "entity_id": "D009396", "entity_type": "Disease", "text_name": "WT" }, { "begin_idx": "752", "end_idx": "754", "entity_id": "D009396", "entity_type": "Disease", "text_name": "WT" }, { "begin_idx": "437", "end_idx": "458", "entity_id": "D012734", "entity_type": "Disease", "text_name": "pseudohermaphroditism" }, { "begin_idx": "720", "end_idx": "744", "entity_id": "D014564", "entity_type": "Disease", "text_name": "urogenital abnormalities" }, { "begin_idx": "356", "end_idx": "376", "entity_id": "D030321", "entity_type": "Disease", "text_name": "Denys-Drash syndrome" }, { "begin_idx": "378", "end_idx": "381", "entity_id": "D030321", "entity_type": "Disease", "text_name": "DDS" }, { "begin_idx": "502", "end_idx": "505", "entity_id": "D030321", "entity_type": "Disease", "text_name": "DDS" }, { "begin_idx": "704", "end_idx": "707", "entity_id": "D030321", "entity_type": "Disease", "text_name": "DDS" }, { "begin_idx": "996", "end_idx": "999", "entity_id": "D030321", "entity_type": "Disease", "text_name": "DDS" }, { "begin_idx": "33", "end_idx": "36", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" }, { "begin_idx": "227", "end_idx": "230", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" }, { "begin_idx": "766", "end_idx": "769", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" }, { "begin_idx": "883", "end_idx": "886", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" }, { "begin_idx": "1013", "end_idx": "1016", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" }, { "begin_idx": "1206", "end_idx": "1209", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" } ]
[ "Yes", "Yes", "No", "No" ]
[ true, true, true, true ]
[ { "begin_idx": "33", "end_idx": "36", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" }, { "begin_idx": "33", "end_idx": "36", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" }, { "begin_idx": "766", "end_idx": "769", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" }, { "begin_idx": "883", "end_idx": "886", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" } ]
[ { "begin_idx": "356", "end_idx": "376", "entity_id": "D030321", "entity_type": "Disease", "text_name": "Denys-Drash syndrome" }, { "begin_idx": "466", "end_idx": "477", "entity_id": "D009396", "entity_type": "Disease", "text_name": "Wilms tumor" }, { "begin_idx": "437", "end_idx": "458", "entity_id": "D012734", "entity_type": "Disease", "text_name": "pseudohermaphroditism" }, { "begin_idx": "416", "end_idx": "419", "entity_id": "C537346", "entity_type": "Disease", "text_name": "DMS" } ]
[ "WT1", "WT1", "WT1", "WT1" ]
[ "Denys-Drash syndrome", "Wilms tumor", "pseudohermaphroditism", "DMS" ]
9535125
Association study of a functional catechol-O-methyltransferase gene polymorphism in Japanese schizophrenics.
Catechol-O-methyltransferase (COMT) is an enzyme which inactivates catecholamine neurotransmitters by methylation, and is considered a candidate for involvement in schizophrenia. A functional COMT gene polymorphism influencing the enzyme activities, the high activity (val-108) and the low activity allele (met-108), was recently confirmed. We investigated a genetic association between schizophrenia and the COMT gene polymorphism in 150 Japanese schizophrenics and controls. We detected the low activity met-108 allele more frequently in schizophrenics than in the controls, and found that subjects sharing the met-108 allele (val/met and met/met) are significantly more common in the patients than in the controls. The results suggest that the low activity met-108 allele may be involved in susceptibility for schizophrenia.
[ { "begin_idx": "93", "end_idx": "107", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenics" }, { "begin_idx": "273", "end_idx": "286", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "496", "end_idx": "509", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "557", "end_idx": "571", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenics" }, { "begin_idx": "649", "end_idx": "663", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenics" }, { "begin_idx": "922", "end_idx": "935", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "34", "end_idx": "62", "entity_id": "1312", "entity_type": "Gene", "text_name": "catechol-O-methyltransferase" }, { "begin_idx": "109", "end_idx": "137", "entity_id": "1312", "entity_type": "Gene", "text_name": "Catechol-O-methyltransferase" }, { "begin_idx": "139", "end_idx": "143", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "301", "end_idx": "305", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" }, { "begin_idx": "518", "end_idx": "522", "entity_id": "1312", "entity_type": "Gene", "text_name": "COMT" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "34", "end_idx": "62", "entity_id": "1312", "entity_type": "Gene", "text_name": "catechol-O-methyltransferase" } ]
[ { "begin_idx": "93", "end_idx": "107", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenics" } ]
[ "catechol-O-methyltransferase" ]
[ "schizophrenics" ]
9536091
Identification of a mutation in liver glycogen phosphorylase in glycogen storage disease type VI.
Glycogen storage disease type VI (GSD6) defines a group of disorders that cause hepatomegaly and hypoglycemia with reduced liver phosphorylase activity. The course of these disorders is generally mild, but definitive diagnosis requires invasive procedures. We analyzed a Mennonite kindred with an autosomal recessive form of GSD6 to determine the molecular defect and develop a non-invasive diagnostic test. Linkage analysis was performed using genetic markers flanking the liver glycogen phosphorylase gene ( PYGL ), which was suspected to be the cause of the disorder on biochemical grounds. Mennonite GSD6 was linked to the PYGL locus with a multipoint LOD score of 4.7. The PYGL gene was analyzed for mutations by sequencing genomic DNA. Sequencing of genomic DNA revealed a splice site abnormality of the intron 13 splice donor. Confirmation of the genomic mutation was performed by sequencing RT-PCR products, which showed heterogeneous PYGL mRNA lacking all or part of exon 13 in affected persons. This study is the first to demonstrate that a mutation in the PYGL gene can cause GSD6. This mutation is estimated to be present on 3% of Mennonite chromosomes and the disease affects 0.1% of that population. Determination of this mutation provides a basis for the development of a simple and non-invasive diagnostic test for the disease and the carrier state in this population and confirms biochemical data showing the importance of this gene in glucose homeostasis.
[ { "begin_idx": "98", "end_idx": "130", "entity_id": "D006013", "entity_type": "Disease", "text_name": "Glycogen storage disease type VI" }, { "begin_idx": "178", "end_idx": "190", "entity_id": "D006529", "entity_type": "Disease", "text_name": "hepatomegaly" }, { "begin_idx": "195", "end_idx": "207", "entity_id": "D007003", "entity_type": "Disease", "text_name": "hypoglycemia" }, { "begin_idx": "89", "end_idx": "96", "entity_id": "D020754", "entity_type": "Disease", "text_name": "type VI" }, { "begin_idx": "132", "end_idx": "136", "entity_id": "5836", "entity_type": "Gene", "text_name": "GSD6" }, { "begin_idx": "423", "end_idx": "427", "entity_id": "5836", "entity_type": "Gene", "text_name": "GSD6" }, { "begin_idx": "608", "end_idx": "612", "entity_id": "5836", "entity_type": "Gene", "text_name": "PYGL" }, { "begin_idx": "702", "end_idx": "706", "entity_id": "5836", "entity_type": "Gene", "text_name": "GSD6" }, { "begin_idx": "725", "end_idx": "729", "entity_id": "5836", "entity_type": "Gene", "text_name": "PYGL" }, { "begin_idx": "776", "end_idx": "780", "entity_id": "5836", "entity_type": "Gene", "text_name": "PYGL" }, { "begin_idx": "1041", "end_idx": "1045", "entity_id": "5836", "entity_type": "Gene", "text_name": "PYGL" }, { "begin_idx": "1165", "end_idx": "1169", "entity_id": "5836", "entity_type": "Gene", "text_name": "PYGL" }, { "begin_idx": "1185", "end_idx": "1189", "entity_id": "5836", "entity_type": "Gene", "text_name": "GSD6" } ]
[ "Yes", "No" ]
[ false, false ]
[ { "begin_idx": "132", "end_idx": "136", "entity_id": "5836", "entity_type": "Gene", "text_name": "GSD6" }, { "begin_idx": "608", "end_idx": "612", "entity_id": "5836", "entity_type": "Gene", "text_name": "PYGL" } ]
[ { "begin_idx": "98", "end_idx": "130", "entity_id": "D006013", "entity_type": "Disease", "text_name": "Glycogen storage disease type VI" }, { "begin_idx": "89", "end_idx": "96", "entity_id": "D020754", "entity_type": "Disease", "text_name": "type VI" } ]
[ "GSD6", "PYGL" ]
[ "Glycogen storage disease type VI", "type VI" ]
9537410
De novo mutations in the CRX homeobox gene associated with Leber congenital amaurosis.
[ { "begin_idx": "59", "end_idx": "85", "entity_id": "D057130", "entity_type": "Disease", "text_name": "Leber congenital amaurosis" }, { "begin_idx": "25", "end_idx": "28", "entity_id": "1406", "entity_type": "Gene", "text_name": "CRX" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "25", "end_idx": "28", "entity_id": "1406", "entity_type": "Gene", "text_name": "CRX" } ]
[ { "begin_idx": "59", "end_idx": "85", "entity_id": "D057130", "entity_type": "Disease", "text_name": "Leber congenital amaurosis" } ]
[ "CRX" ]
[ "Leber congenital amaurosis" ]
9540028
Angiotensinogen T235 and ACE insertion/deletion polymorphisms associated with albuminuria in Chinese type 2 diabetic patients.
OBJECTIVE: Genetic polymorphisms of the renin-angiotensin system (RAS) have been implicated in the pathogenesis of diabetic proteinuria. Ethnic differences in the frequencies of these genotypes have also been reported. To date, most of these studies have been performed in white and Japanese populations. In this study, we examined the associations between albuminuria and RAS genetic polymorphisms in Chinese patients with type 2 diabetes. RESEARCH DESIGN AND METHODS: In a case-control study, the ACE insertion/deletion (I/D) gene, the angiotensinogen (AGT) gene (M235T), and the angiotensin II (AII) type 1 receptor gene (AT1 A1166C) were examined in 110 Chinese type 2 diabetic patients. Increased urinary albumin excretion (UAE) was defined as > or = 30 mg/day on at least two occasions during a 6-week study period. RESULTS: Compared with whites, there were high frequencies of the AGT TT genotype in Chinese control subjects (120/183 = 70%) and type 2 diabetic patients (74/110 = 67%). The frequencies of the MM genotype were 5 and 3%, respectively, and those of the ACE DD genotype were 13 and 10%, respectively. Although 9% of subjects carried the C allele, the AT1 CC genotype was not found in either group. Chinese type 2 diabetic patients with increased albuminuria (n = 56) had higher systolic blood pressure (160 +/- 26 mmHg vs 145 +/- 27 mmHg, P < 0.001) than the normoalbuminuric patients (n = 54). Both the AGT TT genotype (78.6% [44/56] vs. 55.6% [30/54], odds ratio [OR]: 3.0 [1.3-6.8]) and the T allele (88% [99/112] vs. 77% [83/108], OR: 2.5 [1.3-5.4]) were associated with an increased risk of albuminuria. Patients with the AGT TT genotype (n = 74) had higher 24-h UAE than those with the MT or MM genotypes (n = 36) (median: 37.8 mg/day vs. 17.8 mg/day, P < 0.01). This association remained significant in patients with normotension (56 mg/day [n = 19] for patients with the TT genotype vs. 22 mg/day [n = 14] for those with the MT/MM genotype, P = 0.03). The D allele carriers (DD or DI, n = 61) had higher serum ACE activities (75.5 +/- 29 U/l vs. 60.5 +/- 36.3 U/l, P < 0.01) than the noncarriers (II genotype). The median 24-h UAE also tended to be higher in the D allele carriers (38.9 mg/day vs. 21.4 mg/day, P = 0.07). The lowest UAE was observed in patients with the MM/MT/II genotype (16.3 mg/day [n = 18]) and the highest, in patients with the TT/DD/DI genotype (52.3 mg/day [n = 43]). No association was found between the TT genotype or D allele and hypertension. CONCLUSIONS: The high frequencies of the TT genotype and T allele in Chinese populations may contribute to the high prevalence of albuminuria in patients with type 2 diabetes. The possibility of synergism between the AGT TT genotype and the ACE D allele should also be explored.
[ { "begin_idx": "1205", "end_idx": "1207", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "2130", "end_idx": "2132", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "2508", "end_idx": "2510", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "2136", "end_idx": "2138", "entity_id": "C564703", "entity_type": "Disease", "text_name": "DI" }, { "begin_idx": "2511", "end_idx": "2513", "entity_id": "C564703", "entity_type": "Disease", "text_name": "DI" }, { "begin_idx": "78", "end_idx": "89", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "484", "end_idx": "495", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "1393", "end_idx": "1404", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "1743", "end_idx": "1754", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "2756", "end_idx": "2767", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "108", "end_idx": "116", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "242", "end_idx": "250", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "558", "end_idx": "566", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetes" }, { "begin_idx": "800", "end_idx": "808", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "1086", "end_idx": "1094", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "1360", "end_idx": "1368", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "2792", "end_idx": "2800", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetes" }, { "begin_idx": "2612", "end_idx": "2624", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "251", "end_idx": "262", "entity_id": "D011507", "entity_type": "Disease", "text_name": "proteinuria" }, { "begin_idx": "101", "end_idx": "105", "entity_id": "D017827", "entity_type": "Disease", "text_name": "type" }, { "begin_idx": "551", "end_idx": "555", "entity_id": "D017827", "entity_type": "Disease", "text_name": "type" }, { "begin_idx": "793", "end_idx": "797", "entity_id": "D017827", "entity_type": "Disease", "text_name": "type" }, { "begin_idx": "1079", "end_idx": "1083", "entity_id": "D017827", "entity_type": "Disease", "text_name": "type" }, { "begin_idx": "1353", "end_idx": "1357", "entity_id": "D017827", "entity_type": "Disease", "text_name": "type" }, { "begin_idx": "2785", "end_idx": "2789", "entity_id": "D017827", "entity_type": "Disease", "text_name": "type" }, { "begin_idx": "25", "end_idx": "28", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "626", "end_idx": "629", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "1201", "end_idx": "1204", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "2165", "end_idx": "2168", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "2867", "end_idx": "2870", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "0", "end_idx": "15", "entity_id": "183", "entity_type": "Gene", "text_name": "Angiotensinogen" }, { "begin_idx": "665", "end_idx": "680", "entity_id": "183", "entity_type": "Gene", "text_name": "angiotensinogen" }, { "begin_idx": "682", "end_idx": "685", "entity_id": "183", "entity_type": "Gene", "text_name": "AGT" }, { "begin_idx": "1015", "end_idx": "1018", "entity_id": "183", "entity_type": "Gene", "text_name": "AGT" }, { "begin_idx": "1551", "end_idx": "1554", "entity_id": "183", "entity_type": "Gene", "text_name": "AGT" }, { "begin_idx": "1774", "end_idx": "1777", "entity_id": "183", "entity_type": "Gene", "text_name": "AGT" }, { "begin_idx": "2843", "end_idx": "2846", "entity_id": "183", "entity_type": "Gene", "text_name": "AGT" }, { "begin_idx": "709", "end_idx": "745", "entity_id": "185", "entity_type": "Gene", "text_name": "angiotensin II (AII) type 1 receptor" }, { "begin_idx": "752", "end_idx": "755", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" }, { "begin_idx": "1298", "end_idx": "1301", "entity_id": "185", "entity_type": "Gene", "text_name": "AT1" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "0", "end_idx": "15", "entity_id": "183", "entity_type": "Gene", "text_name": "Angiotensinogen" }, { "begin_idx": "626", "end_idx": "629", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" } ]
[ { "begin_idx": "78", "end_idx": "89", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "484", "end_idx": "495", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" } ]
[ "Angiotensinogen", "ACE" ]
[ "albuminuria", "albuminuria" ]
9541180
Endothelial nitric oxide synthase gene polymorphism and coronary heart disease in Japanese NIDDM.
[ { "begin_idx": "56", "end_idx": "78", "entity_id": "D003327", "entity_type": "Disease", "text_name": "coronary heart disease" }, { "begin_idx": "91", "end_idx": "96", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "0", "end_idx": "33", "entity_id": "4846", "entity_type": "Gene", "text_name": "Endothelial nitric oxide synthase" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "0", "end_idx": "33", "entity_id": "4846", "entity_type": "Gene", "text_name": "Endothelial nitric oxide synthase" }, { "begin_idx": "0", "end_idx": "33", "entity_id": "4846", "entity_type": "Gene", "text_name": "Endothelial nitric oxide synthase" } ]
[ { "begin_idx": "91", "end_idx": "96", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "56", "end_idx": "78", "entity_id": "D003327", "entity_type": "Disease", "text_name": "coronary heart disease" } ]
[ "Endothelial nitric oxide synthase", "Endothelial nitric oxide synthase" ]
[ "NIDDM", "coronary heart disease" ]
9544854
Relationship of the angiotensin-converting enzyme gene polymorphism to glucose intolerance, insulin resistance, and hypertension in NIDDM.
The deletion (D) allele of the angiotensin-converting enzyme (ACE) gene insertion/deletion (I/D) polymorphism has been shown to be associated with cardiovascular and renal diseases in diabetes mellitus, but the mechanism underlying this association is not known. In addition, recent studies of the effect of the ACE gene on blood pressure have yielded conflicting results. Therefore, we studied the association of the ACE gene I/D polymorphism with glucose intolerance and insulin resistance, and the contribution of this locus to genetic susceptibility to hypertension in non-insulin-dependent diabetic mellitus (NIDDM). We analysed the ACE genotype in 84 unrelated NIDDM patients with a known disease duration of less than 1 year and in 115 age- and sex-matched controls. The I/D polymorphism was determined by the polymerase chain reaction. There were no differences in ACE genotype distribution and allele frequencies between patients with NIDDM and nondiabetic controls. The frequencies of the D and I alleles in both groups were identical, viz., 0.65 and 0.35, respectively. The NIDDM patients with the DD genotype had significantly higher blood glucose levels in the oral glucose tolerance test than those with the other genotypes; the incremental glucose area under the curve in the order of II, ID, and DD was 7.2+/-2.4, 9.2+/-4.0, and 10.7+/-2.7 mmol/l x h (II vs ID vs DD, P=0.0066 by ANOVA). No significant difference was found between the ACE genotype and serum insulin values. Similarly, there were no differences in body mass index, blood pressure, or serum lipids between the three genotypes. Among the non-diabetic controls, there was no statistically significant association of the I/D polymorphism with serum lipids, blood glucose levels, serum insulin concentrations, or blood pressure values. In conclusion, NIDDM patients with the DD genotype have higher blood glucose levels and are more glucose intolerant; this may help to explain the reported association between the D allele and vascular complications in NIDDM.
[ { "begin_idx": "1248", "end_idx": "1250", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "1451", "end_idx": "1453", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "1519", "end_idx": "1521", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "1992", "end_idx": "1994", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "323", "end_idx": "340", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetes mellitus" }, { "begin_idx": "132", "end_idx": "137", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "712", "end_idx": "751", "entity_id": "D003924", "entity_type": "Disease", "text_name": "non-insulin-dependent diabetic mellitus" }, { "begin_idx": "753", "end_idx": "758", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "806", "end_idx": "811", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1083", "end_idx": "1088", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1224", "end_idx": "1229", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1968", "end_idx": "1973", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "2171", "end_idx": "2176", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "116", "end_idx": "128", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "696", "end_idx": "708", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "305", "end_idx": "319", "entity_id": "D007674", "entity_type": "Disease", "text_name": "renal diseases" }, { "begin_idx": "2145", "end_idx": "2167", "entity_id": "D014652", "entity_type": "Disease", "text_name": "vascular complications" }, { "begin_idx": "71", "end_idx": "90", "entity_id": "D018149", "entity_type": "Disease", "text_name": "glucose intolerance" }, { "begin_idx": "588", "end_idx": "607", "entity_id": "D018149", "entity_type": "Disease", "text_name": "glucose intolerance" }, { "begin_idx": "1382", "end_idx": "1406", "entity_id": "D018149", "entity_type": "Disease", "text_name": "incremental glucose area" }, { "begin_idx": "20", "end_idx": "49", "entity_id": "1636", "entity_type": "Gene", "text_name": "angiotensin-converting enzyme" }, { "begin_idx": "170", "end_idx": "199", "entity_id": "1636", "entity_type": "Gene", "text_name": "angiotensin-converting enzyme" }, { "begin_idx": "201", "end_idx": "204", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "451", "end_idx": "454", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "557", "end_idx": "560", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "777", "end_idx": "780", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "1012", "end_idx": "1015", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "1591", "end_idx": "1594", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "92", "end_idx": "99", "entity_id": "3630", "entity_type": "Gene", "text_name": "insulin" }, { "begin_idx": "612", "end_idx": "619", "entity_id": "3630", "entity_type": "Gene", "text_name": "insulin" }, { "begin_idx": "1614", "end_idx": "1621", "entity_id": "3630", "entity_type": "Gene", "text_name": "insulin" }, { "begin_idx": "1903", "end_idx": "1910", "entity_id": "3630", "entity_type": "Gene", "text_name": "insulin" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "20", "end_idx": "49", "entity_id": "1636", "entity_type": "Gene", "text_name": "angiotensin-converting enzyme" }, { "begin_idx": "612", "end_idx": "619", "entity_id": "3630", "entity_type": "Gene", "text_name": "insulin" } ]
[ { "begin_idx": "712", "end_idx": "751", "entity_id": "D003924", "entity_type": "Disease", "text_name": "non-insulin-dependent diabetic mellitus" }, { "begin_idx": "1083", "end_idx": "1088", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" } ]
[ "angiotensin-converting enzyme", "insulin" ]
[ "non-insulin-dependent diabetic mellitus", "NIDDM" ]
9550358
The Asn9 variant of lipoprotein lipase is associated with the -93G promoter mutation and an increased risk of coronary artery disease. The Regress Study Group.
Two mutations in the lipoprotein lipase (LPL) gene, a T to G transition at position -93 of the proximal promoter region and an Asp9Asn substitution in exon 2, were examined in 762 Dutch males with angiographically-diagnosed coronary artery disease (CAD) and 296 healthy normolipidemic Dutch males. The two mutations exhibited strong linkage disequilibrium (D' = 0.975). A significantly higher proportion of cases (4.86%) than controls (1.37%) carried the -93G/Asn9 allele (p = 0.008). In the combined sample of cases and controls, adjusted mean plasma total cholesterol (TC) levels were significantly higher in -93G/Asn9 carriers (6.20+/-0.13 mmol/l) than in non-carriers (5.93+/-0.03 mmol/l; p = 0.048), while mean high-density lipoprotein cholesterol (HDL-C) levels were lower in carriers (0.88+/-0.03 mmol/l) than in non-carriers (0.98+/-0.01 mmol/l; p = 0.002). There was a trend towards higher triglyceride (TG) levels in carriers (1.96+/-0.14 mmol/l) compared with non-carriers (1.73+/-0.03 mmol/l) (p = 0.08). Additionally, carrier frequencies in tertiles of TC, HDL-C, TG, and LPL activity, suggested an association of the -93G/Asn9 variant with higher TC and TG levels, and with lower HDL-C and LPL activity levels. Logistic regression revealed a significant odds ratio (OR) for the combined -93G/Asn9 genotype in CAD cases relative to controls (OR: 5.36; 95% CI: 1.57-18.24), with age, body mass index (BMI), smoking, and plasma total- and HDL-cholesterol levels included in the model. In conclusion, we show that the LPL Asp9Asn mutation is in non-random association with a T G substitution at position -93 of the proximal promoter region and that the combined -93G/Asn9 genotype predisposes to decreased HDL-C levels and an increased risk of CAD.
[ { "begin_idx": "1059", "end_idx": "1071", "entity_id": "C566031", "entity_type": "Disease", "text_name": "triglyceride" }, { "begin_idx": "1073", "end_idx": "1075", "entity_id": "C566031", "entity_type": "Disease", "text_name": "TG" }, { "begin_idx": "1237", "end_idx": "1239", "entity_id": "C566031", "entity_type": "Disease", "text_name": "TG" }, { "begin_idx": "1328", "end_idx": "1330", "entity_id": "C566031", "entity_type": "Disease", "text_name": "TG" }, { "begin_idx": "110", "end_idx": "133", "entity_id": "D003324", "entity_type": "Disease", "text_name": "coronary artery disease" }, { "begin_idx": "384", "end_idx": "407", "entity_id": "D003324", "entity_type": "Disease", "text_name": "coronary artery disease" }, { "begin_idx": "409", "end_idx": "412", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "1483", "end_idx": "1486", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "1914", "end_idx": "1917", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "20", "end_idx": "38", "entity_id": "4023", "entity_type": "Gene", "text_name": "lipoprotein lipase" }, { "begin_idx": "181", "end_idx": "199", "entity_id": "4023", "entity_type": "Gene", "text_name": "lipoprotein lipase" }, { "begin_idx": "201", "end_idx": "204", "entity_id": "4023", "entity_type": "Gene", "text_name": "LPL" }, { "begin_idx": "1245", "end_idx": "1248", "entity_id": "4023", "entity_type": "Gene", "text_name": "LPL" }, { "begin_idx": "1364", "end_idx": "1367", "entity_id": "4023", "entity_type": "Gene", "text_name": "LPL" }, { "begin_idx": "1688", "end_idx": "1691", "entity_id": "4023", "entity_type": "Gene", "text_name": "LPL" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "20", "end_idx": "38", "entity_id": "4023", "entity_type": "Gene", "text_name": "lipoprotein lipase" }, { "begin_idx": "181", "end_idx": "199", "entity_id": "4023", "entity_type": "Gene", "text_name": "lipoprotein lipase" } ]
[ { "begin_idx": "110", "end_idx": "133", "entity_id": "D003324", "entity_type": "Disease", "text_name": "coronary artery disease" }, { "begin_idx": "1073", "end_idx": "1075", "entity_id": "C566031", "entity_type": "Disease", "text_name": "TG" } ]
[ "lipoprotein lipase", "lipoprotein lipase" ]
[ "coronary artery disease", "TG" ]
9550362
Severe cystic fibrosis associated with a deltaF508/R347H + D979A compound heterozygous genotype.
This report is concerned with twins with cystic fibrosis (CF). They are of mixed parentage: Japanese mother and German father. One case is presented with meconium ileus as a neonate. The other patient did relatively well until the age of 6 years when she was first hospitalized and diagnosed with pulmonary aspergillosis. They have been receiving standard therapies for CF including digestive enzymes, vitamins and periodic antibiotic therapy in the US. At 19 years of age, they were tested for common mutations and one AF508 cystic fibrosis transmembrane conductance regulator (CFTR) allele was found. Further testing of their CFTR gene as well as those of their Japanese mother and grandmother revealed missense mutations in exon 7 (R347H) and exon 16 (D979A). Although the D979A mutant is very rare, this mutation combination seemed to be responsible for severe CF phenotypes.
[ { "begin_idx": "7", "end_idx": "22", "entity_id": "D003550", "entity_type": "Disease", "text_name": "cystic fibrosis" }, { "begin_idx": "138", "end_idx": "153", "entity_id": "D003550", "entity_type": "Disease", "text_name": "cystic fibrosis" }, { "begin_idx": "155", "end_idx": "157", "entity_id": "D003550", "entity_type": "Disease", "text_name": "CF" }, { "begin_idx": "467", "end_idx": "469", "entity_id": "D003550", "entity_type": "Disease", "text_name": "CF" }, { "begin_idx": "623", "end_idx": "638", "entity_id": "D003550", "entity_type": "Disease", "text_name": "cystic fibrosis" }, { "begin_idx": "962", "end_idx": "964", "entity_id": "D003550", "entity_type": "Disease", "text_name": "CF" }, { "begin_idx": "394", "end_idx": "417", "entity_id": "D055732", "entity_type": "Disease", "text_name": "pulmonary aspergillosis" }, { "begin_idx": "676", "end_idx": "680", "entity_id": "1080", "entity_type": "Gene", "text_name": "CFTR" }, { "begin_idx": "725", "end_idx": "729", "entity_id": "1080", "entity_type": "Gene", "text_name": "CFTR" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "676", "end_idx": "680", "entity_id": "1080", "entity_type": "Gene", "text_name": "CFTR" }, { "begin_idx": "725", "end_idx": "729", "entity_id": "1080", "entity_type": "Gene", "text_name": "CFTR" } ]
[ { "begin_idx": "7", "end_idx": "22", "entity_id": "D003550", "entity_type": "Disease", "text_name": "cystic fibrosis" }, { "begin_idx": "394", "end_idx": "417", "entity_id": "D055732", "entity_type": "Disease", "text_name": "pulmonary aspergillosis" } ]
[ "CFTR", "CFTR" ]
[ "cystic fibrosis", "pulmonary aspergillosis" ]
9554745
Mutation and expression analysis of the endoglin gene in hereditary hemorrhagic telangiectasia reveals null alleles.
Hereditary Hemorrhagic Telangiectasia (HHT) is an autosomal dominant disorder characterized by multisystemic vascular dysplasia and recurrent hemorrhage from the sites of vascular lesions. Two genes have been identified for HHT. Endoglin, a TGF-beta binding protein which maps to chromosome 9q3, is the gene for HHT1. The type and location of most of the previously described mutations in the endoglin (ENG) gene suggested a dominant-negative model of receptor-complex dysfunction for the molecular basis of this disorder. In this article we describe 11 novel ENG mutations in HHT kindreds, which include missense and splice-site mutations. Two identical missense mutations in unrelated families disrupt the start codon of the gene. In addition, some frameshift and nonsense mutations lead to very low or undetectable levels of transcript from the mutant allele. These combined data suggest that the nature of most ENG mutations is to create a null (nonfunctional) allele, and that there is no requirement for the synthesis of a truncated endoglin protein in the pathogenesis of HHT.
[ { "begin_idx": "259", "end_idx": "269", "entity_id": "D006470", "entity_type": "Disease", "text_name": "hemorrhage" }, { "begin_idx": "569", "end_idx": "597", "entity_id": "D008107", "entity_type": "Disease", "text_name": "receptor-complex dysfunction" }, { "begin_idx": "57", "end_idx": "94", "entity_id": "D013683", "entity_type": "Disease", "text_name": "hereditary hemorrhagic telangiectasia" }, { "begin_idx": "117", "end_idx": "154", "entity_id": "D013683", "entity_type": "Disease", "text_name": "Hereditary Hemorrhagic Telangiectasia" }, { "begin_idx": "156", "end_idx": "159", "entity_id": "D013683", "entity_type": "Disease", "text_name": "HHT" }, { "begin_idx": "341", "end_idx": "344", "entity_id": "D013683", "entity_type": "Disease", "text_name": "HHT" }, { "begin_idx": "694", "end_idx": "697", "entity_id": "D013683", "entity_type": "Disease", "text_name": "HHT" }, { "begin_idx": "1196", "end_idx": "1199", "entity_id": "D013683", "entity_type": "Disease", "text_name": "HHT" }, { "begin_idx": "212", "end_idx": "244", "entity_id": "D014652", "entity_type": "Disease", "text_name": "multisystemic vascular dysplasia" }, { "begin_idx": "288", "end_idx": "304", "entity_id": "D014652", "entity_type": "Disease", "text_name": "vascular lesions" }, { "begin_idx": "167", "end_idx": "194", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal dominant disorder" }, { "begin_idx": "40", "end_idx": "48", "entity_id": "2022", "entity_type": "Gene", "text_name": "endoglin" }, { "begin_idx": "346", "end_idx": "354", "entity_id": "2022", "entity_type": "Gene", "text_name": "Endoglin" }, { "begin_idx": "429", "end_idx": "433", "entity_id": "2022", "entity_type": "Gene", "text_name": "HHT1" }, { "begin_idx": "510", "end_idx": "518", "entity_id": "2022", "entity_type": "Gene", "text_name": "endoglin" }, { "begin_idx": "520", "end_idx": "523", "entity_id": "2022", "entity_type": "Gene", "text_name": "ENG" }, { "begin_idx": "677", "end_idx": "680", "entity_id": "2022", "entity_type": "Gene", "text_name": "ENG" }, { "begin_idx": "1032", "end_idx": "1035", "entity_id": "2022", "entity_type": "Gene", "text_name": "ENG" }, { "begin_idx": "1156", "end_idx": "1164", "entity_id": "2022", "entity_type": "Gene", "text_name": "endoglin" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "40", "end_idx": "48", "entity_id": "2022", "entity_type": "Gene", "text_name": "endoglin" }, { "begin_idx": "1032", "end_idx": "1035", "entity_id": "2022", "entity_type": "Gene", "text_name": "ENG" } ]
[ { "begin_idx": "57", "end_idx": "94", "entity_id": "D013683", "entity_type": "Disease", "text_name": "hereditary hemorrhagic telangiectasia" }, { "begin_idx": "212", "end_idx": "244", "entity_id": "D014652", "entity_type": "Disease", "text_name": "multisystemic vascular dysplasia" } ]
[ "endoglin", "ENG" ]
[ "hereditary hemorrhagic telangiectasia", "multisystemic vascular dysplasia" ]
9554749
Clustering of private mutations in the congenital chloride diarrhea/down-regulated in adenoma gene.
An inherited defect in intestinal anion exchange, congenital chloride diarrhea (CLD), was recently shown to be caused by mutations in the down-regulated in adenoma (DRA) gene. A three base pair deletion resulting in the loss of an amino acid valine (V317del) in the predicted CLD/DRA protein was shown to be responsible for all CLD cases in a Finnish founder population. Two additional mutations, H124L and 344delT, were found in Polish CLD patients. Here, we screened for additional mutations in a set of 14 CLD families of Polish, Swedish, North American, and Finnish origin using primers that allowed mutation searches directly from genomic DNA samples. We found eight novel mutations in the CLD/DRA gene. The mutations included two transversions, one transition, one insertion, and four small deletions. Of 11 sequence alterations detected so far, nine lie clustered in three short segments that are 49 bp, 39 bp, and 65 bp in size, respectively. These short segments span only 6.7% of the total cDNA length, suggesting functional importance or mutation-prone DNA regions of the corresponding CLD/DRA protein domains.
[ { "begin_idx": "39", "end_idx": "67", "entity_id": "C536210", "entity_type": "Disease", "text_name": "congenital chloride diarrhea" }, { "begin_idx": "150", "end_idx": "178", "entity_id": "C536210", "entity_type": "Disease", "text_name": "congenital chloride diarrhea" }, { "begin_idx": "180", "end_idx": "183", "entity_id": "C536210", "entity_type": "Disease", "text_name": "CLD" }, { "begin_idx": "376", "end_idx": "379", "entity_id": "C536210", "entity_type": "Disease", "text_name": "CLD" }, { "begin_idx": "428", "end_idx": "431", "entity_id": "C536210", "entity_type": "Disease", "text_name": "CLD" }, { "begin_idx": "537", "end_idx": "540", "entity_id": "C536210", "entity_type": "Disease", "text_name": "CLD" }, { "begin_idx": "609", "end_idx": "612", "entity_id": "C536210", "entity_type": "Disease", "text_name": "CLD" }, { "begin_idx": "795", "end_idx": "798", "entity_id": "C536210", "entity_type": "Disease", "text_name": "CLD" }, { "begin_idx": "1197", "end_idx": "1200", "entity_id": "C536210", "entity_type": "Disease", "text_name": "CLD" }, { "begin_idx": "86", "end_idx": "93", "entity_id": "D000236", "entity_type": "Disease", "text_name": "adenoma" }, { "begin_idx": "103", "end_idx": "119", "entity_id": "D030342", "entity_type": "Disease", "text_name": "inherited defect" }, { "begin_idx": "238", "end_idx": "269", "entity_id": "1811", "entity_type": "Gene", "text_name": "down-regulated in adenoma (DRA)" }, { "begin_idx": "380", "end_idx": "383", "entity_id": "1811", "entity_type": "Gene", "text_name": "DRA" }, { "begin_idx": "799", "end_idx": "802", "entity_id": "1811", "entity_type": "Gene", "text_name": "DRA" }, { "begin_idx": "1201", "end_idx": "1204", "entity_id": "1811", "entity_type": "Gene", "text_name": "DRA" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "238", "end_idx": "269", "entity_id": "1811", "entity_type": "Gene", "text_name": "down-regulated in adenoma (DRA)" }, { "begin_idx": "380", "end_idx": "383", "entity_id": "1811", "entity_type": "Gene", "text_name": "DRA" } ]
[ { "begin_idx": "39", "end_idx": "67", "entity_id": "C536210", "entity_type": "Disease", "text_name": "congenital chloride diarrhea" }, { "begin_idx": "103", "end_idx": "119", "entity_id": "D030342", "entity_type": "Disease", "text_name": "inherited defect" } ]
[ "down-regulated in adenoma (DRA)", "DRA" ]
[ "congenital chloride diarrhea", "inherited defect" ]
9563925
Treatment of chronic hepatitis C with interferon with or without ursodeoxycholic acid: a randomized prospective trial.
The only effective and approved therapy for chronic hepatitis C is interferon-alpha. Because sustained response rates with interferon alone are disappointingly low, multidrug treatment regimens are currently being investigated. Ursodeoxycholic acid has been used in other chronic liver diseases and can limit hepatocyte injury. To evaluate the potential benefit of ursodeoxycholic acid in combination with interferon-alpha for the treatment of chronic hepatitis C, we conducted a prospective, double-blinded, randomized, placebo-controlled trial comparing the combination therapy of interferon-alpha 2b and ursodeoxycholic acid with interferon alone. Thirty-one patients with chronic hepatitis C were randomized to receive 3 million units of interferon-alpha 2b subcutaneously three times per week and either 13 to 15 mg/kg/day ursodeoxycholic acid or placebo orally for 6 months. The 6-month treatment period was followed by 6 months of observation. Biochemical normalization at the end of treatment occurred in 5 of 14 (36%) patients receiving monotherapy versus 8 of 15 (53%) patients (p = 0.34) receiving combination therapy. No patient treated with interferon alone had a sustained biochemical response 6 months after therapy; however, 3 of 12 patients (25%) treated with combination interferon and ursodeoxycholic acid maintained biochemical normalization at 6 months after therapy (p = 0.08). No difference in liver histology or clearance of hepatitis C viral RNA was noted 6 months after treatment. We conclude that combination therapy with ursodeoxycholic acid and interferon-alpha 2b was no more effective than interferon monotherapy in inducing a biochemical response in previously untreated patients with chronic hepatitis C. Ursodeoxycholic acid, however, may be useful in prolonging the biochemical response to interferon therapy.
[ { "begin_idx": "1568", "end_idx": "1579", "entity_id": "D006526", "entity_type": "Disease", "text_name": "hepatitis C" }, { "begin_idx": "399", "end_idx": "413", "entity_id": "D008107", "entity_type": "Disease", "text_name": "liver diseases" }, { "begin_idx": "0", "end_idx": "32", "entity_id": "D019698", "entity_type": "Disease", "text_name": "Treatment of chronic hepatitis C" }, { "begin_idx": "163", "end_idx": "182", "entity_id": "D019698", "entity_type": "Disease", "text_name": "chronic hepatitis C" }, { "begin_idx": "563", "end_idx": "582", "entity_id": "D019698", "entity_type": "Disease", "text_name": "chronic hepatitis C" }, { "begin_idx": "795", "end_idx": "814", "entity_id": "D019698", "entity_type": "Disease", "text_name": "chronic hepatitis C" }, { "begin_idx": "1836", "end_idx": "1855", "entity_id": "D019698", "entity_type": "Disease", "text_name": "chronic hepatitis C" }, { "begin_idx": "702", "end_idx": "721", "entity_id": "3440", "entity_type": "Gene", "text_name": "interferon-alpha 2b" }, { "begin_idx": "861", "end_idx": "880", "entity_id": "3440", "entity_type": "Gene", "text_name": "interferon-alpha 2b" }, { "begin_idx": "1693", "end_idx": "1712", "entity_id": "3440", "entity_type": "Gene", "text_name": "interferon-alpha 2b" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "702", "end_idx": "721", "entity_id": "3440", "entity_type": "Gene", "text_name": "interferon-alpha 2b" }, { "begin_idx": "1693", "end_idx": "1712", "entity_id": "3440", "entity_type": "Gene", "text_name": "interferon-alpha 2b" } ]
[ { "begin_idx": "0", "end_idx": "32", "entity_id": "D019698", "entity_type": "Disease", "text_name": "Treatment of chronic hepatitis C" }, { "begin_idx": "1568", "end_idx": "1579", "entity_id": "D006526", "entity_type": "Disease", "text_name": "hepatitis C" } ]
[ "interferon-alpha 2b", "interferon-alpha 2b" ]
[ "Treatment of chronic hepatitis C", "hepatitis C" ]
9566422
Novel muscle chloride channel (CLCN1) mutations in myotonia congenita with various modes of inheritance including incomplete dominance and penetrance.
Autosomal-dominant and -recessive myotonia congenita are caused by mutations in the skeletal muscle voltage-gated chloride channel gene (CLCN1). We searched for mutations in this gene in 20 unrelated families with myotonia congenita. We identified 11 different mutations in 10 families. Two of five new mutations (Ala313Thr and Ile556Asn) were both autosomal recessive and dominant with either reduced penetrance or incomplete dominance. Mutations in the CLCN1 gene do not therefore necessarily behave in a classic Mendelian manner.
[ { "begin_idx": "51", "end_idx": "69", "entity_id": "D009224", "entity_type": "Disease", "text_name": "myotonia congenita" }, { "begin_idx": "151", "end_idx": "203", "entity_id": "D009224", "entity_type": "Disease", "text_name": "Autosomal-dominant and -recessive myotonia congenita" }, { "begin_idx": "365", "end_idx": "383", "entity_id": "D009224", "entity_type": "Disease", "text_name": "myotonia congenita" }, { "begin_idx": "31", "end_idx": "36", "entity_id": "1180", "entity_type": "Gene", "text_name": "CLCN1" }, { "begin_idx": "288", "end_idx": "293", "entity_id": "1180", "entity_type": "Gene", "text_name": "CLCN1" }, { "begin_idx": "606", "end_idx": "611", "entity_id": "1180", "entity_type": "Gene", "text_name": "CLCN1" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "31", "end_idx": "36", "entity_id": "1180", "entity_type": "Gene", "text_name": "CLCN1" } ]
[ { "begin_idx": "151", "end_idx": "203", "entity_id": "D009224", "entity_type": "Disease", "text_name": "Autosomal-dominant and -recessive myotonia congenita" } ]
[ "CLCN1" ]
[ "Autosomal-dominant and -recessive myotonia congenita" ]
9568693
Decreased tolbutamide-stimulated insulin secretion in healthy subjects with sequence variants in the high-affinity sulfonylurea receptor gene.
The high-affinity sulfonylurea receptor (SUR1) is, as a subunit of the ATP-sensitive potassium channel, an important regulator of insulin secretion in the pancreatic beta-cell. The aim of this study was to examine if genetic variability of the SUR1 gene was associated with NIDDM or altered pancreatic beta-cell function. Mutational analysis of all the 39 SUR1 exons, including intron-exon boundaries, in 63 NIDDM patients revealed two missense variants, five silent variants in the coding region, and four intron variants. The two missense variants (Asp673Asn and Ser1369Ala) and two sequence variants (ACC-->ACT, Thr759Thr and a c-->t intron variant in position -3 of the exon 16 splice acceptor site) were examined for association with NIDDM and for a possible influence on insulin and C-peptide secretion after intravenous glucose and tolbutamide loads in a random sample of unrelated, healthy, young Danish Caucasians. The Asp673Asn variant in exon 14 was only identified in one NIDDM patient, and the allelic frequency of the Ser1369Ala was similar among 247 control subjects (0.38 [95% CI 0.34-0.42]) and 406 NIDDM patients (0.40 [0.37-0.43]). The allelic frequency of the silent exon 18 Thr775Thr variant was 0.051 (0.035-0.067) in NIDDM patients (n=392) and 0.027 (0.013-0.041) in control subjects (n=246; chi2=4.99, P=0.03). The allelic frequency of the intron variant was similar among NIDDM patients (0.45 [0.42-0.48]) and control subjects (0.44 [0.40-0.48]). Of 386 NIDDM patients, 17 had the combined genotype exon 18 C/T and intron -3c/-3t (0.044 [0.024-0.064]), whereas 3 of 243 control subjects had the same combined genotype (0.012 [0-0.026]; chi2=4.87, P=0.03; odds ratio: 3.69 [1.07-12.71]). Of 380 unrelated, healthy, young Danish Caucasians, 10 (0.026 [0.010-0.042]) had the combined at-risk genotype. These subjects had, on average, a 50% reduction in serum C-peptide and a 40% reduction in serum insulin responses upon tolbutamide injection (P=0.002 and P=0.05, respectively) but normal serum C-peptide and insulin responses upon glucose injection. In conclusion, a silent polymorphism in exon 18 of the SUR1 gene is associated with NIDDM in a Danish Caucasian population. In combination with an intron variant, the association is higher, and young, healthy carriers of the intragenic combination have reduced serum C-peptide and insulin responses to a tolbutamide load.
[ { "begin_idx": "417", "end_idx": "422", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "551", "end_idx": "556", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "882", "end_idx": "887", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1127", "end_idx": "1132", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1259", "end_idx": "1264", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1383", "end_idx": "1388", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1540", "end_idx": "1545", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1622", "end_idx": "1627", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "2300", "end_idx": "2305", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "184", "end_idx": "188", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "387", "end_idx": "391", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "499", "end_idx": "503", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "2271", "end_idx": "2275", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "184", "end_idx": "188", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" } ]
[ { "begin_idx": "417", "end_idx": "422", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" } ]
[ "SUR1" ]
[ "NIDDM" ]
9585598
Systematic analysis of molecular defects in the ferrochelatase gene from patients with erythropoietic protoporphyria.
Erythropoietic protoporphyria (EPP; MIM 177000) is an inherited disorder caused by partial deficiency of ferrochelatase (FECH), the last enzyme in the heme biosynthetic pathway. In EPP patients, the FECH deficiency causes accumulation of free protoporphyrin in the erythron, associated with a painful skin photosensitivity. In rare cases, the massive accumulation of protoporphyrin in hepatocytes may lead to a rapidly progressive liver failure. The mode of inheritance in EPP is complex and can be either autosomal dominant with low clinical penetrance, as it is in most cases, or autosomal recessive. To acquire an in-depth knowledge of the genetic basis of EPP, we conducted a systematic mutation analysis of the FECH gene, following a procedure that combines the exon-by-exon denaturing-gradient-gel-electrophoresis screening of the FECH genomic DNA and direct sequencing. Twenty different mutations, 15 of which are newly described here, have been characterized in 26 of 29 EPP patients of Swiss and French origin. All the EPP patients, including those with liver complications, were heterozygous for the mutations identified in the FECH gene. The deleterious effect of all missense mutations has been assessed by bacterial expression of the respective FECH cDNAs generated by site-directed mutagenesis. Mutations leading to a null allele were a common feature among three EPP pedigrees with liver complications. Our systematic molecular study has resulted in a significant enlargement of the mutation repertoire in the FECH gene and has shed new light on the hereditary behavior of EPP.
[ { "begin_idx": "1181", "end_idx": "1200", "entity_id": "D008107", "entity_type": "Disease", "text_name": "liver complications" }, { "begin_idx": "1496", "end_idx": "1534", "entity_id": "D008107", "entity_type": "Disease", "text_name": "EPP pedigrees with liver complications" }, { "begin_idx": "411", "end_idx": "418", "entity_id": "D010146", "entity_type": "Disease", "text_name": "painful" }, { "begin_idx": "549", "end_idx": "562", "entity_id": "D017093", "entity_type": "Disease", "text_name": "liver failure" }, { "begin_idx": "172", "end_idx": "190", "entity_id": "D030342", "entity_type": "Disease", "text_name": "inherited disorder" }, { "begin_idx": "87", "end_idx": "116", "entity_id": "D046351", "entity_type": "Disease", "text_name": "erythropoietic protoporphyria" }, { "begin_idx": "118", "end_idx": "147", "entity_id": "D046351", "entity_type": "Disease", "text_name": "Erythropoietic protoporphyria" }, { "begin_idx": "149", "end_idx": "152", "entity_id": "D046351", "entity_type": "Disease", "text_name": "EPP" }, { "begin_idx": "299", "end_idx": "302", "entity_id": "D046351", "entity_type": "Disease", "text_name": "EPP" }, { "begin_idx": "317", "end_idx": "332", "entity_id": "D046351", "entity_type": "Disease", "text_name": "FECH deficiency" }, { "begin_idx": "591", "end_idx": "594", "entity_id": "D046351", "entity_type": "Disease", "text_name": "EPP" }, { "begin_idx": "778", "end_idx": "781", "entity_id": "D046351", "entity_type": "Disease", "text_name": "EPP" }, { "begin_idx": "1097", "end_idx": "1100", "entity_id": "D046351", "entity_type": "Disease", "text_name": "EPP" }, { "begin_idx": "1146", "end_idx": "1149", "entity_id": "D046351", "entity_type": "Disease", "text_name": "EPP" }, { "begin_idx": "1706", "end_idx": "1709", "entity_id": "D046351", "entity_type": "Disease", "text_name": "EPP" }, { "begin_idx": "48", "end_idx": "62", "entity_id": "2235", "entity_type": "Gene", "text_name": "ferrochelatase" }, { "begin_idx": "223", "end_idx": "237", "entity_id": "2235", "entity_type": "Gene", "text_name": "ferrochelatase" }, { "begin_idx": "239", "end_idx": "243", "entity_id": "2235", "entity_type": "Gene", "text_name": "FECH" }, { "begin_idx": "834", "end_idx": "838", "entity_id": "2235", "entity_type": "Gene", "text_name": "FECH" }, { "begin_idx": "955", "end_idx": "959", "entity_id": "2235", "entity_type": "Gene", "text_name": "FECH" }, { "begin_idx": "1256", "end_idx": "1260", "entity_id": "2235", "entity_type": "Gene", "text_name": "FECH" }, { "begin_idx": "1376", "end_idx": "1380", "entity_id": "2235", "entity_type": "Gene", "text_name": "FECH" }, { "begin_idx": "1643", "end_idx": "1647", "entity_id": "2235", "entity_type": "Gene", "text_name": "FECH" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "48", "end_idx": "62", "entity_id": "2235", "entity_type": "Gene", "text_name": "ferrochelatase" }, { "begin_idx": "223", "end_idx": "237", "entity_id": "2235", "entity_type": "Gene", "text_name": "ferrochelatase" } ]
[ { "begin_idx": "87", "end_idx": "116", "entity_id": "D046351", "entity_type": "Disease", "text_name": "erythropoietic protoporphyria" }, { "begin_idx": "549", "end_idx": "562", "entity_id": "D017093", "entity_type": "Disease", "text_name": "liver failure" } ]
[ "ferrochelatase", "ferrochelatase" ]
[ "erythropoietic protoporphyria", "liver failure" ]
9588463
An association between NIDDM and a GAA trinucleotide repeat polymorphism in the X25/frataxin (Friedreich's ataxia) gene.
Friedreich's ataxia is the most common hereditary ataxia and is frequently associated with disturbances of glucose metabolism. This autosomal recessive disease is caused by the decreased expression of a mitochondrial protein, frataxin, encoded by the X25 gene. Homozygous expansion of a GAA repeat in the first intron of X25 inhibits frataxin expression and is associated with clinical disease. We evaluated whether heterozygous expansions of the triplet repeat in the frataxin gene X25 may be associated with NIDDM in two genetically distinct populations--one in Germany (n = 358) and the other in the U.S. (n = 292)--using a polymerase chain reaction-based assay. Intermediate expansions (10-36 repeats), which are longer than normal but not sufficient for the appearance of the ataxia phenotype, were found in 24.7 and 27.3% of these two NIDDM cohorts compared with 7.6 and 6.3% of the matched control subjects (both P < 0.001). The odds ratios were 3.36 (95% CI 1.72-6.55) for the German group and 4.01 (2.08-7.74) for the U.S. group. Therefore, we conclude that the X25/frataxin GAA repeat polymorphism is associated with NIDDM in a frequency higher than any other mutation heretofore described. Further studies are needed to elucidate the possible role of frataxin in the pathogenesis of NIDDM.
[ { "begin_idx": "902", "end_idx": "908", "entity_id": "D001259", "entity_type": "Disease", "text_name": "ataxia" }, { "begin_idx": "23", "end_idx": "28", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "631", "end_idx": "636", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "962", "end_idx": "967", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1248", "end_idx": "1253", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1415", "end_idx": "1420", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "94", "end_idx": "113", "entity_id": "D005621", "entity_type": "Disease", "text_name": "Friedreich's ataxia" }, { "begin_idx": "121", "end_idx": "140", "entity_id": "D005621", "entity_type": "Disease", "text_name": "Friedreich's ataxia" }, { "begin_idx": "160", "end_idx": "177", "entity_id": "D013132", "entity_type": "Disease", "text_name": "hereditary ataxia" }, { "begin_idx": "253", "end_idx": "280", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive disease" }, { "begin_idx": "80", "end_idx": "92", "entity_id": "2395", "entity_type": "Gene", "text_name": "X25/frataxin" }, { "begin_idx": "347", "end_idx": "355", "entity_id": "2395", "entity_type": "Gene", "text_name": "frataxin" }, { "begin_idx": "372", "end_idx": "375", "entity_id": "2395", "entity_type": "Gene", "text_name": "X25" }, { "begin_idx": "442", "end_idx": "445", "entity_id": "2395", "entity_type": "Gene", "text_name": "X25" }, { "begin_idx": "455", "end_idx": "463", "entity_id": "2395", "entity_type": "Gene", "text_name": "frataxin" }, { "begin_idx": "590", "end_idx": "598", "entity_id": "2395", "entity_type": "Gene", "text_name": "frataxin" }, { "begin_idx": "604", "end_idx": "607", "entity_id": "2395", "entity_type": "Gene", "text_name": "X25" }, { "begin_idx": "1192", "end_idx": "1195", "entity_id": "2395", "entity_type": "Gene", "text_name": "X25" }, { "begin_idx": "1196", "end_idx": "1204", "entity_id": "2395", "entity_type": "Gene", "text_name": "frataxin" }, { "begin_idx": "1383", "end_idx": "1391", "entity_id": "2395", "entity_type": "Gene", "text_name": "frataxin" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "80", "end_idx": "92", "entity_id": "2395", "entity_type": "Gene", "text_name": "X25/frataxin" }, { "begin_idx": "1383", "end_idx": "1391", "entity_id": "2395", "entity_type": "Gene", "text_name": "frataxin" } ]
[ { "begin_idx": "23", "end_idx": "28", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "121", "end_idx": "140", "entity_id": "D005621", "entity_type": "Disease", "text_name": "Friedreich's ataxia" } ]
[ "X25/frataxin", "frataxin" ]
[ "NIDDM", "Friedreich's ataxia" ]
9596955
[Relationship between angiotensin 1 converting enzyme gene polymorphism and diabetic nephropathy].
OBJECTIVE: To clarify if ACE gene I/D polymorphism attributes to the development of non-insulin dependent diabetes mellitus (NIDDM) and renal complication. METHODS: A fragment of 287 bp Alu sequence in inron 16 of ACE gene was used as I/D polymorphic marker. After PCR amplification of DNA fragment, 12% non-denatured polyacrylamide gel electrophoresis was undertaken to analyze the PCR products. RESULTS: Allele D was the prominent one (frequency 0.75) in the subgroup of DN which was complicated in the earliest stage of NIDDM without HTN, CHD and diabetic retinopathy. However, the frequency of allele D was low (0.39) in the subgroup of NIDDM without DN, HTN, CHD and diabetic retinopathy, in which the duration of each subject was over 5 years. It was significant to compare these two mentioned subgroups (P < 0.02, continuity-adjusted X2test, P < 0.05). The distribution of genotype in the whole NIDDM group without HTN and CHD (69 cases) was very similar to the normal control (110 subjects) (I 0.60 to 0.59, D 0.40 to 0.41). CONCLUSION: The I/D polymorphism of ACE gene may be the factor of genetic predisposition of early-onset diabetic nephropathy in Chinese NIDDM.
[ { "begin_idx": "649", "end_idx": "669", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic retinopathy" }, { "begin_idx": "771", "end_idx": "791", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic retinopathy" }, { "begin_idx": "183", "end_idx": "222", "entity_id": "D003924", "entity_type": "Disease", "text_name": "non-insulin dependent diabetes mellitus" }, { "begin_idx": "224", "end_idx": "229", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "622", "end_idx": "627", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "740", "end_idx": "745", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1001", "end_idx": "1006", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1268", "end_idx": "1273", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "76", "end_idx": "96", "entity_id": "D003928", "entity_type": "Disease", "text_name": "diabetic nephropathy" }, { "begin_idx": "1236", "end_idx": "1256", "entity_id": "D003928", "entity_type": "Disease", "text_name": "diabetic nephropathy" }, { "begin_idx": "235", "end_idx": "253", "entity_id": "D007674", "entity_type": "Disease", "text_name": "renal complication" }, { "begin_idx": "124", "end_idx": "127", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "313", "end_idx": "316", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "1168", "end_idx": "1171", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "124", "end_idx": "127", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "313", "end_idx": "316", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" } ]
[ { "begin_idx": "183", "end_idx": "222", "entity_id": "D003924", "entity_type": "Disease", "text_name": "non-insulin dependent diabetes mellitus" }, { "begin_idx": "649", "end_idx": "669", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic retinopathy" } ]
[ "ACE", "ACE" ]
[ "non-insulin dependent diabetes mellitus", "diabetic retinopathy" ]
9598899
The effect of thalidomide and 2 analogs on collagen induced arthritis.
OBJECTIVE: Thalidomide has been described as an inhibitor of both angiogenesis (which may account for its teratogenic effects on limb bud formation) and tumor necrosis factor-alpha (TNF-alpha) production. We evaluated its therapeutic potential in collagen induced arthritis (CIA), a rat model of rheumatoid arthritis (RA). METHODS: Rats were administered orally 200 mg/kg/day thalidomide (n = 10) or either of 2 analogs, EM-12 (n = 9) or supidimide (n = 9). An additional group was given thalidomide (n = 10) at 200 mg/kg twice daily, and a control group (n = 13) was given vehicle only. At completion of the protocols, serum levels of TNF-alpha and vascular endothelial growth factor (VEGF) were measured. RESULTS: Suppression of inflammatory synovitis by clinical and radiographic criteria was significantly lower in all experimental protocols except the lower dose thalidomide group. The EM-12 analog was the most efficacious, and twice daily thalidomide was better than once daily. The incidence of arthritis onset was comparable among all groups. Strong cell mediated and humoral responses to type II collagen, measured by a radiometric delayed type hypersensitivity assay and anti-type II collagen IgG ELISA, respectively, were similar in the experimental and control groups. TNF-alpha and VEGF levels were increased in all rats immunized with collagen compared to naive controls. CONCLUSION: Thalidomide and its analogs can suppress the clinical severity of rat CIA, but the mechanism of action is not a result of TNF-alpha or VEGF downregulation.
[ { "begin_idx": "1074", "end_idx": "1083", "entity_id": "D001168", "entity_type": "Disease", "text_name": "arthritis" }, { "begin_idx": "43", "end_idx": "69", "entity_id": "D001169", "entity_type": "Disease", "text_name": "collagen induced arthritis" }, { "begin_idx": "318", "end_idx": "344", "entity_id": "D001169", "entity_type": "Disease", "text_name": "collagen induced arthritis" }, { "begin_idx": "346", "end_idx": "349", "entity_id": "D001169", "entity_type": "Disease", "text_name": "CIA" }, { "begin_idx": "1540", "end_idx": "1543", "entity_id": "D001169", "entity_type": "Disease", "text_name": "CIA" }, { "begin_idx": "367", "end_idx": "387", "entity_id": "D001172", "entity_type": "Disease", "text_name": "rheumatoid arthritis" }, { "begin_idx": "389", "end_idx": "391", "entity_id": "D001172", "entity_type": "Disease", "text_name": "RA" }, { "begin_idx": "1226", "end_idx": "1242", "entity_id": "D004342", "entity_type": "Disease", "text_name": "hypersensitivity" }, { "begin_idx": "815", "end_idx": "824", "entity_id": "D013585", "entity_type": "Disease", "text_name": "synovitis" }, { "begin_idx": "224", "end_idx": "251", "entity_id": "7124", "entity_type": "Gene", "text_name": "tumor necrosis factor-alpha" }, { "begin_idx": "253", "end_idx": "262", "entity_id": "7124", "entity_type": "Gene", "text_name": "TNF-alpha" }, { "begin_idx": "707", "end_idx": "716", "entity_id": "7124", "entity_type": "Gene", "text_name": "TNF-alpha" }, { "begin_idx": "1353", "end_idx": "1362", "entity_id": "7124", "entity_type": "Gene", "text_name": "TNF-alpha" }, { "begin_idx": "1592", "end_idx": "1601", "entity_id": "7124", "entity_type": "Gene", "text_name": "TNF-alpha" }, { "begin_idx": "721", "end_idx": "755", "entity_id": "7422", "entity_type": "Gene", "text_name": "vascular endothelial growth factor" }, { "begin_idx": "757", "end_idx": "761", "entity_id": "7422", "entity_type": "Gene", "text_name": "VEGF" }, { "begin_idx": "1367", "end_idx": "1371", "entity_id": "7422", "entity_type": "Gene", "text_name": "VEGF" }, { "begin_idx": "1605", "end_idx": "1609", "entity_id": "7422", "entity_type": "Gene", "text_name": "VEGF" } ]
[ "Yes", "Yes", "No", "No" ]
[ true, true, false, false ]
[ { "begin_idx": "721", "end_idx": "755", "entity_id": "7422", "entity_type": "Gene", "text_name": "vascular endothelial growth factor" }, { "begin_idx": "224", "end_idx": "251", "entity_id": "7124", "entity_type": "Gene", "text_name": "tumor necrosis factor-alpha" }, { "begin_idx": "721", "end_idx": "755", "entity_id": "7422", "entity_type": "Gene", "text_name": "vascular endothelial growth factor" }, { "begin_idx": "1592", "end_idx": "1601", "entity_id": "7124", "entity_type": "Gene", "text_name": "TNF-alpha" } ]
[ { "begin_idx": "43", "end_idx": "69", "entity_id": "D001169", "entity_type": "Disease", "text_name": "collagen induced arthritis" }, { "begin_idx": "43", "end_idx": "69", "entity_id": "D001169", "entity_type": "Disease", "text_name": "collagen induced arthritis" }, { "begin_idx": "1074", "end_idx": "1083", "entity_id": "D001168", "entity_type": "Disease", "text_name": "arthritis" }, { "begin_idx": "815", "end_idx": "824", "entity_id": "D013585", "entity_type": "Disease", "text_name": "synovitis" } ]
[ "vascular endothelial growth factor", "tumor necrosis factor-alpha", "vascular endothelial growth factor", "TNF-alpha" ]
[ "collagen induced arthritis", "collagen induced arthritis", "arthritis", "synovitis" ]
9609997
High proportion of missense mutations of the BRCA1 and BRCA2 genes in Japanese breast cancer families.
Mutations in either of two recently identified genes, BRCA1 and BRCA2, are thought to be responsible for approximately two-thirds of all cases of autosomal-dominantly inherited breast cancer. To examine the nature and frequency of BRCA1 and BRCA2 mutations in Japanese families exhibiting a high incidence of breast cancer, we screened 78 unrelated families in this category for mutations of these two genes. Examining the entire coding sequences as well as exon-intron boundaries of both genes by polymerase chain reaction (PCR) single-strand conformation polymorphism (SSCP) and multiplex-SSCP analysis, we identified possible disease-causing alterations in BRCA1 among affected members of 15 families and in BRCA2 in another 14 families. In 15 of those 29 families, the affected individuals carried missense mutations, although most germline mutations reported worldwide have been deletions or nonsense mutations. Our results, indicating that missense mutations of BRCA1 and BRCA2 tend to predominate over frameshifts or nonsense mutations in Japanese breast cancer families, will contribute significantly to an understanding of mammary tumorigenesis in Japan, and will be of vital importance for future genetic testing.
[ { "begin_idx": "79", "end_idx": "92", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "412", "end_idx": "425", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "1158", "end_idx": "1171", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "249", "end_idx": "293", "entity_id": "D061325", "entity_type": "Disease", "text_name": "autosomal-dominantly inherited breast cancer" }, { "begin_idx": "45", "end_idx": "50", "entity_id": "672", "entity_type": "Gene", "text_name": "BRCA1" }, { "begin_idx": "157", "end_idx": "162", "entity_id": "672", "entity_type": "Gene", "text_name": "BRCA1" }, { "begin_idx": "334", "end_idx": "339", "entity_id": "672", "entity_type": "Gene", "text_name": "BRCA1" }, { "begin_idx": "763", "end_idx": "768", "entity_id": "672", "entity_type": "Gene", "text_name": "BRCA1" }, { "begin_idx": "1071", "end_idx": "1076", "entity_id": "672", "entity_type": "Gene", "text_name": "BRCA1" }, { "begin_idx": "55", "end_idx": "60", "entity_id": "675", "entity_type": "Gene", "text_name": "BRCA2" }, { "begin_idx": "167", "end_idx": "172", "entity_id": "675", "entity_type": "Gene", "text_name": "BRCA2" }, { "begin_idx": "344", "end_idx": "349", "entity_id": "675", "entity_type": "Gene", "text_name": "BRCA2" }, { "begin_idx": "814", "end_idx": "819", "entity_id": "675", "entity_type": "Gene", "text_name": "BRCA2" }, { "begin_idx": "1081", "end_idx": "1086", "entity_id": "675", "entity_type": "Gene", "text_name": "BRCA2" } ]
[ "Yes", "Yes", "No", "No" ]
[ true, true, true, true ]
[ { "begin_idx": "55", "end_idx": "60", "entity_id": "675", "entity_type": "Gene", "text_name": "BRCA2" }, { "begin_idx": "45", "end_idx": "50", "entity_id": "672", "entity_type": "Gene", "text_name": "BRCA1" }, { "begin_idx": "55", "end_idx": "60", "entity_id": "675", "entity_type": "Gene", "text_name": "BRCA2" }, { "begin_idx": "344", "end_idx": "349", "entity_id": "675", "entity_type": "Gene", "text_name": "BRCA2" } ]
[ { "begin_idx": "79", "end_idx": "92", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "79", "end_idx": "92", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "249", "end_idx": "293", "entity_id": "D061325", "entity_type": "Disease", "text_name": "autosomal-dominantly inherited breast cancer" }, { "begin_idx": "249", "end_idx": "293", "entity_id": "D061325", "entity_type": "Disease", "text_name": "autosomal-dominantly inherited breast cancer" } ]
[ "BRCA2", "BRCA1", "BRCA2", "BRCA2" ]
[ "breast cancer", "breast cancer", "autosomal-dominantly inherited breast cancer", "autosomal-dominantly inherited breast cancer" ]
9612607
Effect of zidovudine therapy in patients with HIV infection on endogenous interferon plasma levels and the hepatic cytochrome P450 enzyme system.
In this study, we wanted to investigate if there are differences in endogenous interferon (IFN) plasma levels in patients with different stages of HIV infections before and after therapy with zidovudine (ZDV) and determined the influence of ZDV therapy on the hepatic monooxygenase system by measuring the antipyrine pharmacokinetics. Therefore we investigated the endogenous IFN plasma levels in patients with asymptomatic HIV infection (CDC/WHO A1, n = 10) and patients with AIDS (CDC/WHO C3, n = 10). In AIDS plasma IFN-alpha and IFN-gamma levels are elevated (15.6 +/- 5.8 U/ml; 2.1 +/- 0.7 U/ml) compared to patients with an asymptomatic HIV infection (6.1 +/- 3.3 U/ml; 0.6 +/- 0.3 U/ml). The antipyrine clearance was significantly reduced in the group of AIDS patients (43.1 +/- 7.2 ml/min compared to 56.4 +/- 8.7 ml/min). In a second study with 11 patients in stage CDC/WHO A1/2 and CDC/ WHO B/C3 each, we studied the effect of a 14-day administration of ZDV on the endogenous plasma IFN levels and the CYP450 enzyme activity using the antipyrine pharmacokinetics as a parameter. We investigated the antipyrine clearance, clearance to metabolite and half-life by using HPLC. IFNs were measured by RIA or ELISA, respectively. In the first group no significant alterations of antipyrine kinetics or plasma IFN levels were observed after treatment with ZDV. In contrast to these results, we found a significant decrease in IFN-alpha and IFN-gamma (19.8 +/- 3.6 U/ml, 4.6 +/- 1.5 U/ml before; 7.9 +/- 2.6 U/ml, 1.9 +/- 1.3 U/ml after administration of ZDV), a decrease in antipyrine half-life, an elevation of the antipyrine clearance (49.8 +/- 15.7 ml/min, 57.3 +/- 13.7 ml/min) and an elevation of the clearances to metabolite.
[ { "begin_idx": "623", "end_idx": "627", "entity_id": "D000163", "entity_type": "Disease", "text_name": "AIDS" }, { "begin_idx": "653", "end_idx": "657", "entity_id": "D000163", "entity_type": "Disease", "text_name": "AIDS" }, { "begin_idx": "908", "end_idx": "912", "entity_id": "D000163", "entity_type": "Disease", "text_name": "AIDS" }, { "begin_idx": "46", "end_idx": "59", "entity_id": "D015658", "entity_type": "Disease", "text_name": "HIV infection" }, { "begin_idx": "293", "end_idx": "307", "entity_id": "D015658", "entity_type": "Disease", "text_name": "HIV infections" }, { "begin_idx": "557", "end_idx": "583", "entity_id": "D015658", "entity_type": "Disease", "text_name": "asymptomatic HIV infection" }, { "begin_idx": "776", "end_idx": "802", "entity_id": "D015658", "entity_type": "Disease", "text_name": "asymptomatic HIV infection" }, { "begin_idx": "1029", "end_idx": "1051", "entity_id": "28887", "entity_type": "Gene", "text_name": "A1/2 and CDC/ WHO B/C3" }, { "begin_idx": "665", "end_idx": "674", "entity_id": "3439", "entity_type": "Gene", "text_name": "IFN-alpha" }, { "begin_idx": "1575", "end_idx": "1584", "entity_id": "3439", "entity_type": "Gene", "text_name": "IFN-alpha" }, { "begin_idx": "679", "end_idx": "688", "entity_id": "3458", "entity_type": "Gene", "text_name": "IFN-gamma" }, { "begin_idx": "1589", "end_idx": "1598", "entity_id": "3458", "entity_type": "Gene", "text_name": "IFN-gamma" }, { "begin_idx": "1029", "end_idx": "1051", "entity_id": "597", "entity_type": "Gene", "text_name": "A1/2 and CDC/ WHO B/C3" } ]
[ "Yes", "Yes", "No", "No" ]
[ true, true, true, false ]
[ { "begin_idx": "665", "end_idx": "674", "entity_id": "3439", "entity_type": "Gene", "text_name": "IFN-alpha" }, { "begin_idx": "679", "end_idx": "688", "entity_id": "3458", "entity_type": "Gene", "text_name": "IFN-gamma" }, { "begin_idx": "679", "end_idx": "688", "entity_id": "3458", "entity_type": "Gene", "text_name": "IFN-gamma" }, { "begin_idx": "1029", "end_idx": "1051", "entity_id": "597", "entity_type": "Gene", "text_name": "A1/2 and CDC/ WHO B/C3" } ]
[ { "begin_idx": "623", "end_idx": "627", "entity_id": "D000163", "entity_type": "Disease", "text_name": "AIDS" }, { "begin_idx": "623", "end_idx": "627", "entity_id": "D000163", "entity_type": "Disease", "text_name": "AIDS" }, { "begin_idx": "46", "end_idx": "59", "entity_id": "D015658", "entity_type": "Disease", "text_name": "HIV infection" }, { "begin_idx": "46", "end_idx": "59", "entity_id": "D015658", "entity_type": "Disease", "text_name": "HIV infection" } ]
[ "IFN-alpha", "IFN-gamma", "IFN-gamma", "A1/2 and CDC/ WHO B/C3" ]
[ "AIDS", "AIDS", "HIV infection", "HIV infection" ]
9613851
Association study of structural mutations of the tyrosine hydroxylase gene with schizophrenia and Parkinson's disease.
Tyrosine hydroxylase (TH) gene is the rate-limiting enzyme in the synthesis of catecholamines. Functional polymorphisms of the TH gene may be involved in the pathogenesis of neuropsychiatric diseases such as schizophrenia, affective disorders, and Parkinsonism. This study examined a possible association of two polymorphisms, both of which result in an amino acid change of the TH protein, with schizophrenia and Parkinson's disease (PD). The Val81Met polymorphism is a common variation, although its effect on the enzyme expression is unclear. Leu205Pro polymorphism is a rare mutation that is reported to cause Parkinsonism in infancy for individuals who are homozygous for the mutated type. We genotyped a Japanese sample of 194 schizophrenics, 99 patients with PD, and 161 controls for the Val81Met polymorphism by using mis-match PCR and digestion by the restriction enzyme BalI. There was no significant allelic or genotypic association of the Val81Met polymorphism with schizophrenia or PD. The Leu205Pro polymorphism was examined by using PCR and digestion by AluI; however, there was no individual who carried the mutated type of Pro205 among 50 schizophrenics or 50 patients with PD. Thus we obtained no evidence for the involvement of the two structural mutations of the TH gene in the pathogenesis of schizophrenia or PD.
[ { "begin_idx": "293", "end_idx": "318", "entity_id": "D001523", "entity_type": "Disease", "text_name": "neuropsychiatric diseases" }, { "begin_idx": "98", "end_idx": "117", "entity_id": "D010300", "entity_type": "Disease", "text_name": "Parkinson's disease" }, { "begin_idx": "533", "end_idx": "552", "entity_id": "D010300", "entity_type": "Disease", "text_name": "Parkinson's disease" }, { "begin_idx": "554", "end_idx": "556", "entity_id": "D010300", "entity_type": "Disease", "text_name": "PD" }, { "begin_idx": "885", "end_idx": "887", "entity_id": "D010300", "entity_type": "Disease", "text_name": "PD" }, { "begin_idx": "1114", "end_idx": "1116", "entity_id": "D010300", "entity_type": "Disease", "text_name": "PD" }, { "begin_idx": "1310", "end_idx": "1312", "entity_id": "D010300", "entity_type": "Disease", "text_name": "PD" }, { "begin_idx": "1450", "end_idx": "1452", "entity_id": "D010300", "entity_type": "Disease", "text_name": "PD" }, { "begin_idx": "367", "end_idx": "379", "entity_id": "D010302", "entity_type": "Disease", "text_name": "Parkinsonism" }, { "begin_idx": "733", "end_idx": "745", "entity_id": "D010302", "entity_type": "Disease", "text_name": "Parkinsonism" }, { "begin_idx": "80", "end_idx": "93", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "327", "end_idx": "340", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "515", "end_idx": "528", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "1097", "end_idx": "1110", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "1433", "end_idx": "1446", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenia" }, { "begin_idx": "342", "end_idx": "361", "entity_id": "D019964", "entity_type": "Disease", "text_name": "affective disorders" }, { "begin_idx": "49", "end_idx": "69", "entity_id": "7054", "entity_type": "Gene", "text_name": "tyrosine hydroxylase" }, { "begin_idx": "119", "end_idx": "139", "entity_id": "7054", "entity_type": "Gene", "text_name": "Tyrosine hydroxylase" }, { "begin_idx": "141", "end_idx": "143", "entity_id": "7054", "entity_type": "Gene", "text_name": "TH" }, { "begin_idx": "246", "end_idx": "248", "entity_id": "7054", "entity_type": "Gene", "text_name": "TH" }, { "begin_idx": "498", "end_idx": "500", "entity_id": "7054", "entity_type": "Gene", "text_name": "TH" }, { "begin_idx": "1402", "end_idx": "1404", "entity_id": "7054", "entity_type": "Gene", "text_name": "TH" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "49", "end_idx": "69", "entity_id": "7054", "entity_type": "Gene", "text_name": "tyrosine hydroxylase" }, { "begin_idx": "141", "end_idx": "143", "entity_id": "7054", "entity_type": "Gene", "text_name": "TH" } ]
[ { "begin_idx": "98", "end_idx": "117", "entity_id": "D010300", "entity_type": "Disease", "text_name": "Parkinson's disease" }, { "begin_idx": "293", "end_idx": "318", "entity_id": "D001523", "entity_type": "Disease", "text_name": "neuropsychiatric diseases" } ]
[ "tyrosine hydroxylase", "TH" ]
[ "Parkinson's disease", "neuropsychiatric diseases" ]
9618169
Genetic heterogeneity in familial hyperinsulinism.
Familial hyperinsulinism (HI) is a disorder characterized by dysregulation of insulin secretion and profound hypoglycemia. Mutations in both the Kir6.2 and sulfonylurea receptor (SUR1) genes have been associated with the autosomal recessive form of this disorder. In this study, the spectrum and frequency of SUR1 mutations in HI and their significance to clinical manifestations of the disease were investigated by screening 45 HI probands of various ethnic origins for mutations in the SUR1 gene. Single-strand conformation polymorphism (SSCP) and nucleotide sequence analyses of genomic DNA revealed a total of 17 novel and three previously described mutations in SUR1 . The novel mutations comprised one nonsense and 10 missense mutations, two deletions, three mutations in consensus splice-site sequences and an in-frame insertion of six nucleotides. One mutation occurred in the first nucleotide binding domain (NBF-1) of the SUR1 molecule and another eight mutations were located in the second nucleotide binding domain (NBF-2), including two at highly conserved amino acid residues within the Walker A sequence motif. The majority of the remaining mutations was distributed throughout the three putative transmembrane domains of the SUR1 protein. With the exception of the 3993-9G-->A mutation, which was detected on 4.5% (4/88) disease chromosomes, allelic frequencies for the identified mutations varied between 1.1 and 2.3% for HI chromosomes, indicating that each mutation was rare within the patient cohort. The clinical manifestations of HI in those patients homozygous for mutations in the SUR1 gene are described. In contrast with the allelic homogeneity of HI previously described in Ashkenazi Jewish patients, these findings suggest that a large degree of allelic heterogeneity at the SUR1 locus exists in non-Ashkenazi HI patients. These data have important implications for genetic counseling and prenatal diagnosis of HI, and also provide a basis to further elucidate the molecular mechanisms underlying the pathophysiology of this disease.
[ { "begin_idx": "77", "end_idx": "79", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" }, { "begin_idx": "378", "end_idx": "380", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" }, { "begin_idx": "480", "end_idx": "482", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" }, { "begin_idx": "1490", "end_idx": "1492", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" }, { "begin_idx": "1603", "end_idx": "1605", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" }, { "begin_idx": "1725", "end_idx": "1727", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" }, { "begin_idx": "1889", "end_idx": "1891", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" }, { "begin_idx": "1990", "end_idx": "1992", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" }, { "begin_idx": "160", "end_idx": "172", "entity_id": "D007003", "entity_type": "Disease", "text_name": "hypoglycemia" }, { "begin_idx": "25", "end_idx": "49", "entity_id": "D044903", "entity_type": "Disease", "text_name": "familial hyperinsulinism" }, { "begin_idx": "51", "end_idx": "75", "entity_id": "D044903", "entity_type": "Disease", "text_name": "Familial hyperinsulinism" }, { "begin_idx": "196", "end_idx": "202", "entity_id": "3767", "entity_type": "Gene", "text_name": "Kir6.2" }, { "begin_idx": "230", "end_idx": "234", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "360", "end_idx": "364", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "539", "end_idx": "543", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "718", "end_idx": "722", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "983", "end_idx": "987", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "1292", "end_idx": "1296", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "1656", "end_idx": "1660", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "1854", "end_idx": "1858", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" } ]
[ "Yes", "No" ]
[ false, false ]
[ { "begin_idx": "230", "end_idx": "234", "entity_id": "6833", "entity_type": "Gene", "text_name": "SUR1" }, { "begin_idx": "196", "end_idx": "202", "entity_id": "3767", "entity_type": "Gene", "text_name": "Kir6.2" } ]
[ { "begin_idx": "25", "end_idx": "49", "entity_id": "D044903", "entity_type": "Disease", "text_name": "familial hyperinsulinism" }, { "begin_idx": "1990", "end_idx": "1992", "entity_id": "D006946", "entity_type": "Disease", "text_name": "HI" } ]
[ "SUR1", "Kir6.2" ]
[ "familial hyperinsulinism", "HI" ]
9619639
Association of apolipoprotein E genotype and intronic polymorphism of the presenilin-1 gene with Alzheimer's disease in elderly Taiwan Chinese.
We studied the apoliprotein E (apo-E) allele frequencies and intronic polymorphism of the presenilin-1 (PS-1) gene in 55 patients with late-onset Alzheimer's disease (AD) and 93 age- and sex-matched controls. The apoE epsilon4 allele frequency was significantly higher in the AD group than in the control group (0.255 versus 0.070, P<0.0001). The odds ratio for AD in individuals with either one or two epsilon4 alleles was 5.22 (95% confidence interval: 2.32-11.70). The polymorphism within the region composing intron 3' to exon 8 of the PS-1 gene showed a similar distribution between AD patients and controls. This is the first study on the intronic polymorphism of the PS-1 gene in Chinese. Our results support an association between apoE epislon4 and AD in Chinese, but not between the intronic polymorphism of the PS-1 gene and AD. However, the allele frequency of apo-E epsilon4 among Chinese is lower than that among Caucasians. The interaction between apo-E and PS-1 genotypes is still unclear.
[ { "begin_idx": "97", "end_idx": "116", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "290", "end_idx": "309", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "311", "end_idx": "313", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "420", "end_idx": "422", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "506", "end_idx": "508", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "732", "end_idx": "734", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "901", "end_idx": "903", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "979", "end_idx": "981", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "15", "end_idx": "31", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "159", "end_idx": "173", "entity_id": "348", "entity_type": "Gene", "text_name": "apoliprotein E" }, { "begin_idx": "175", "end_idx": "180", "entity_id": "348", "entity_type": "Gene", "text_name": "apo-E" }, { "begin_idx": "357", "end_idx": "361", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE" }, { "begin_idx": "883", "end_idx": "887", "entity_id": "348", "entity_type": "Gene", "text_name": "apoE" }, { "begin_idx": "1016", "end_idx": "1021", "entity_id": "348", "entity_type": "Gene", "text_name": "apo-E" }, { "begin_idx": "1106", "end_idx": "1111", "entity_id": "348", "entity_type": "Gene", "text_name": "apo-E" }, { "begin_idx": "74", "end_idx": "86", "entity_id": "5663", "entity_type": "Gene", "text_name": "presenilin-1" }, { "begin_idx": "234", "end_idx": "246", "entity_id": "5663", "entity_type": "Gene", "text_name": "presenilin-1" }, { "begin_idx": "248", "end_idx": "252", "entity_id": "5663", "entity_type": "Gene", "text_name": "PS-1" }, { "begin_idx": "684", "end_idx": "688", "entity_id": "5663", "entity_type": "Gene", "text_name": "PS-1" }, { "begin_idx": "818", "end_idx": "822", "entity_id": "5663", "entity_type": "Gene", "text_name": "PS-1" }, { "begin_idx": "965", "end_idx": "969", "entity_id": "5663", "entity_type": "Gene", "text_name": "PS-1" }, { "begin_idx": "1116", "end_idx": "1120", "entity_id": "5663", "entity_type": "Gene", "text_name": "PS-1" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "15", "end_idx": "31", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "74", "end_idx": "86", "entity_id": "5663", "entity_type": "Gene", "text_name": "presenilin-1" } ]
[ { "begin_idx": "97", "end_idx": "116", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "420", "end_idx": "422", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" } ]
[ "apolipoprotein E", "presenilin-1" ]
[ "Alzheimer's disease", "AD" ]
9627112
Down-regulation of T1A12/mac25, a novel insulin-like growth factor binding protein related gene, is associated with disease progression in breast carcinomas.
To define genes that are essential to the initiation and progression of breast cancer we utilized subtractive hybridization and differential display cloning techniques and isolated over 950 cDNAs from breast cell-lines derived from matched normal and tumor tissue. Of these, 102 cDNAs were characterized by DNA sequencing and Northern blot analysis. GenBank searches showed that one of these genes, T1A12 is identical to mac25, an insulin-like growth factor-binding protein related gene. Antibodies generated against the C-terminal region of the T1A12/mac25 protein were used to investigate its expression in 60 primary breast tissues. Sections of 12 benign, 16 ductal carcinoma in situ and 32 infiltrating ductal carcinoma specimens were examined. Strong immunoperoxidase staining was observed in luminal epithelial cells of normal lobules and ducts, in apocrine cells of cysts and fibroadenomas. Moderate to weak protein expression was found in hyperplastic and DCIS cells, but no specific staining was detected in invasive carcinoma cells. FISH mapping using a PAC clone localized the T1A12/mac25 gene to 4q12-13. Microsatellite length polymorphism was studied using markers for 4q in paired normal and tumor breast tissues. Thirty-three per cent (10/30) of the samples were found to be polymorphic with D4S189 and D4S231 microsatellite markers and LOH was detected in 50% (5/10) of these informative samples. Our data indicate that T1A12/mac25 expression is abrogated during breast cancer progression concomitant with loss of heterozygosity on chromosome 4q. T1A12/mac25 may therefore have a tumor suppressor-like function and its expression could indicate a disease with a more favorable status, having a better prognosis.
[ { "begin_idx": "139", "end_idx": "156", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast carcinomas" }, { "begin_idx": "230", "end_idx": "243", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer" }, { "begin_idx": "1637", "end_idx": "1662", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer progression" }, { "begin_idx": "1184", "end_idx": "1193", "entity_id": "D002277", "entity_type": "Disease", "text_name": "carcinoma" }, { "begin_idx": "1122", "end_idx": "1126", "entity_id": "D002285", "entity_type": "Disease", "text_name": "DCIS" }, { "begin_idx": "409", "end_idx": "414", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" }, { "begin_idx": "1364", "end_idx": "1369", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" }, { "begin_idx": "1754", "end_idx": "1759", "entity_id": "D009369", "entity_type": "Disease", "text_name": "tumor" }, { "begin_idx": "1041", "end_idx": "1054", "entity_id": "D018226", "entity_type": "Disease", "text_name": "fibroadenomas" }, { "begin_idx": "820", "end_idx": "836", "entity_id": "D044584", "entity_type": "Disease", "text_name": "ductal carcinoma" }, { "begin_idx": "865", "end_idx": "881", "entity_id": "D044584", "entity_type": "Disease", "text_name": "ductal carcinoma" }, { "begin_idx": "25", "end_idx": "30", "entity_id": "3490", "entity_type": "Gene", "text_name": "mac25" }, { "begin_idx": "579", "end_idx": "584", "entity_id": "3490", "entity_type": "Gene", "text_name": "mac25" }, { "begin_idx": "710", "end_idx": "715", "entity_id": "3490", "entity_type": "Gene", "text_name": "mac25" }, { "begin_idx": "1252", "end_idx": "1257", "entity_id": "3490", "entity_type": "Gene", "text_name": "mac25" }, { "begin_idx": "1600", "end_idx": "1605", "entity_id": "3490", "entity_type": "Gene", "text_name": "mac25" }, { "begin_idx": "1727", "end_idx": "1732", "entity_id": "3490", "entity_type": "Gene", "text_name": "mac25" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "25", "end_idx": "30", "entity_id": "3490", "entity_type": "Gene", "text_name": "mac25" }, { "begin_idx": "710", "end_idx": "715", "entity_id": "3490", "entity_type": "Gene", "text_name": "mac25" } ]
[ { "begin_idx": "1637", "end_idx": "1662", "entity_id": "D001943", "entity_type": "Disease", "text_name": "breast cancer progression" }, { "begin_idx": "865", "end_idx": "881", "entity_id": "D044584", "entity_type": "Disease", "text_name": "ductal carcinoma" } ]
[ "mac25", "mac25" ]
[ "breast cancer progression", "ductal carcinoma" ]
9633815
Spectrum of mutations in the fumarylacetoacetate hydrolase gene of tyrosinemia type 1 patients in northwestern Europe and Mediterranean countries.
Hereditary tyrosinemia type 1 (HT1) is a rare metabolic disease caused by a deficient activity of the enzyme fumarylacetoacetase (FAH). To investigate the molecular heterogeneity of tyrosinemia, the geographic distribution and the genotype-phenotype relationship, we have analyzed the FAH genotype of 25 HT1 patients. Mutation screening was performed by PCR amplification of exons 1-14 of the FAH gene, followed by SSCP analysis and direct sequencing of the amplified exons. Fourteen different mutations were found, of which seven were novel, viz. three missense mutations (G158D, P261L, F405H), a deletion of three nucleotides causing a deletion of serine (DEL366S) and three splice site mutations: IVS2+1(g-t), IVS6-1(g-c), IVS8-1(g-c). The splice site mutations IVS6-1(g-t) and IVS12+5(g-a) were frequently found in countries around the Mediterranean and northwestern Europe, respectively. No clear correlation between the genotype and the three major HT1 subtypes could be established.
[ { "begin_idx": "193", "end_idx": "210", "entity_id": "D008659", "entity_type": "Disease", "text_name": "metabolic disease" }, { "begin_idx": "67", "end_idx": "85", "entity_id": "D020176", "entity_type": "Disease", "text_name": "tyrosinemia type 1" }, { "begin_idx": "147", "end_idx": "176", "entity_id": "D020176", "entity_type": "Disease", "text_name": "Hereditary tyrosinemia type 1" }, { "begin_idx": "178", "end_idx": "181", "entity_id": "D020176", "entity_type": "Disease", "text_name": "HT1" }, { "begin_idx": "329", "end_idx": "340", "entity_id": "D020176", "entity_type": "Disease", "text_name": "tyrosinemia" }, { "begin_idx": "451", "end_idx": "454", "entity_id": "D020176", "entity_type": "Disease", "text_name": "HT1" }, { "begin_idx": "1102", "end_idx": "1105", "entity_id": "D020176", "entity_type": "Disease", "text_name": "HT1" }, { "begin_idx": "277", "end_idx": "280", "entity_id": "2184", "entity_type": "Gene", "text_name": "FAH" }, { "begin_idx": "432", "end_idx": "435", "entity_id": "2184", "entity_type": "Gene", "text_name": "FAH" }, { "begin_idx": "540", "end_idx": "543", "entity_id": "2184", "entity_type": "Gene", "text_name": "FAH" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "277", "end_idx": "280", "entity_id": "2184", "entity_type": "Gene", "text_name": "FAH" }, { "begin_idx": "540", "end_idx": "543", "entity_id": "2184", "entity_type": "Gene", "text_name": "FAH" } ]
[ { "begin_idx": "147", "end_idx": "176", "entity_id": "D020176", "entity_type": "Disease", "text_name": "Hereditary tyrosinemia type 1" }, { "begin_idx": "193", "end_idx": "210", "entity_id": "D008659", "entity_type": "Disease", "text_name": "metabolic disease" } ]
[ "FAH", "FAH" ]
[ "Hereditary tyrosinemia type 1", "metabolic disease" ]
9633819
Different missense mutations in histidine-108 of lysosomal acid lipase cause cholesteryl ester storage disease in unrelated compound heterozygous and hemizygous individuals.
Cholesteryl ester storage disease (CESD) and Wolman disease (WD) are both autosomal recessive disorders associated with reduced activity of lysosomal acid lipase (LAL), that leads to the tissue accumulation of cholesteryl esters in endosomes and lysosomes. WD is caused by genetic defects of LAL that leave no residual enzymatic activity, while in CESD patients a residual LAL activity can be identified. We have analyzed the LAL cDNA in three CESD patients from two nonrelated families and identified the mutations responsible for the disease. The associated genetic defects characterized revealed compound heterozygosity for a splice defect leading to skipping of exon 8, due to a G-->A transition at position -1 of the exon 8 splice donor site, and a point mutation leading to a Hisl08Pro change (CAT-->CCT) in two patients (siblings) with mild CESD phenotype. A further CESD patient was hemizygous for a His108-->Arg missense mutation (CAT-->CGT) in combination with a partial deletion of the LAL gene and was affected more severely. Expression of the LAL enzymes with the His108-->Pro and His108-->Arg mutation in insect cells revealed residual enzymatic activities of 4.6% versus 2.7%, respectively, compared with controls. Therefore, His108 seems to play a crucial role in folding or catalytic activity of the lysosomal acid lipase. This is the first description of two different, naturally occurring mutations involving the same amino acid residue in the lysosomal acid lipase in unrelated CESD patients. Moreover, our results demonstrate that the variable manifestation of CESD can be explained by mutation-dependent, variable inactivation of the LAL enzyme.
[ { "begin_idx": "77", "end_idx": "110", "entity_id": "C531854", "entity_type": "Disease", "text_name": "cholesteryl ester storage disease" }, { "begin_idx": "174", "end_idx": "207", "entity_id": "C531854", "entity_type": "Disease", "text_name": "Cholesteryl ester storage disease" }, { "begin_idx": "209", "end_idx": "213", "entity_id": "C531854", "entity_type": "Disease", "text_name": "CESD" }, { "begin_idx": "522", "end_idx": "526", "entity_id": "C531854", "entity_type": "Disease", "text_name": "CESD" }, { "begin_idx": "618", "end_idx": "622", "entity_id": "C531854", "entity_type": "Disease", "text_name": "CESD" }, { "begin_idx": "1022", "end_idx": "1026", "entity_id": "C531854", "entity_type": "Disease", "text_name": "CESD" }, { "begin_idx": "1048", "end_idx": "1052", "entity_id": "C531854", "entity_type": "Disease", "text_name": "CESD" }, { "begin_idx": "1672", "end_idx": "1676", "entity_id": "C531854", "entity_type": "Disease", "text_name": "CESD" }, { "begin_idx": "1756", "end_idx": "1760", "entity_id": "C531854", "entity_type": "Disease", "text_name": "CESD" }, { "begin_idx": "219", "end_idx": "233", "entity_id": "D015223", "entity_type": "Disease", "text_name": "Wolman disease" }, { "begin_idx": "235", "end_idx": "237", "entity_id": "D015223", "entity_type": "Disease", "text_name": "WD" }, { "begin_idx": "431", "end_idx": "433", "entity_id": "D015223", "entity_type": "Disease", "text_name": "WD" }, { "begin_idx": "248", "end_idx": "277", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive disorders" }, { "begin_idx": "337", "end_idx": "340", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" }, { "begin_idx": "466", "end_idx": "469", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" }, { "begin_idx": "547", "end_idx": "550", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" }, { "begin_idx": "600", "end_idx": "603", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" }, { "begin_idx": "1171", "end_idx": "1174", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" }, { "begin_idx": "1230", "end_idx": "1233", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" }, { "begin_idx": "1830", "end_idx": "1833", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "337", "end_idx": "340", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" }, { "begin_idx": "547", "end_idx": "550", "entity_id": "3988", "entity_type": "Gene", "text_name": "LAL" } ]
[ { "begin_idx": "219", "end_idx": "233", "entity_id": "D015223", "entity_type": "Disease", "text_name": "Wolman disease" }, { "begin_idx": "522", "end_idx": "526", "entity_id": "C531854", "entity_type": "Disease", "text_name": "CESD" } ]
[ "LAL", "LAL" ]
[ "Wolman disease", "CESD" ]
9643286
A polymorphism of the CC16 gene is associated with an increased risk of asthma.
Several quantitative traits associated with the asthma phenotype have been linked to markers on chromosome 11q13, although the gene responsible has yet to be well established. The gene for Clara cell secretory protein (CC16) is an ideal candidate for involvement in an inherited predisposition to asthma because of its chromosomal location, the role of the CC16 protein in controlling airway inflammation, and differences in levels of the protein between asthmatics and healthy controls. All three CC16 exons were screened in an unselected population of 266 subjects from 76 families and a cohort of 52 severely asthmatic children. A combination of single strand conformational polymorphism (SSCP) analysis, heteroduplex analysis, DNA sequencing, and restriction digestion was used. Mutation detection methods identified an adenine to guanine substitution in the CC16 gene at position 38 (A38G) downstream from the transcription initiation site within the non-coding region of exon 1. In the unselected population, 43.6% were homozygous for the polymorphic sequence (38GG) and 46.2% were heterozygous (38AG). All the asthmatic and unaffected children from both populations were selected for an unmatched case control analysis consisting of 67 asthmatic and 46 unaffected subjects. Those homozygous for the published sequence (38AA) had a 6.9-fold increased risk of developing asthma (p=0.049) and heterozygotes (38AG) a 4.2-fold increased risk (p=0.028). Modelling of genotype as a continuous covariate indicated evidence of a significant linear trend across the three genotypes (odds ratio=2.84 per unit increase in genotype code, p=0.018). These associations were independent of age, gender, and tobacco smoke exposure. These data and the known anti-inflammatory role of CC16 in the respiratory tract suggest that alteration to the gene at position 38 may contribute to asthma.
[ { "begin_idx": "72", "end_idx": "78", "entity_id": "D001249", "entity_type": "Disease", "text_name": "asthma" }, { "begin_idx": "128", "end_idx": "134", "entity_id": "D001249", "entity_type": "Disease", "text_name": "asthma" }, { "begin_idx": "377", "end_idx": "383", "entity_id": "D001249", "entity_type": "Disease", "text_name": "asthma" }, { "begin_idx": "1456", "end_idx": "1462", "entity_id": "D001249", "entity_type": "Disease", "text_name": "asthma" }, { "begin_idx": "1952", "end_idx": "1958", "entity_id": "D001249", "entity_type": "Disease", "text_name": "asthma" }, { "begin_idx": "692", "end_idx": "701", "entity_id": "D013224", "entity_type": "Disease", "text_name": "asthmatic" }, { "begin_idx": "1197", "end_idx": "1206", "entity_id": "D013224", "entity_type": "Disease", "text_name": "asthmatic" }, { "begin_idx": "1323", "end_idx": "1332", "entity_id": "D013224", "entity_type": "Disease", "text_name": "asthmatic" }, { "begin_idx": "22", "end_idx": "26", "entity_id": "7356", "entity_type": "Gene", "text_name": "CC16" }, { "begin_idx": "299", "end_idx": "303", "entity_id": "7356", "entity_type": "Gene", "text_name": "CC16" }, { "begin_idx": "437", "end_idx": "441", "entity_id": "7356", "entity_type": "Gene", "text_name": "CC16" }, { "begin_idx": "578", "end_idx": "582", "entity_id": "7356", "entity_type": "Gene", "text_name": "CC16" }, { "begin_idx": "943", "end_idx": "947", "entity_id": "7356", "entity_type": "Gene", "text_name": "CC16" }, { "begin_idx": "1853", "end_idx": "1857", "entity_id": "7356", "entity_type": "Gene", "text_name": "CC16" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "22", "end_idx": "26", "entity_id": "7356", "entity_type": "Gene", "text_name": "CC16" }, { "begin_idx": "1853", "end_idx": "1857", "entity_id": "7356", "entity_type": "Gene", "text_name": "CC16" } ]
[ { "begin_idx": "72", "end_idx": "78", "entity_id": "D001249", "entity_type": "Disease", "text_name": "asthma" }, { "begin_idx": "1323", "end_idx": "1332", "entity_id": "D013224", "entity_type": "Disease", "text_name": "asthmatic" } ]
[ "CC16", "CC16" ]
[ "asthma", "asthmatic" ]
9650766
A novel mutation of the beta-glucocerebrosidase gene associated with neurologic manifestations in three sibs.
We report on a sibship in which three members were affected by Gaucher disease. Molecular analysis of the patients showed homozygosity for a novel mutation (C5390G) of the beta-glucocerebrosidase gene, resulting in the substitution of the arginine 353 with a glycine. Western blot analysis showed a reduced amount of beta-glucocerebrosidase-related polypeptides in fibroblasts. The phenotype resulting from this mutation is characterized by visceral and skeletal manifestations. In addition, the presence of seizures and electrophysiological abnormalities only in the 3 patients and in none of the other unaffected sibs suggests that the mutation is responsible for neurologic involvement.
[ { "begin_idx": "173", "end_idx": "188", "entity_id": "D005776", "entity_type": "Disease", "text_name": "Gaucher disease" }, { "begin_idx": "618", "end_idx": "626", "entity_id": "D012640", "entity_type": "Disease", "text_name": "seizures" }, { "begin_idx": "24", "end_idx": "47", "entity_id": "2629", "entity_type": "Gene", "text_name": "beta-glucocerebrosidase" }, { "begin_idx": "282", "end_idx": "305", "entity_id": "2629", "entity_type": "Gene", "text_name": "beta-glucocerebrosidase" }, { "begin_idx": "427", "end_idx": "450", "entity_id": "2629", "entity_type": "Gene", "text_name": "beta-glucocerebrosidase" } ]
[ "Yes", "No" ]
[ false, false ]
[ { "begin_idx": "24", "end_idx": "47", "entity_id": "2629", "entity_type": "Gene", "text_name": "beta-glucocerebrosidase" }, { "begin_idx": "282", "end_idx": "305", "entity_id": "2629", "entity_type": "Gene", "text_name": "beta-glucocerebrosidase" } ]
[ { "begin_idx": "173", "end_idx": "188", "entity_id": "D005776", "entity_type": "Disease", "text_name": "Gaucher disease" }, { "begin_idx": "618", "end_idx": "626", "entity_id": "D012640", "entity_type": "Disease", "text_name": "seizures" } ]
[ "beta-glucocerebrosidase", "beta-glucocerebrosidase" ]
[ "Gaucher disease", "seizures" ]
9654205
LDL-R and Apo-B-100 gene mutations in Polish familial hypercholesterolemias.
A group of 30 Polish families with clinical signs of familial hypercholesterolemia was studied for the presence of germ-line mutations in the LDL-R and ApoB-100 genes. Screening of the LDL-R gene was performed at the genomic DNA level by single-strand conformation polymorphism analysis of all 18 exons and extended by sequencing of polymerase chain reaction (PCR) products showing abnormalities. The occurrence of large LDL-R gene alterations was evaluated by analysis of restriction enzyme patterns on Southern blots and using the long-PCR technique. The ApoB-100 gene was studied by combined allele-specific and asymmetric PCR for the occurrence of the common B-3500 missense mutation G to A at nucleotide position 10,708. Germ-line mutations were found in 17 families. In 12 of them LDL-R gene mutations were detected. Three of 11 different mutations had previously been described in other populations (3-bp deletion of codon 197; Ser156Leu; Gly571Glu). Of the mutations not previously recognized and identified in Polish families, there were three small deletions (2-bp deletion AG at codon 291; 4-bp deletion CCCT at codons 661-662; 1-bp deletion A at codon 830), and four point mutations (Arg239Stop, Cys331Stop, Asn543Ser, Gln665Stop). Additionally, one large (approximately 1-kb) LDL-R gene deletion between exons 6 and 9 was identified. In five families, the B-3500 mutation within the ApoB-100 gene was revealed.
[ { "begin_idx": "45", "end_idx": "75", "entity_id": "D006938", "entity_type": "Disease", "text_name": "familial hypercholesterolemias" }, { "begin_idx": "130", "end_idx": "159", "entity_id": "D006938", "entity_type": "Disease", "text_name": "familial hypercholesterolemia" }, { "begin_idx": "10", "end_idx": "19", "entity_id": "338", "entity_type": "Gene", "text_name": "Apo-B-100" }, { "begin_idx": "229", "end_idx": "237", "entity_id": "338", "entity_type": "Gene", "text_name": "ApoB-100" }, { "begin_idx": "634", "end_idx": "642", "entity_id": "338", "entity_type": "Gene", "text_name": "ApoB-100" }, { "begin_idx": "1473", "end_idx": "1481", "entity_id": "338", "entity_type": "Gene", "text_name": "ApoB-100" }, { "begin_idx": "0", "end_idx": "5", "entity_id": "3949", "entity_type": "Gene", "text_name": "LDL-R" }, { "begin_idx": "219", "end_idx": "224", "entity_id": "3949", "entity_type": "Gene", "text_name": "LDL-R" }, { "begin_idx": "262", "end_idx": "267", "entity_id": "3949", "entity_type": "Gene", "text_name": "LDL-R" }, { "begin_idx": "498", "end_idx": "503", "entity_id": "3949", "entity_type": "Gene", "text_name": "LDL-R" }, { "begin_idx": "864", "end_idx": "869", "entity_id": "3949", "entity_type": "Gene", "text_name": "LDL-R" }, { "begin_idx": "1366", "end_idx": "1371", "entity_id": "3949", "entity_type": "Gene", "text_name": "LDL-R" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "0", "end_idx": "5", "entity_id": "3949", "entity_type": "Gene", "text_name": "LDL-R" }, { "begin_idx": "10", "end_idx": "19", "entity_id": "338", "entity_type": "Gene", "text_name": "Apo-B-100" } ]
[ { "begin_idx": "45", "end_idx": "75", "entity_id": "D006938", "entity_type": "Disease", "text_name": "familial hypercholesterolemias" }, { "begin_idx": "45", "end_idx": "75", "entity_id": "D006938", "entity_type": "Disease", "text_name": "familial hypercholesterolemias" } ]
[ "LDL-R", "Apo-B-100" ]
[ "familial hypercholesterolemias", "familial hypercholesterolemias" ]
9655392
Smad2 role in mesoderm formation, left-right patterning and craniofacial development.
Signalling by the transforming growth factor-beta (TGF-beta) superfamily of proteins depends on the phosphorylation and activation of SMAD proteins by heteromeric complexes of ligand-specific type I and type II receptors with serine/threonine-kinase activity. The vertebrate SMAD family includes at least nine members, of which Smad2 has been shown to mediate signalling by activin and TGF-beta. In Xenopus, Smad2 can induce dorsal mesoderm, mimicking Vg-1, activin and nodal. Here we investigate the function of Smad2 in mammalian development by generating two independent Smad2 mutant alleles in mice by gene targeting. We show that homozygous mutant embryos fail to form an organized egg cylinder and lack mesoderm, like mutant mice lacking nodal or ActRIB, the gene encoding the activin type-I receptor. About 20 per cent of Smad2 heterozygous embryos have severe gastrulation defects and lack mandibles or eyes, indicating that the gene dosage of Smad2 is critical for signalling. Mice trans-heterozygous for both Smad2 and nodal mutations display a range of phenotypes, including gastrulation defects, complex craniofacial abnormalities such as cyclopia, and defects in left-right patterning, indicating that Smad2 may mediate nodal signalling in these developmental processes. Our results show that Smad2 function is essential for early development and for several patterning processes in mice.
[ { "begin_idx": "954", "end_idx": "974", "entity_id": "D000013", "entity_type": "Disease", "text_name": "gastrulation defects" }, { "begin_idx": "1172", "end_idx": "1192", "entity_id": "D000013", "entity_type": "Disease", "text_name": "gastrulation defects" }, { "begin_idx": "1237", "end_idx": "1245", "entity_id": "D016142", "entity_type": "Disease", "text_name": "cyclopia" }, { "begin_idx": "1202", "end_idx": "1228", "entity_id": "D019465", "entity_type": "Disease", "text_name": "craniofacial abnormalities" }, { "begin_idx": "0", "end_idx": "5", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "414", "end_idx": "419", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "599", "end_idx": "604", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "660", "end_idx": "665", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "915", "end_idx": "920", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "1038", "end_idx": "1043", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "1105", "end_idx": "1110", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "1301", "end_idx": "1306", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "1392", "end_idx": "1397", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "494", "end_idx": "499", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "538", "end_idx": "542", "entity_id": "677862", "entity_type": "Gene", "text_name": "Vg-1" }, { "begin_idx": "137", "end_idx": "145", "entity_id": "7040", "entity_type": "Gene", "text_name": "TGF-beta" }, { "begin_idx": "472", "end_idx": "480", "entity_id": "7040", "entity_type": "Gene", "text_name": "TGF-beta" }, { "begin_idx": "839", "end_idx": "845", "entity_id": "91", "entity_type": "Gene", "text_name": "ActRIB" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "0", "end_idx": "5", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" }, { "begin_idx": "1105", "end_idx": "1110", "entity_id": "4087", "entity_type": "Gene", "text_name": "Smad2" } ]
[ { "begin_idx": "1202", "end_idx": "1228", "entity_id": "D019465", "entity_type": "Disease", "text_name": "craniofacial abnormalities" }, { "begin_idx": "1172", "end_idx": "1192", "entity_id": "D000013", "entity_type": "Disease", "text_name": "gastrulation defects" } ]
[ "Smad2", "Smad2" ]
[ "craniofacial abnormalities", "gastrulation defects" ]
9691087
Mutations in the liver glycogen synthase gene in children with hypoglycemia due to glycogen storage disease type 0.
Glycogen storage disease type 0 (GSD-0) is a rare form of fasting hypoglycemia presenting in infancy or early childhood and accompanied by high blood ketones and low alanine and lactate concentrations. Although feeding relieves symptoms, it often results in postprandial hyperglycemia and hyperlactatemia. The glycogen synthase (GS) activity has been low or immeasurable in liver biopsies, whereas the liver glycogen content has been only moderately decreased. To investigate whether mutations in the liver GS gene (GYS2) on chromosome 12p12.2 were involved in GSD-0, we determined the exon-intron structure of the GYS2 gene and examined nine affected children from five families for linkage of GSD-0 to the GYS2 gene. Mutation screening of the 16 GYS2 exons was done by single-strand conformational polymorphism (SSCP) and direct sequencing. Liver GS deficiency was diagnosed from liver biopsies (GS activity and glycogen content). GS activity in the liver of the affected children was extremely low or nil, resulting in subnormal glycogen content. After suggestive linkage to the GYS2 gene had been established (LOD score = 2.9; P < 0.01), mutation screening revealed several different mutations in these families, including a premature stop codon in exon 5 (Arg246X), a 5'-donor splice site mutation in intron 6 (G+1T--> CT), and missense mutations Asn39Ser, Ala339Pro, His446Asp, Pro479Gln, Ser483Pro, and Met491Arg. Seven of the affected children carried mutations on both alleles. The mutations could not be found in 200 healthy persons. Expression of the mutated enzymes in COS7 cells indicated severely impaired GS activity. In conclusion, the results demonstrate that GSD-0 is caused by different mutations in the GYS2 gene.
[ { "begin_idx": "959", "end_idx": "978", "entity_id": "C565485", "entity_type": "Disease", "text_name": "Liver GS deficiency" }, { "begin_idx": "23", "end_idx": "40", "entity_id": "C566917", "entity_type": "Disease", "text_name": "glycogen synthase" }, { "begin_idx": "83", "end_idx": "114", "entity_id": "C566917", "entity_type": "Disease", "text_name": "glycogen storage disease type 0" }, { "begin_idx": "116", "end_idx": "147", "entity_id": "C566917", "entity_type": "Disease", "text_name": "Glycogen storage disease type 0" }, { "begin_idx": "149", "end_idx": "154", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GSD-0" }, { "begin_idx": "426", "end_idx": "443", "entity_id": "C566917", "entity_type": "Disease", "text_name": "glycogen synthase" }, { "begin_idx": "445", "end_idx": "447", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GS" }, { "begin_idx": "623", "end_idx": "625", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GS" }, { "begin_idx": "677", "end_idx": "682", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GSD-0" }, { "begin_idx": "811", "end_idx": "816", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GSD-0" }, { "begin_idx": "1014", "end_idx": "1016", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GS" }, { "begin_idx": "1049", "end_idx": "1051", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GS" }, { "begin_idx": "1736", "end_idx": "1738", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GS" }, { "begin_idx": "1793", "end_idx": "1798", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GSD-0" }, { "begin_idx": "387", "end_idx": "400", "entity_id": "D006943", "entity_type": "Disease", "text_name": "hyperglycemia" }, { "begin_idx": "63", "end_idx": "75", "entity_id": "D007003", "entity_type": "Disease", "text_name": "hypoglycemia" }, { "begin_idx": "182", "end_idx": "194", "entity_id": "D007003", "entity_type": "Disease", "text_name": "hypoglycemia" }, { "begin_idx": "405", "end_idx": "420", "entity_id": "D065906", "entity_type": "Disease", "text_name": "hyperlactatemia" }, { "begin_idx": "632", "end_idx": "636", "entity_id": "2998", "entity_type": "Gene", "text_name": "GYS2" }, { "begin_idx": "731", "end_idx": "735", "entity_id": "2998", "entity_type": "Gene", "text_name": "GYS2" }, { "begin_idx": "824", "end_idx": "828", "entity_id": "2998", "entity_type": "Gene", "text_name": "GYS2" }, { "begin_idx": "864", "end_idx": "868", "entity_id": "2998", "entity_type": "Gene", "text_name": "GYS2" }, { "begin_idx": "1198", "end_idx": "1202", "entity_id": "2998", "entity_type": "Gene", "text_name": "GYS2" }, { "begin_idx": "1839", "end_idx": "1843", "entity_id": "2998", "entity_type": "Gene", "text_name": "GYS2" } ]
[ "Yes", "No" ]
[ false, true ]
[ { "begin_idx": "632", "end_idx": "636", "entity_id": "2998", "entity_type": "Gene", "text_name": "GYS2" }, { "begin_idx": "824", "end_idx": "828", "entity_id": "2998", "entity_type": "Gene", "text_name": "GYS2" } ]
[ { "begin_idx": "959", "end_idx": "978", "entity_id": "C565485", "entity_type": "Disease", "text_name": "Liver GS deficiency" }, { "begin_idx": "1049", "end_idx": "1051", "entity_id": "C566917", "entity_type": "Disease", "text_name": "GS" } ]
[ "GYS2", "GYS2" ]
[ "Liver GS deficiency", "GS" ]
9697698
Febrile seizures and generalized epilepsy associated with a mutation in the Na+-channel beta1 subunit gene SCN1B.
Febrile seizures affect approximately 3% of all children under six years of age and are by far the most common seizure disorder. A small proportion of children with febrile seizures later develop ongoing epilepsy with afebrile seizures. Segregation analysis suggests the majority of cases have complex inheritance but rare families show apparent autosomal dominant inheritance. Two putative loci have been mapped (FEB1 and FEB2), but specific genes have not yet been identified. We recently described a clinical subset, termed generalized epilepsy with febrile seizures plus (GEFS+), in which many family members have seizures with fever that may persist beyond six years of age or be associated with afebrile generalized seizures. We now report linkage, in another large GEFS+ family, to chromosome region 19q13.1 and identification of a mutation in the voltage-gated sodium (Na+)-channel beta1 subunit gene (SCN1B). The mutation changes a conserved cysteine residue disrupting a putative disulfide bridge which normally maintains an extracellular immunoglobulin-like fold. Co-expression of the mutant beta1 subunit with a brain Na+-channel alpha subunit in Xenopus laevis oocytes demonstrates that the mutation interferes with the ability of the subunit to modulate channel-gating kinetics consistent with a loss-of-function allele. This observation develops the theme that idiopathic epilepsies are a family of channelopathies and raises the possibility of involvement of other Na+-channel subunit genes in febrile seizures and generalized epilepsies with complex inheritance patterns.
[ { "begin_idx": "641", "end_idx": "688", "entity_id": "C565809", "entity_type": "Disease", "text_name": "generalized epilepsy with febrile seizures plus" }, { "begin_idx": "318", "end_idx": "326", "entity_id": "D004827", "entity_type": "Disease", "text_name": "epilepsy" }, { "begin_idx": "1501", "end_idx": "1511", "entity_id": "D004827", "entity_type": "Disease", "text_name": "epilepsies" }, { "begin_idx": "21", "end_idx": "41", "entity_id": "D004829", "entity_type": "Disease", "text_name": "generalized epilepsy" }, { "begin_idx": "1645", "end_idx": "1667", "entity_id": "D004829", "entity_type": "Disease", "text_name": "generalized epilepsies" }, { "begin_idx": "746", "end_idx": "751", "entity_id": "D005334", "entity_type": "Disease", "text_name": "fever" }, { "begin_idx": "0", "end_idx": "16", "entity_id": "D012640", "entity_type": "Disease", "text_name": "Febrile seizures" }, { "begin_idx": "114", "end_idx": "130", "entity_id": "D012640", "entity_type": "Disease", "text_name": "Febrile seizures" }, { "begin_idx": "225", "end_idx": "232", "entity_id": "D012640", "entity_type": "Disease", "text_name": "seizure" }, { "begin_idx": "279", "end_idx": "295", "entity_id": "D012640", "entity_type": "Disease", "text_name": "febrile seizures" }, { "begin_idx": "332", "end_idx": "349", "entity_id": "D012640", "entity_type": "Disease", "text_name": "afebrile seizures" }, { "begin_idx": "732", "end_idx": "740", "entity_id": "D012640", "entity_type": "Disease", "text_name": "seizures" }, { "begin_idx": "836", "end_idx": "844", "entity_id": "D012640", "entity_type": "Disease", "text_name": "seizures" }, { "begin_idx": "1624", "end_idx": "1640", "entity_id": "D012640", "entity_type": "Disease", "text_name": "febrile seizures" }, { "begin_idx": "1528", "end_idx": "1543", "entity_id": "D053447", "entity_type": "Disease", "text_name": "channelopathies" }, { "begin_idx": "528", "end_idx": "532", "entity_id": "2233", "entity_type": "Gene", "text_name": "FEB1" }, { "begin_idx": "537", "end_idx": "541", "entity_id": "2234", "entity_type": "Gene", "text_name": "FEB2" }, { "begin_idx": "107", "end_idx": "112", "entity_id": "6324", "entity_type": "Gene", "text_name": "SCN1B" }, { "begin_idx": "1024", "end_idx": "1029", "entity_id": "6324", "entity_type": "Gene", "text_name": "SCN1B" } ]
[ "Yes", "No" ]
[ false, false ]
[ { "begin_idx": "107", "end_idx": "112", "entity_id": "6324", "entity_type": "Gene", "text_name": "SCN1B" }, { "begin_idx": "528", "end_idx": "532", "entity_id": "2233", "entity_type": "Gene", "text_name": "FEB1" } ]
[ { "begin_idx": "641", "end_idx": "688", "entity_id": "C565809", "entity_type": "Disease", "text_name": "generalized epilepsy with febrile seizures plus" }, { "begin_idx": "114", "end_idx": "130", "entity_id": "D012640", "entity_type": "Disease", "text_name": "Febrile seizures" } ]
[ "SCN1B", "FEB1" ]
[ "generalized epilepsy with febrile seizures plus", "Febrile seizures" ]
9702742
Homozygosity at the dopamine D3 receptor gene is associated with opiate dependence.
Anatomical, pharmacological and human post-mortem studies suggest the dopamine D3 receptor (DRD3) gene as a candidate for drug dependence. We thus performed an association study of the Bal I polymorphism at the DRD3 gene, including 54 opiate addicts and 70 controls. Opiate addicts had a higher sensation-seeking score (on the Z ckerman scale) than controls (P = 0.001), particularly a subgroup (70%) who had a distinctly higher score, exceeding 24. There were no marked differences in genotypes between patients as a whole and controls. However, patients with a sensation-seeking score above 24 were more frequently homozygotes for both alleles than patients with a sensation-seeking score under 24 (P = 0.038) or controls (P = 0.034). Although obtained in a sample of limited size, these results suggest that the DRD3 gene may have a role in drug dependence susceptibility in individuals with high sensation-seeking scores. This hypothesis is consistent with the role of DRD3 in mediating responses to drugs of abuse in animals and the association of homozygosity at the Bal I polymorphism with drug abuse in schizophrenic patients (see companion article by Krebs et al).
[ { "begin_idx": "65", "end_idx": "82", "entity_id": "D009293", "entity_type": "Disease", "text_name": "opiate dependence" }, { "begin_idx": "1196", "end_idx": "1209", "entity_id": "D012559", "entity_type": "Disease", "text_name": "schizophrenic" }, { "begin_idx": "206", "end_idx": "221", "entity_id": "D019966", "entity_type": "Disease", "text_name": "drug dependence" }, { "begin_idx": "929", "end_idx": "944", "entity_id": "D019966", "entity_type": "Disease", "text_name": "drug dependence" }, { "begin_idx": "1182", "end_idx": "1192", "entity_id": "D019966", "entity_type": "Disease", "text_name": "drug abuse" }, { "begin_idx": "176", "end_idx": "180", "entity_id": "1814", "entity_type": "Gene", "text_name": "DRD3" }, { "begin_idx": "295", "end_idx": "299", "entity_id": "1814", "entity_type": "Gene", "text_name": "DRD3" }, { "begin_idx": "900", "end_idx": "904", "entity_id": "1814", "entity_type": "Gene", "text_name": "DRD3" }, { "begin_idx": "1058", "end_idx": "1062", "entity_id": "1814", "entity_type": "Gene", "text_name": "DRD3" } ]
[ "Yes", "No" ]
[ false, true ]
[ { "begin_idx": "176", "end_idx": "180", "entity_id": "1814", "entity_type": "Gene", "text_name": "DRD3" }, { "begin_idx": "900", "end_idx": "904", "entity_id": "1814", "entity_type": "Gene", "text_name": "DRD3" } ]
[ { "begin_idx": "65", "end_idx": "82", "entity_id": "D009293", "entity_type": "Disease", "text_name": "opiate dependence" }, { "begin_idx": "929", "end_idx": "944", "entity_id": "D019966", "entity_type": "Disease", "text_name": "drug dependence" } ]
[ "DRD3", "DRD3" ]
[ "opiate dependence", "drug dependence" ]
9713438
Association between angiotensin II receptor gene polymorphism and serum angiotensin converting enzyme (SACE) activity in patients with sarcoidosis.
[ { "begin_idx": "135", "end_idx": "146", "entity_id": "D012507", "entity_type": "Disease", "text_name": "sarcoidosis" }, { "begin_idx": "72", "end_idx": "101", "entity_id": "1636", "entity_type": "Gene", "text_name": "angiotensin converting enzyme" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "72", "end_idx": "101", "entity_id": "1636", "entity_type": "Gene", "text_name": "angiotensin converting enzyme" } ]
[ { "begin_idx": "135", "end_idx": "146", "entity_id": "D012507", "entity_type": "Disease", "text_name": "sarcoidosis" } ]
[ "angiotensin converting enzyme" ]
[ "sarcoidosis" ]
9716139
Association of the platelet glycoprotein IIIa PlA1/A2 gene polymorphism to coronary artery disease but not to nonfatal myocardial infarction in low risk patients.
BACKGROUND: The platelet membrane glycoprotein IIb/IIIa functions as a receptor for fibrinogen and von Willebrand factor during platelet aggregation. In a small case-control study, evidence has been presented that the PlA2 allele of the platelet glycoprotein GPIIIa PlA/A2 gene polymorphism might be an independent risk factor for acute myocardial infarction (MI). METHODS AND RESULTS: We explored the association of the PlA1A2 to the severity of coronary artery disease (CAD), as assessed angiographically in 2252 male individuals, and to myocardial infarction (MI). The severity of coronary heart disease (CHD) was also estimated by calculating a CHD score according to Gensini. The PlA genotype was determined by allele specific restriction digestion. Relation of the PlA2 allele to CAD: In the total population, the frequency of the PlA2 allele was not associated to the presence or to the extent of CAD. Also the CHD scores of PlA1/PlA2 genotypes were essentially the same. However, after exclusion of individuals with high BMI (> or =26.9 kg/m2) and/or low apoAI (< 1.43 g/l) PlA2PlA2 carriers had clearly higher CHD scores than PlA1PlA1 genotypes: PlA1PlA2 heterozygotes had intermediate values (p <0.05). After division of the study population into one group of individuals without any angiographic signs of CAD (CHD score = 0) and into another group of patients with severe CAD (CHD score (> or = 120), a strong association of the PlA2 allele with severe CAD was also found in the same low risk groups: e.g. exclusion of persons with high BMI and low apoAI resulted in an Odds ratio of 5.37 (1.46-19.7) (p <0.02). Relation of the PlA2 allele to MI: No association was found between PlA1/PlA2 genotypes and risk of MI neither in the total population nor in low risk subgroups. CONCLUSIONS: Whereas no difference in the distribution of allele and genotype frequencies between controls and survivors of MI could be detected, the PlA2 allele is associated with CHD in low risk patients.
[ { "begin_idx": "291", "end_idx": "311", "entity_id": "D001791", "entity_type": "Disease", "text_name": "platelet aggregation" }, { "begin_idx": "75", "end_idx": "98", "entity_id": "D003324", "entity_type": "Disease", "text_name": "coronary artery disease" }, { "begin_idx": "610", "end_idx": "633", "entity_id": "D003324", "entity_type": "Disease", "text_name": "coronary artery disease" }, { "begin_idx": "635", "end_idx": "638", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "949", "end_idx": "952", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "1067", "end_idx": "1070", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "1479", "end_idx": "1482", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "1546", "end_idx": "1549", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "1627", "end_idx": "1630", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" }, { "begin_idx": "747", "end_idx": "769", "entity_id": "D003327", "entity_type": "Disease", "text_name": "coronary heart disease" }, { "begin_idx": "771", "end_idx": "774", "entity_id": "D003327", "entity_type": "Disease", "text_name": "CHD" }, { "begin_idx": "812", "end_idx": "815", "entity_id": "D003327", "entity_type": "Disease", "text_name": "CHD" }, { "begin_idx": "1081", "end_idx": "1084", "entity_id": "D003327", "entity_type": "Disease", "text_name": "CHD" }, { "begin_idx": "1282", "end_idx": "1285", "entity_id": "D003327", "entity_type": "Disease", "text_name": "CHD" }, { "begin_idx": "1484", "end_idx": "1487", "entity_id": "D003327", "entity_type": "Disease", "text_name": "CHD" }, { "begin_idx": "1551", "end_idx": "1554", "entity_id": "D003327", "entity_type": "Disease", "text_name": "CHD" }, { "begin_idx": "2129", "end_idx": "2132", "entity_id": "D003327", "entity_type": "Disease", "text_name": "CHD" }, { "begin_idx": "119", "end_idx": "140", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "500", "end_idx": "521", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "523", "end_idx": "525", "entity_id": "D009203", "entity_type": "Disease", "text_name": "MI" }, { "begin_idx": "703", "end_idx": "724", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "726", "end_idx": "728", "entity_id": "D009203", "entity_type": "Disease", "text_name": "MI" }, { "begin_idx": "1817", "end_idx": "1819", "entity_id": "D009203", "entity_type": "Disease", "text_name": "MI" }, { "begin_idx": "1886", "end_idx": "1888", "entity_id": "D009203", "entity_type": "Disease", "text_name": "MI" }, { "begin_idx": "2072", "end_idx": "2074", "entity_id": "D009203", "entity_type": "Disease", "text_name": "MI" }, { "begin_idx": "262", "end_idx": "276", "entity_id": "D014842", "entity_type": "Disease", "text_name": "von Willebrand" }, { "begin_idx": "247", "end_idx": "257", "entity_id": "2244", "entity_type": "Gene", "text_name": "fibrinogen" }, { "begin_idx": "46", "end_idx": "53", "entity_id": "25833", "entity_type": "Gene", "text_name": "PlA1/A2" }, { "begin_idx": "1095", "end_idx": "1099", "entity_id": "25833", "entity_type": "Gene", "text_name": "PlA1" }, { "begin_idx": "1854", "end_idx": "1858", "entity_id": "25833", "entity_type": "Gene", "text_name": "PlA1" }, { "begin_idx": "179", "end_idx": "213", "entity_id": "3674", "entity_type": "Gene", "text_name": "platelet membrane glycoprotein IIb" }, { "begin_idx": "422", "end_idx": "428", "entity_id": "3690", "entity_type": "Gene", "text_name": "GPIIIa" }, { "begin_idx": "381", "end_idx": "385", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" }, { "begin_idx": "934", "end_idx": "938", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" }, { "begin_idx": "1000", "end_idx": "1004", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" }, { "begin_idx": "1100", "end_idx": "1104", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" }, { "begin_idx": "1603", "end_idx": "1607", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" }, { "begin_idx": "1802", "end_idx": "1806", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" }, { "begin_idx": "1859", "end_idx": "1863", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" }, { "begin_idx": "2098", "end_idx": "2102", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "422", "end_idx": "428", "entity_id": "3690", "entity_type": "Gene", "text_name": "GPIIIa" }, { "begin_idx": "1603", "end_idx": "1607", "entity_id": "5320", "entity_type": "Gene", "text_name": "PlA2" } ]
[ { "begin_idx": "119", "end_idx": "140", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "1479", "end_idx": "1482", "entity_id": "D003324", "entity_type": "Disease", "text_name": "CAD" } ]
[ "GPIIIa", "PlA2" ]
[ "myocardial infarction", "CAD" ]
9718352
Dihydropyrimidinase deficiency: structural organization, chromosomal localization, and mutation analysis of the human dihydropyrimidinase gene.
Dihydropyrimidinase (DHP) deficiency (MIM 222748) is characterized by dihydropyrimidinuria and is associated with a variable clinical phenotype. This disease might be associated with a risk of 5-fluorouracil toxicity, although no cases have been reported. We present here both the molecular characterization of the human DHP gene and, for the first time, the mutations causing DHP deficiency. The human DHP gene spans >80 kb and consists of 10 exons. It has been assigned to 8q22, by FISH. We performed mutation analysis of genomic DNA in one symptomatic and five asymptomatic individuals presenting with dihydropyrimidinuria. We identified one frameshift mutation and five missense mutations. Two related Japanese adult subjects were homozygous for the Q334R substitution, whereas two other, unrelated Japanese infant subjects were heterozygous for the same mutation, but this mutation is not common in the Japanese population. A Caucasian pediatric patient exhibiting epileptic attacks, dysmorphic features, and severe developmental delay was homozygous for W360R. Using a eukaryotic expression system, we showed that all mutations reduced enzyme activity significantly, indicating that these are crucial DHP deficiency-causing mutations. There was no significant difference, in residual activity, between mutations observed in the symptomatic and those observed in the asymptomatic individuals.
[ { "begin_idx": "0", "end_idx": "30", "entity_id": "C562815", "entity_type": "Disease", "text_name": "Dihydropyrimidinase deficiency" }, { "begin_idx": "144", "end_idx": "180", "entity_id": "C562815", "entity_type": "Disease", "text_name": "Dihydropyrimidinase (DHP) deficiency" }, { "begin_idx": "521", "end_idx": "535", "entity_id": "C562815", "entity_type": "Disease", "text_name": "DHP deficiency" }, { "begin_idx": "1351", "end_idx": "1365", "entity_id": "C562815", "entity_type": "Disease", "text_name": "DHP deficiency" }, { "begin_idx": "1133", "end_idx": "1152", "entity_id": "D000013", "entity_type": "Disease", "text_name": "dysmorphic features" }, { "begin_idx": "1165", "end_idx": "1184", "entity_id": "D002658", "entity_type": "Disease", "text_name": "developmental delay" }, { "begin_idx": "1114", "end_idx": "1131", "entity_id": "D004827", "entity_type": "Disease", "text_name": "epileptic attacks" }, { "begin_idx": "214", "end_idx": "234", "entity_id": "D054067", "entity_type": "Disease", "text_name": "dihydropyrimidinuria" }, { "begin_idx": "749", "end_idx": "769", "entity_id": "D054067", "entity_type": "Disease", "text_name": "dihydropyrimidinuria" }, { "begin_idx": "352", "end_idx": "360", "entity_id": "D064420", "entity_type": "Disease", "text_name": "toxicity" }, { "begin_idx": "118", "end_idx": "137", "entity_id": "1807", "entity_type": "Gene", "text_name": "dihydropyrimidinase" }, { "begin_idx": "165", "end_idx": "168", "entity_id": "1807", "entity_type": "Gene", "text_name": "DHP" }, { "begin_idx": "465", "end_idx": "468", "entity_id": "1807", "entity_type": "Gene", "text_name": "DHP" }, { "begin_idx": "547", "end_idx": "550", "entity_id": "1807", "entity_type": "Gene", "text_name": "DHP" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "118", "end_idx": "137", "entity_id": "1807", "entity_type": "Gene", "text_name": "dihydropyrimidinase" }, { "begin_idx": "547", "end_idx": "550", "entity_id": "1807", "entity_type": "Gene", "text_name": "DHP" } ]
[ { "begin_idx": "144", "end_idx": "180", "entity_id": "C562815", "entity_type": "Disease", "text_name": "Dihydropyrimidinase (DHP) deficiency" }, { "begin_idx": "352", "end_idx": "360", "entity_id": "D064420", "entity_type": "Disease", "text_name": "toxicity" } ]
[ "dihydropyrimidinase", "DHP" ]
[ "Dihydropyrimidinase (DHP) deficiency", "toxicity" ]
9730698
Angiotensin-converting enzyme insertion/deletion polymorphism and diabetic albuminuria in patients with NIDDM followed Up for 9 years.
Nephropathy is a major cause of premature morbidity and mortality in patients with non-insulin-dependent diabetes mellitus (NIDDM). The insertion/deletion (I/D) polymorphism of angiotensin-converting enzyme (ACE) is a genetic determinant of plasma ACE levels. Recent studies have found I/D polymorphism of the ACE gene to be associated with nephropathy in NIDDM. This association has not been evaluated in prospective studies. We, therefore, studied the relationship between ACE gene I/D polymorphism and diabetic albuminuria and glomerular filtration rate (GFR) in 83 NIDDM patients followed up for 9 years. At baseline, 29% (24 of 83) of the diabetic patients had an increased (>30 mg/24 h) urinary albumin excretion rate (UAER) and the prevalence of albuminuria at the 9-year examination was 35% (29 of 83). During the follow-up period, systolic blood pressure (p = 0.044), prevalence of hypertension (p < 0.01), and fasting blood glucose levels (p < 0.01) increased, while high-density lipoprotein cholesterol (p < 0.01) decreased. The declines of GFR during the follow-up period were 8.5, 14.1, and 16.3% within genotype groups of II, ID, and DD, respectively (p values for decreases: NS for II, <0.001 for ID, and <0.001 for DD). Patients with the DD genotype tended to have a steeper decrease of GFR, but the change was not statistically significant between the genotype groups. The increases of UAER during the follow-up period were 35.1, 8.3, and 122.4% within genotype groups of II, ID, and DD, respectively, but p values for all increases were not significant. Parallel to GFR, patients with the DD genotype tended to have a steeper increase of UAER, but the change was not statistically significant between the genotype groups. There were no differences in the ACE genotype distribution and allele frequencies between the patients with or without albuminuria either at follow-up or in cross-sectional settings. In conclusion, this 9-year follow-up study does not support the hypothesis that the ACE I/D polymorphism is a major genetic marker of diabetic nephropathy in NIDDM patients.
[ { "begin_idx": "1283", "end_idx": "1285", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "1366", "end_idx": "1368", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "1389", "end_idx": "1391", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "1636", "end_idx": "1638", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "1742", "end_idx": "1744", "entity_id": "C536170", "entity_type": "Disease", "text_name": "DD" }, { "begin_idx": "75", "end_idx": "86", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "649", "end_idx": "660", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "888", "end_idx": "899", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "1994", "end_idx": "2005", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" }, { "begin_idx": "66", "end_idx": "74", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "640", "end_idx": "648", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "779", "end_idx": "787", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetic" }, { "begin_idx": "104", "end_idx": "109", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "218", "end_idx": "257", "entity_id": "D003924", "entity_type": "Disease", "text_name": "non-insulin-dependent diabetes mellitus" }, { "begin_idx": "259", "end_idx": "264", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "491", "end_idx": "496", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "704", "end_idx": "709", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "2216", "end_idx": "2221", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "2192", "end_idx": "2212", "entity_id": "D003928", "entity_type": "Disease", "text_name": "diabetic nephropathy" }, { "begin_idx": "1026", "end_idx": "1038", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "135", "end_idx": "146", "entity_id": "D007674", "entity_type": "Disease", "text_name": "Nephropathy" }, { "begin_idx": "476", "end_idx": "487", "entity_id": "D007674", "entity_type": "Disease", "text_name": "nephropathy" }, { "begin_idx": "343", "end_idx": "346", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "383", "end_idx": "386", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "445", "end_idx": "448", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "610", "end_idx": "613", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "1908", "end_idx": "1911", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "2142", "end_idx": "2145", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "343", "end_idx": "346", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" }, { "begin_idx": "383", "end_idx": "386", "entity_id": "1636", "entity_type": "Gene", "text_name": "ACE" } ]
[ { "begin_idx": "218", "end_idx": "257", "entity_id": "D003924", "entity_type": "Disease", "text_name": "non-insulin-dependent diabetes mellitus" }, { "begin_idx": "888", "end_idx": "899", "entity_id": "D000419", "entity_type": "Disease", "text_name": "albuminuria" } ]
[ "ACE", "ACE" ]
[ "non-insulin-dependent diabetes mellitus", "albuminuria" ]
9736777
ClC-1 chloride channel mutations in myotonia congenita: variable penetrance of mutations shifting the voltage dependence.
Mutations in the ClC-1 muscle chloride channel cause either recessive or dominant myotonia congenita. Using a systematic screening procedure, we have now identified four novel missense mutations in dominant (V286A, F307S) and recessive myotonia (V236L, G285E), and have analysed the effect of these and other recently described mutations (A313T, I556N) on channel properties in the Xenopus oocyte expression system. Mutations V286A, F307S and A313T displayed a 'classical' dominant phenotype: their voltage dependence was shifted towards positive potentials and displayed a dominant-negative effect by significantly imparting a voltage shift on mutant-wild-type heteromeric channels as found in heterozygous patients. In contrast, the recessive mutation V236L also shifted the voltage dependence to positive values, but co-expression with wild-type ClC-1 gave almost wild-type currents. I556N, a mutation found in patients with benign dominant myotonia, drastically shifts the voltage dependence, but only a slight shift is seen when co-expressed with wild-type ClC-1. Thus, the voltage dependence of mutant heteromeric channels is not always intermediate between those of the constituent homomeric channel subunits, a conclusion further supported by mixing different ClC-1 mutants. These complex interactions correlate clinically with various inheritance patterns, ranging from autosomal dominant with various degrees of penetrance to autosomal recessive.
[ { "begin_idx": "348", "end_idx": "366", "entity_id": "D009222", "entity_type": "Disease", "text_name": "recessive myotonia" }, { "begin_idx": "1050", "end_idx": "1074", "entity_id": "D009222", "entity_type": "Disease", "text_name": "benign dominant myotonia" }, { "begin_idx": "36", "end_idx": "54", "entity_id": "D009224", "entity_type": "Disease", "text_name": "myotonia congenita" }, { "begin_idx": "182", "end_idx": "222", "entity_id": "D009224", "entity_type": "Disease", "text_name": "recessive or dominant myotonia congenita" }, { "begin_idx": "0", "end_idx": "5", "entity_id": "1180", "entity_type": "Gene", "text_name": "ClC-1" }, { "begin_idx": "139", "end_idx": "144", "entity_id": "1180", "entity_type": "Gene", "text_name": "ClC-1" }, { "begin_idx": "971", "end_idx": "976", "entity_id": "1180", "entity_type": "Gene", "text_name": "ClC-1" }, { "begin_idx": "1184", "end_idx": "1189", "entity_id": "1180", "entity_type": "Gene", "text_name": "ClC-1" }, { "begin_idx": "1390", "end_idx": "1395", "entity_id": "1180", "entity_type": "Gene", "text_name": "ClC-1" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "0", "end_idx": "5", "entity_id": "1180", "entity_type": "Gene", "text_name": "ClC-1" }, { "begin_idx": "0", "end_idx": "5", "entity_id": "1180", "entity_type": "Gene", "text_name": "ClC-1" } ]
[ { "begin_idx": "182", "end_idx": "222", "entity_id": "D009224", "entity_type": "Disease", "text_name": "recessive or dominant myotonia congenita" }, { "begin_idx": "1050", "end_idx": "1074", "entity_id": "D009222", "entity_type": "Disease", "text_name": "benign dominant myotonia" } ]
[ "ClC-1", "ClC-1" ]
[ "recessive or dominant myotonia congenita", "benign dominant myotonia" ]
9740618
Association between a polymorphism in the G protein beta3 subunit gene and lower renin and elevated diastolic blood pressure levels.
Gi proteins mediate the intracellular effects of many vasoactive and proliferative stimuli. Recently such signaling was found to be enhanced in cultured cells of some hypertensive subjects. A polymorphism at position 825 (C-->T) of the G protein beta3 subunit gene (GNB3) was strictly related to this phenotype. The aim of the present investigation was to test the association between this polymorphism and blood pressure and plasma renin levels in humans. A population-based sample (n=608) was analyzed by questionnaire and characterized for blood pressure; plasma renin, prorenin, and aldosterone levels; and Gbeta3 C825T allele status. In individuals without antihypertensive medication (n=474; age range, 52 to 67 years), the polymorphism was mildly associated with diastolic blood pressure (CC: 88.6+/-0.3 mm Hg, n=218; versus CT: 90.1+/-0.7 mm Hg, n=209; versus TT: 91.8+/-1.7 mm Hg, n=47; P=0.02 for trend) but not with systolic blood pressure. Furthermore, the 825T allele was also significantly associated with lower renin and prorenin levels, whereas the aldosterone to renin ratio was elevated in these subjects. Significant associations between the 825T allele and diastolic blood pressure, plasma renin, and prorenin levels (inverse), and the aldosterone to renin ratio persisted after adjustment for age, gender, body mass index, and systolic blood pressure. Finally, when the entire sample was considered and an adjustment was made for covariates, the presence of arterial hypertension and the use of antihypertensive medication were both 1. 8-fold higher in the TT than in the CC genotype group (P<0.05 and P=0.06, respectively). This observation, if replicated in further studies, suggests a molecular mechanism that unifies a higher diastolic blood pressure, a lower renin level, and an elevated aldosterone to renin ratio, ie, a combination of features frequently found in patients with arterial hypertension.
[ { "begin_idx": "300", "end_idx": "312", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertensive" }, { "begin_idx": "1621", "end_idx": "1633", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "2048", "end_idx": "2060", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "42", "end_idx": "57", "entity_id": "2784", "entity_type": "Gene", "text_name": "G protein beta3" }, { "begin_idx": "369", "end_idx": "384", "entity_id": "2784", "entity_type": "Gene", "text_name": "G protein beta3" }, { "begin_idx": "399", "end_idx": "403", "entity_id": "2784", "entity_type": "Gene", "text_name": "GNB3" }, { "begin_idx": "81", "end_idx": "86", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" }, { "begin_idx": "566", "end_idx": "571", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" }, { "begin_idx": "699", "end_idx": "704", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" }, { "begin_idx": "1159", "end_idx": "1164", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" }, { "begin_idx": "1213", "end_idx": "1218", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" }, { "begin_idx": "1343", "end_idx": "1348", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" }, { "begin_idx": "1404", "end_idx": "1409", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" }, { "begin_idx": "1918", "end_idx": "1923", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" }, { "begin_idx": "1962", "end_idx": "1967", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" } ]
[ "Yes", "No" ]
[ false, true ]
[ { "begin_idx": "42", "end_idx": "57", "entity_id": "2784", "entity_type": "Gene", "text_name": "G protein beta3" }, { "begin_idx": "566", "end_idx": "571", "entity_id": "5972", "entity_type": "Gene", "text_name": "renin" } ]
[ { "begin_idx": "300", "end_idx": "312", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertensive" }, { "begin_idx": "2048", "end_idx": "2060", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" } ]
[ "G protein beta3", "renin" ]
[ "hypertensive", "hypertension" ]
9748691
160Thr mutation in the rhodopsin gene associated with retinitis pigmentosa.
Mutations in the rhodopsin gene were studied in 23 unrelated Spanish patients with sporadic retinitis pigmentosa (RP). A codon 160 Thr C-->A transition was found in 4 of the 23 patients vs. none of the 159 controls (p < 0.001) suggesting that this mutation may be an informative marker in RP.
[ { "begin_idx": "54", "end_idx": "74", "entity_id": "D012174", "entity_type": "Disease", "text_name": "retinitis pigmentosa" }, { "begin_idx": "159", "end_idx": "188", "entity_id": "D012174", "entity_type": "Disease", "text_name": "sporadic retinitis pigmentosa" }, { "begin_idx": "190", "end_idx": "192", "entity_id": "D012174", "entity_type": "Disease", "text_name": "RP" }, { "begin_idx": "365", "end_idx": "367", "entity_id": "D012174", "entity_type": "Disease", "text_name": "RP" }, { "begin_idx": "23", "end_idx": "32", "entity_id": "6010", "entity_type": "Gene", "text_name": "rhodopsin" }, { "begin_idx": "93", "end_idx": "102", "entity_id": "6010", "entity_type": "Gene", "text_name": "rhodopsin" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "23", "end_idx": "32", "entity_id": "6010", "entity_type": "Gene", "text_name": "rhodopsin" } ]
[ { "begin_idx": "159", "end_idx": "188", "entity_id": "D012174", "entity_type": "Disease", "text_name": "sporadic retinitis pigmentosa" } ]
[ "rhodopsin" ]
[ "sporadic retinitis pigmentosa" ]
9752691
[Aldehyde dehydrogenase 2(ALDH2) gene polymorphism in NIDDM patients with chronic renal failure].
In order to investigate the influence of aldehyde dehydrogenase 2(ALDH2) genotype in the pathogenesis of nephropathy due to non-insulin dependent diabetes mellitus (NIDDM), genotyping of ALDH2 was measured using the PCR-RFLP method in patients with NIDDM on chronic hemodialysis (HD). The results were as follows; 1) The frequency of active ALDH2 was 63% and that of inactive ALDH2 was 37%. 2) The percentage of active ALDH2 was significantly higher in patients with alcohol tolerance than that in those without it (38%). 3) The estimated amount of alcohol consumption in the past was 506 +/- 720 g/week in the active ALDH2 group, and 156 +/- 288 g/week in the inactive ALDH2 group, showing a significant difference between the two groups. 4) Interdialytic body weight gain was larger in patients with active ALDH2 than in those with inactive ALDH2. Since the frequency of active ALDH2 was similar to that in patients without nephropathy, these results do not support the hypothesis that ALDH2 gene polymorphism is involved in the development and persistence of chronic renal failure due to NIDDM. However, salt and water craving in dialysis patients may be influenced partially by an active ALDH2 gene.
[ { "begin_idx": "54", "end_idx": "59", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "222", "end_idx": "261", "entity_id": "D003924", "entity_type": "Disease", "text_name": "non-insulin dependent diabetes mellitus" }, { "begin_idx": "263", "end_idx": "268", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "347", "end_idx": "352", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "1189", "end_idx": "1194", "entity_id": "D003924", "entity_type": "Disease", "text_name": "NIDDM" }, { "begin_idx": "203", "end_idx": "214", "entity_id": "D007674", "entity_type": "Disease", "text_name": "nephropathy" }, { "begin_idx": "1024", "end_idx": "1035", "entity_id": "D007674", "entity_type": "Disease", "text_name": "nephropathy" }, { "begin_idx": "74", "end_idx": "95", "entity_id": "D051437", "entity_type": "Disease", "text_name": "chronic renal failure" }, { "begin_idx": "1160", "end_idx": "1181", "entity_id": "D051437", "entity_type": "Disease", "text_name": "chronic renal failure" }, { "begin_idx": "1", "end_idx": "25", "entity_id": "217", "entity_type": "Gene", "text_name": "Aldehyde dehydrogenase 2" }, { "begin_idx": "26", "end_idx": "31", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "139", "end_idx": "163", "entity_id": "217", "entity_type": "Gene", "text_name": "aldehyde dehydrogenase 2" }, { "begin_idx": "164", "end_idx": "169", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "285", "end_idx": "290", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "439", "end_idx": "444", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "474", "end_idx": "479", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "517", "end_idx": "522", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "716", "end_idx": "721", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "768", "end_idx": "773", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "907", "end_idx": "912", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "941", "end_idx": "946", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "978", "end_idx": "983", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "1086", "end_idx": "1091", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" }, { "begin_idx": "1290", "end_idx": "1295", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "1", "end_idx": "25", "entity_id": "217", "entity_type": "Gene", "text_name": "Aldehyde dehydrogenase 2" }, { "begin_idx": "164", "end_idx": "169", "entity_id": "217", "entity_type": "Gene", "text_name": "ALDH2" } ]
[ { "begin_idx": "222", "end_idx": "261", "entity_id": "D003924", "entity_type": "Disease", "text_name": "non-insulin dependent diabetes mellitus" }, { "begin_idx": "74", "end_idx": "95", "entity_id": "D051437", "entity_type": "Disease", "text_name": "chronic renal failure" } ]
[ "Aldehyde dehydrogenase 2", "ALDH2" ]
[ "non-insulin dependent diabetes mellitus", "chronic renal failure" ]
9756053
Association of alcohol or other drug dependence with alleles of the mu opioid receptor gene (OPRM1).
Opioidergic neurotransmission and, specifically, the mu opioid receptor have been implicated in the reinforcing effects of a variety of drugs of abuse. Consequently, the present study examined the association of a polymorphic (CA)n repeat at the OPRM1 locus (the gene coding for the mu opioid receptor) to alcohol or drug dependence in 320 Caucasian and 108 African-American substance-dependent or control subjects. Among Caucasians, suggestion of a modest association, which could be interpreted as statistically significant (p = 0.03), was observed between OPRM1 alleles and substance (alcohol, cocaine, or opioid) dependence. Analysis by specific substance showed only a trend level association to alcohol dependence. Comparisons among African Americans yielded no evidence for association. Further studies of the association between alleles of the OPRM1 gene and substance dependence appear warranted, particularly if they use a family-based approach to control for population stratification. Phenotypes other than a broad diagnostic categorization, such as opioid antagonist effects on drinking behavior in alcoholics, may provide more consistent evidence of a role for OPRM1 in behavioral variability.
[ { "begin_idx": "802", "end_idx": "820", "entity_id": "D000437", "entity_type": "Disease", "text_name": "alcohol dependence" }, { "begin_idx": "32", "end_idx": "47", "entity_id": "D019966", "entity_type": "Disease", "text_name": "drug dependence" }, { "begin_idx": "418", "end_idx": "433", "entity_id": "D019966", "entity_type": "Disease", "text_name": "drug dependence" }, { "begin_idx": "718", "end_idx": "728", "entity_id": "D019966", "entity_type": "Disease", "text_name": "dependence" }, { "begin_idx": "978", "end_idx": "988", "entity_id": "D019966", "entity_type": "Disease", "text_name": "dependence" }, { "begin_idx": "93", "end_idx": "98", "entity_id": "4988", "entity_type": "Gene", "text_name": "OPRM1" }, { "begin_idx": "347", "end_idx": "352", "entity_id": "4988", "entity_type": "Gene", "text_name": "OPRM1" }, { "begin_idx": "660", "end_idx": "665", "entity_id": "4988", "entity_type": "Gene", "text_name": "OPRM1" }, { "begin_idx": "953", "end_idx": "958", "entity_id": "4988", "entity_type": "Gene", "text_name": "OPRM1" }, { "begin_idx": "1276", "end_idx": "1281", "entity_id": "4988", "entity_type": "Gene", "text_name": "OPRM1" } ]
[ "Yes", "No" ]
[ false, true ]
[ { "begin_idx": "93", "end_idx": "98", "entity_id": "4988", "entity_type": "Gene", "text_name": "OPRM1" }, { "begin_idx": "1276", "end_idx": "1281", "entity_id": "4988", "entity_type": "Gene", "text_name": "OPRM1" } ]
[ { "begin_idx": "802", "end_idx": "820", "entity_id": "D000437", "entity_type": "Disease", "text_name": "alcohol dependence" }, { "begin_idx": "978", "end_idx": "988", "entity_id": "D019966", "entity_type": "Disease", "text_name": "dependence" } ]
[ "OPRM1", "OPRM1" ]
[ "alcohol dependence", "dependence" ]
9758611
Familial eosinophilia maps to the cytokine gene cluster on human chromosomal region 5q31-q33.
Familial eosinophilia (FE) is an autosomal dominant disorder characterized by peripheral hypereosinophilia of unidentifiable cause with or without other organ involvement. To localize the gene for FE, we performed a genomewide search in a large U.S. kindred, using 312 different polymorphic markers. Seventeen affected subjects, 28 unaffected bloodline relatives, and 8 spouses were genotyped. The initial linkage results from the genome scan provided evidence for linkage on chromosome 5q31-q33. Additional genotyping of genetic markers located in this specific region demonstrated significant evidence that the FE locus is situated between the chromosome 5q markers D5S642 and D5S816 (multipoint LOD score of 6.49). Notably, this region contains the cytokine gene cluster, which includes three genes-namely, those for interleukin (IL)-3, IL-5, and granulocyte/macrophage colony-stimulating factor (GM-CSF)-whose products play important roles in the development and proliferation of eosinophils. These three cytokine genes were screened for potential disease-specific mutations by resequencing of a subgroup of individuals from the present kindred. No functional sequence polymorphisms were found within the promoter, the exons, or the introns of any of these genes or within the IL-3/GM-CSF enhancer, suggesting that the primary defect in FE is not caused by a mutation in any one of these genes but, rather, is caused by another gene in the area.
[ { "begin_idx": "0", "end_idx": "21", "entity_id": "C562722", "entity_type": "Disease", "text_name": "Familial eosinophilia" }, { "begin_idx": "94", "end_idx": "115", "entity_id": "C562722", "entity_type": "Disease", "text_name": "Familial eosinophilia" }, { "begin_idx": "117", "end_idx": "119", "entity_id": "C562722", "entity_type": "Disease", "text_name": "FE" }, { "begin_idx": "291", "end_idx": "293", "entity_id": "C562722", "entity_type": "Disease", "text_name": "FE" }, { "begin_idx": "707", "end_idx": "709", "entity_id": "C562722", "entity_type": "Disease", "text_name": "FE" }, { "begin_idx": "1435", "end_idx": "1437", "entity_id": "C562722", "entity_type": "Disease", "text_name": "FE" }, { "begin_idx": "183", "end_idx": "200", "entity_id": "D004802", "entity_type": "Disease", "text_name": "hypereosinophilia" }, { "begin_idx": "127", "end_idx": "154", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal dominant disorder" }, { "begin_idx": "944", "end_idx": "992", "entity_id": "1437", "entity_type": "Gene", "text_name": "granulocyte/macrophage colony-stimulating factor" }, { "begin_idx": "994", "end_idx": "1000", "entity_id": "1437", "entity_type": "Gene", "text_name": "GM-CSF" }, { "begin_idx": "1380", "end_idx": "1386", "entity_id": "1437", "entity_type": "Gene", "text_name": "GM-CSF" }, { "begin_idx": "914", "end_idx": "932", "entity_id": "3562", "entity_type": "Gene", "text_name": "interleukin (IL)-3" }, { "begin_idx": "1375", "end_idx": "1379", "entity_id": "3562", "entity_type": "Gene", "text_name": "IL-3" }, { "begin_idx": "934", "end_idx": "938", "entity_id": "3567", "entity_type": "Gene", "text_name": "IL-5" } ]
[ "Yes", "No" ]
[ false, true ]
[ { "begin_idx": "934", "end_idx": "938", "entity_id": "3567", "entity_type": "Gene", "text_name": "IL-5" }, { "begin_idx": "944", "end_idx": "992", "entity_id": "1437", "entity_type": "Gene", "text_name": "granulocyte/macrophage colony-stimulating factor" } ]
[ { "begin_idx": "183", "end_idx": "200", "entity_id": "D004802", "entity_type": "Disease", "text_name": "hypereosinophilia" }, { "begin_idx": "117", "end_idx": "119", "entity_id": "C562722", "entity_type": "Disease", "text_name": "FE" } ]
[ "IL-5", "granulocyte/macrophage colony-stimulating factor" ]
[ "hypereosinophilia", "FE" ]
9758617
Mutation in the human acetylcholinesterase-associated collagen gene, COLQ, is responsible for congenital myasthenic syndrome with end-plate acetylcholinesterase deficiency (Type Ic).
Congenital myasthenic syndrome (CMS) with end-plate acetylcholinesterase (AChE) deficiency is a rare autosomal recessive disease, recently classified as CMS type Ic (CMS-Ic). It is characterized by onset in childhood, generalized weakness increased by exertion, refractoriness to anticholinesterase drugs, and morphological abnormalities of the neuromuscular junctions (NMJs). The collagen-tailed form of AChE, which is normally concentrated at NMJs, is composed of catalytic tetramers associated with a specific collagen, COLQ. In CMS-Ic patients, these collagen-tailed forms are often absent. We studied a large family comprising 11 siblings, 6 of whom are affected by a mild form of CMS-Ic. The muscles of the patients contained collagen-tailed AChE. We first excluded the ACHE gene (7q22) as potential culprit, by linkage analysis; then we mapped COLQ to chromosome 3p24.2. By analyzing 3p24.2 markers located close to the gene, we found that the six affected patients were homozygous for an interval of 14 cM between D3S1597 and D3S2338. We determined the COLQ coding sequence and found that the patients present a homozygous missense mutation, Y431S, in the conserved C-terminal domain of COLQ. This mutation is thought to disturb the attachment of collagen-tailed AChE to the NMJ, thus constituting the first genetic defect causing CMS-Ic.
[ { "begin_idx": "336", "end_idx": "347", "entity_id": "C566415", "entity_type": "Disease", "text_name": "CMS type Ic" }, { "begin_idx": "349", "end_idx": "355", "entity_id": "C566415", "entity_type": "Disease", "text_name": "CMS-Ic" }, { "begin_idx": "715", "end_idx": "721", "entity_id": "C566415", "entity_type": "Disease", "text_name": "CMS-Ic" }, { "begin_idx": "869", "end_idx": "875", "entity_id": "C566415", "entity_type": "Disease", "text_name": "CMS-Ic" }, { "begin_idx": "1522", "end_idx": "1528", "entity_id": "C566415", "entity_type": "Disease", "text_name": "CMS-Ic" }, { "begin_idx": "130", "end_idx": "171", "entity_id": "D007676", "entity_type": "Disease", "text_name": "end-plate acetylcholinesterase deficiency" }, { "begin_idx": "507", "end_idx": "551", "entity_id": "D009468", "entity_type": "Disease", "text_name": "abnormalities of the neuromuscular junctions" }, { "begin_idx": "94", "end_idx": "124", "entity_id": "D020294", "entity_type": "Disease", "text_name": "congenital myasthenic syndrome" }, { "begin_idx": "183", "end_idx": "213", "entity_id": "D020294", "entity_type": "Disease", "text_name": "Congenital myasthenic syndrome" }, { "begin_idx": "215", "end_idx": "218", "entity_id": "D020294", "entity_type": "Disease", "text_name": "CMS" }, { "begin_idx": "284", "end_idx": "311", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive disease" }, { "begin_idx": "1499", "end_idx": "1513", "entity_id": "D030342", "entity_type": "Disease", "text_name": "genetic defect" }, { "begin_idx": "235", "end_idx": "255", "entity_id": "43", "entity_type": "Gene", "text_name": "acetylcholinesterase" }, { "begin_idx": "257", "end_idx": "261", "entity_id": "43", "entity_type": "Gene", "text_name": "AChE" }, { "begin_idx": "588", "end_idx": "592", "entity_id": "43", "entity_type": "Gene", "text_name": "AChE" }, { "begin_idx": "931", "end_idx": "935", "entity_id": "43", "entity_type": "Gene", "text_name": "AChE" }, { "begin_idx": "959", "end_idx": "963", "entity_id": "43", "entity_type": "Gene", "text_name": "ACHE" }, { "begin_idx": "1454", "end_idx": "1458", "entity_id": "43", "entity_type": "Gene", "text_name": "AChE" }, { "begin_idx": "22", "end_idx": "62", "entity_id": "8292", "entity_type": "Gene", "text_name": "acetylcholinesterase-associated collagen" }, { "begin_idx": "69", "end_idx": "73", "entity_id": "8292", "entity_type": "Gene", "text_name": "COLQ" }, { "begin_idx": "706", "end_idx": "710", "entity_id": "8292", "entity_type": "Gene", "text_name": "COLQ" }, { "begin_idx": "1034", "end_idx": "1038", "entity_id": "8292", "entity_type": "Gene", "text_name": "COLQ" }, { "begin_idx": "1244", "end_idx": "1248", "entity_id": "8292", "entity_type": "Gene", "text_name": "COLQ" }, { "begin_idx": "1378", "end_idx": "1382", "entity_id": "8292", "entity_type": "Gene", "text_name": "COLQ" } ]
[ "Yes", "No" ]
[ false, true ]
[ { "begin_idx": "22", "end_idx": "62", "entity_id": "8292", "entity_type": "Gene", "text_name": "acetylcholinesterase-associated collagen" }, { "begin_idx": "1244", "end_idx": "1248", "entity_id": "8292", "entity_type": "Gene", "text_name": "COLQ" } ]
[ { "begin_idx": "336", "end_idx": "347", "entity_id": "C566415", "entity_type": "Disease", "text_name": "CMS type Ic" }, { "begin_idx": "183", "end_idx": "213", "entity_id": "D020294", "entity_type": "Disease", "text_name": "Congenital myasthenic syndrome" } ]
[ "acetylcholinesterase-associated collagen", "COLQ" ]
[ "CMS type Ic", "Congenital myasthenic syndrome" ]
9762601
Hunter disease in the Spanish population: molecular analysis in 31 families.
Mucopolysaccharidosis type II (Hunter disease) is an X-linked disorder due to deficiency of the lysosomal enzyme iduronate 2-sulphatase. Here we report an update of molecular studies in 31 Spanish families with Hunter disease. We found a total of 22 novel small mutations (7 reported previously by our group), and 4 large deletions or rearrangements. Particularly relevant are two mutations, one showing an alternatively spliced product although the normal splice site is conserved; the other mutation results in an amino acid change that most likely modifies regulation of expression of the IDS gene. Except for large gene alterations and for the G374sp mutation already described, we could not establish a clear phenotype-genotype correlation. Mutation G374sp is the point mutation most frequent in our population (10%) and is always associated with mild phenotype. Our molecular analyses carried out in a relatively large series of patients with Hunter disease contribute to the identification of new mutations and reinforce the conclusions drawn in other populations about the genotype-phenotype correlation and the gene distribution of mutations.
[ { "begin_idx": "0", "end_idx": "14", "entity_id": "D004194", "entity_type": "Disease", "text_name": "Hunter disease" }, { "begin_idx": "108", "end_idx": "122", "entity_id": "D004194", "entity_type": "Disease", "text_name": "Hunter disease" }, { "begin_idx": "288", "end_idx": "302", "entity_id": "D004194", "entity_type": "Disease", "text_name": "Hunter disease" }, { "begin_idx": "1026", "end_idx": "1040", "entity_id": "D004194", "entity_type": "Disease", "text_name": "Hunter disease" }, { "begin_idx": "77", "end_idx": "106", "entity_id": "D016532", "entity_type": "Disease", "text_name": "Mucopolysaccharidosis type II" }, { "begin_idx": "130", "end_idx": "147", "entity_id": "D040181", "entity_type": "Disease", "text_name": "X-linked disorder" }, { "begin_idx": "669", "end_idx": "672", "entity_id": "3423", "entity_type": "Gene", "text_name": "IDS" } ]
[ "Yes", "No" ]
[ false, false ]
[ { "begin_idx": "669", "end_idx": "672", "entity_id": "3423", "entity_type": "Gene", "text_name": "IDS" }, { "begin_idx": "669", "end_idx": "672", "entity_id": "3423", "entity_type": "Gene", "text_name": "IDS" } ]
[ { "begin_idx": "77", "end_idx": "106", "entity_id": "D016532", "entity_type": "Disease", "text_name": "Mucopolysaccharidosis type II" }, { "begin_idx": "288", "end_idx": "302", "entity_id": "D004194", "entity_type": "Disease", "text_name": "Hunter disease" } ]
[ "IDS", "IDS" ]
[ "Mucopolysaccharidosis type II", "Hunter disease" ]
9769329
The effect of novel polymorphisms in the interleukin-6 (IL-6) gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis.
During active disease, patients with systemic-onset juvenile chronic arthritis (S-JCA) demonstrate a rise and fall in serum interleukin-6 (IL-6) that parallels the classic quotidian fever. To investigate the possibility that this cytokine profile results from a difference in the control of IL-6 expression, we examined the 5' flanking region of the IL-6 gene for polymorphisms. A G/C polymorphism was detected at position -174. In a group of 383 healthy men and women from a general practice in North London, the frequency of the C allele was 0.403 (95% confidence interval 0.37-0.44). In comparison, 92 patients with S-JCA had a different overall genotype frequency, especially those with onset of disease at < 5 yr of age. This was mainly due to the statistically significant lower frequency of the CC genotype in this subgroup. When comparing constructs of the 5' flanking region (-550-+61 bp) in a luciferase reporter vector transiently transfected into HeLa cells, the -174C construct showed 0.624+/-0.15-fold lower expression than the -174G construct. After stimulation with LPS or IL-1, expression from the -174C construct did not significantly change after 24 h, whereas expression from the -174G construct increased by 2.35+/-0.10- and 3.60+/-0.26-fold, respectively, compared with the unstimulated level. Plasma levels of IL-6 were also measured in 102 of the healthy subjects, and the C allele was found to be associated with significantly lower levels of plasma IL-6. These results suggest that there is a genetically determined difference in the degree of the IL-6 response to stressful stimuli between individuals. The reduced frequency of the potentially protective CC genotype in young S-JCA patients may contribute to its pathogenesis. Similarly the individual's IL-6 genotype may be highly relevant in other conditions where IL-6 has been implicated, such as atherosclerosis.
[ { "begin_idx": "1262", "end_idx": "1265", "entity_id": "C536528", "entity_type": "Disease", "text_name": "LPS" }, { "begin_idx": "152", "end_idx": "178", "entity_id": "D001171", "entity_type": "Disease", "text_name": "juvenile chronic arthritis" }, { "begin_idx": "232", "end_idx": "258", "entity_id": "D001171", "entity_type": "Disease", "text_name": "juvenile chronic arthritis" }, { "begin_idx": "187", "end_idx": "201", "entity_id": "D004194", "entity_type": "Disease", "text_name": "active disease" }, { "begin_idx": "2058", "end_idx": "2073", "entity_id": "D050197", "entity_type": "Disease", "text_name": "atherosclerosis" }, { "begin_idx": "1269", "end_idx": "1273", "entity_id": "3552", "entity_type": "Gene", "text_name": "IL-1" }, { "begin_idx": "41", "end_idx": "54", "entity_id": "3569", "entity_type": "Gene", "text_name": "interleukin-6" }, { "begin_idx": "56", "end_idx": "60", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "70", "end_idx": "74", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "100", "end_idx": "104", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "304", "end_idx": "317", "entity_id": "3569", "entity_type": "Gene", "text_name": "interleukin-6" }, { "begin_idx": "319", "end_idx": "323", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "471", "end_idx": "475", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "530", "end_idx": "534", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "1513", "end_idx": "1517", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "1655", "end_idx": "1659", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "1754", "end_idx": "1758", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "1961", "end_idx": "1965", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" }, { "begin_idx": "2024", "end_idx": "2028", "entity_id": "3569", "entity_type": "Gene", "text_name": "IL-6" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "41", "end_idx": "54", "entity_id": "3569", "entity_type": "Gene", "text_name": "interleukin-6" }, { "begin_idx": "41", "end_idx": "54", "entity_id": "3569", "entity_type": "Gene", "text_name": "interleukin-6" } ]
[ { "begin_idx": "152", "end_idx": "178", "entity_id": "D001171", "entity_type": "Disease", "text_name": "juvenile chronic arthritis" }, { "begin_idx": "1262", "end_idx": "1265", "entity_id": "C536528", "entity_type": "Disease", "text_name": "LPS" } ]
[ "interleukin-6", "interleukin-6" ]
[ "juvenile chronic arthritis", "LPS" ]
9771706
A gene encoding a transmembrane protein is mutated in patients with diabetes mellitus and optic atrophy (Wolfram syndrome).
Wolfram syndrome (WFS; OMIM 222300) is an autosomal recessive neurodegenerative disorder defined by young-onset non-immune insulin-dependent diabetes mellitus and progressive optic atrophy. Linkage to markers on chromosome 4p was confirmed in five families. On the basis of meiotic recombinants and disease-associated haplotypes, the WFS gene was localized to a BAC/P1 contig of less than 250 kb. Mutations in a novel gene (WFS1) encoding a putative transmembrane protein were found in all affected individuals in six WFS families, and these mutations were associated with the disease phenotype. WFS1 appears to function in survival of islet beta-cells and neurons.
[ { "begin_idx": "68", "end_idx": "85", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetes mellitus" }, { "begin_idx": "247", "end_idx": "282", "entity_id": "D003922", "entity_type": "Disease", "text_name": "insulin-dependent diabetes mellitus" }, { "begin_idx": "90", "end_idx": "103", "entity_id": "D009896", "entity_type": "Disease", "text_name": "optic atrophy" }, { "begin_idx": "299", "end_idx": "312", "entity_id": "D009896", "entity_type": "Disease", "text_name": "optic atrophy" }, { "begin_idx": "105", "end_idx": "121", "entity_id": "D014929", "entity_type": "Disease", "text_name": "Wolfram syndrome" }, { "begin_idx": "124", "end_idx": "140", "entity_id": "D014929", "entity_type": "Disease", "text_name": "Wolfram syndrome" }, { "begin_idx": "142", "end_idx": "145", "entity_id": "D014929", "entity_type": "Disease", "text_name": "WFS" }, { "begin_idx": "458", "end_idx": "461", "entity_id": "D014929", "entity_type": "Disease", "text_name": "WFS" }, { "begin_idx": "642", "end_idx": "645", "entity_id": "D014929", "entity_type": "Disease", "text_name": "WFS" }, { "begin_idx": "166", "end_idx": "212", "entity_id": "D020271", "entity_type": "Disease", "text_name": "autosomal recessive neurodegenerative disorder" }, { "begin_idx": "548", "end_idx": "552", "entity_id": "7466", "entity_type": "Gene", "text_name": "WFS1" }, { "begin_idx": "720", "end_idx": "724", "entity_id": "7466", "entity_type": "Gene", "text_name": "WFS1" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "548", "end_idx": "552", "entity_id": "7466", "entity_type": "Gene", "text_name": "WFS1" }, { "begin_idx": "720", "end_idx": "724", "entity_id": "7466", "entity_type": "Gene", "text_name": "WFS1" } ]
[ { "begin_idx": "105", "end_idx": "121", "entity_id": "D014929", "entity_type": "Disease", "text_name": "Wolfram syndrome" }, { "begin_idx": "90", "end_idx": "103", "entity_id": "D009896", "entity_type": "Disease", "text_name": "optic atrophy" } ]
[ "WFS1", "WFS1" ]
[ "Wolfram syndrome", "optic atrophy" ]
9787075
Murine CASK is disrupted in a sex-linked cleft palate mouse mutant.
A transgenic mouse insertional mutant displayed the phenotype of altered cranial morphology with sex-linked cleft palate. We have cloned the disrupted genomic X-linked locus and report the identification of the mCASK gene. The gene is transcribed to produce two messages of 4.5 and 9.5 kb expressed during development and in adult tissues, particularly the brain. We describe the isolation of two differentially spliced mouse cDNAs from the locus (mCASK-A and mCASK-B). The mCASK-B cDNA probably represents the full-length product of the 4.5-kb transcript. The identical N-termini of the predicted encoded proteins (mCASK-A and -B) are highly homologous to Ca2+/calmodulin-dependent protein kinase II, while the deduced C-terminus of mCASK-B is highly homologous to a family of multidomain proteins containing a guanylate kinase motif, the MAGUK proteins. mCASK-B is a new member of an emerging family of genes in which the encoded proteins combine these domains, termed here, the CAMGUKs, including rat CASK, Caenorhabditis elegans lin-2, and Drosophila caki/camguk. The CAMGUKs are likely to be effectors in signal transduction as regulatory partners of transmembrane molecules, modulated by calcium and nucleotides. The transgene in this mutant mouse line integrated into an intron that bisects the encoded calmodulin-binding domain, a potentially important regulatory domain of the predicted protein, generating hybrid transcripts.
[ { "begin_idx": "30", "end_idx": "53", "entity_id": "D002972", "entity_type": "Disease", "text_name": "sex-linked cleft palate" }, { "begin_idx": "165", "end_idx": "188", "entity_id": "D002972", "entity_type": "Disease", "text_name": "sex-linked cleft palate" }, { "begin_idx": "7", "end_idx": "11", "entity_id": "8573", "entity_type": "Gene", "text_name": "CASK" }, { "begin_idx": "279", "end_idx": "284", "entity_id": "8573", "entity_type": "Gene", "text_name": "mCASK" }, { "begin_idx": "516", "end_idx": "521", "entity_id": "8573", "entity_type": "Gene", "text_name": "mCASK" }, { "begin_idx": "528", "end_idx": "533", "entity_id": "8573", "entity_type": "Gene", "text_name": "mCASK" }, { "begin_idx": "542", "end_idx": "547", "entity_id": "8573", "entity_type": "Gene", "text_name": "mCASK" }, { "begin_idx": "684", "end_idx": "689", "entity_id": "8573", "entity_type": "Gene", "text_name": "mCASK" }, { "begin_idx": "802", "end_idx": "807", "entity_id": "8573", "entity_type": "Gene", "text_name": "mCASK" }, { "begin_idx": "924", "end_idx": "929", "entity_id": "8573", "entity_type": "Gene", "text_name": "mCASK" }, { "begin_idx": "1072", "end_idx": "1076", "entity_id": "8573", "entity_type": "Gene", "text_name": "CASK" }, { "begin_idx": "1101", "end_idx": "1106", "entity_id": "8573", "entity_type": "Gene", "text_name": "lin-2" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "279", "end_idx": "284", "entity_id": "8573", "entity_type": "Gene", "text_name": "mCASK" } ]
[ { "begin_idx": "30", "end_idx": "53", "entity_id": "D002972", "entity_type": "Disease", "text_name": "sex-linked cleft palate" } ]
[ "mCASK" ]
[ "sex-linked cleft palate" ]
9788719
Analysis of the transactivation domain of the androgen receptor in patients with male infertility.
Genetic defects of the human androgen receptor (AR) can cause a wide spectrum of androgen insensitivity syndromes (AIS) in XY individuals ranging from phenotypic females, to defective spermatogenesis in otherwise normal males. We screened the non-polymorphic regions of exon 1, transactivation domain (TAD), of the AR gene in 153 subjects with varying degrees of defective spermatogenesis of unknown aetiology, and compared them to 100 healthy fertile controls. Three different single-strand conformation polymorphisms were detected and sequencing of the mutant fragments revealed three G-->A transitions in codons 210, 211 and 214. The first two mutations were polymorphisms and the transition in codon 211 was related to ethnic origin occurring in 10-15% of Indian or Middle-Eastern subjects, but not in the majority of Chinese. The third mutation resulted in a non-conservative glycine to arginine substitution at codon 214 (G214R) and was associated with approximately 20% lower transactivation capacity compared to the wild-type (WT). This study, the first screening of the AR TAD for subtle mutations, in a large group of males with defective spermatogenesis, has uncovered novel polymorphisms which may be useful in ethnic studies. Although a possible pathogenic mutation was uncovered, mutations of the nonpolymorphic portions of the TAD of the AR do not appear to have a major role in the aetiology of idiopathic male infertility.
[ { "begin_idx": "81", "end_idx": "97", "entity_id": "D007248", "entity_type": "Disease", "text_name": "male infertility" }, { "begin_idx": "1521", "end_idx": "1537", "entity_id": "D007248", "entity_type": "Disease", "text_name": "male infertility" }, { "begin_idx": "180", "end_idx": "212", "entity_id": "D013734", "entity_type": "Disease", "text_name": "androgen insensitivity syndromes" }, { "begin_idx": "214", "end_idx": "217", "entity_id": "D013734", "entity_type": "Disease", "text_name": "AIS" }, { "begin_idx": "1123", "end_idx": "1132", "entity_id": "D017827", "entity_type": "Disease", "text_name": "wild-type" }, { "begin_idx": "1134", "end_idx": "1136", "entity_id": "D017827", "entity_type": "Disease", "text_name": "WT" }, { "begin_idx": "46", "end_idx": "63", "entity_id": "367", "entity_type": "Gene", "text_name": "androgen receptor" }, { "begin_idx": "128", "end_idx": "145", "entity_id": "367", "entity_type": "Gene", "text_name": "androgen receptor" }, { "begin_idx": "147", "end_idx": "149", "entity_id": "367", "entity_type": "Gene", "text_name": "AR" }, { "begin_idx": "414", "end_idx": "416", "entity_id": "367", "entity_type": "Gene", "text_name": "AR" }, { "begin_idx": "1452", "end_idx": "1454", "entity_id": "367", "entity_type": "Gene", "text_name": "AR" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "46", "end_idx": "63", "entity_id": "367", "entity_type": "Gene", "text_name": "androgen receptor" }, { "begin_idx": "414", "end_idx": "416", "entity_id": "367", "entity_type": "Gene", "text_name": "AR" } ]
[ { "begin_idx": "180", "end_idx": "212", "entity_id": "D013734", "entity_type": "Disease", "text_name": "androgen insensitivity syndromes" }, { "begin_idx": "1521", "end_idx": "1537", "entity_id": "D007248", "entity_type": "Disease", "text_name": "male infertility" } ]
[ "androgen receptor", "AR" ]
[ "androgen insensitivity syndromes", "male infertility" ]
9788975
Inhibition of norepinephrine-induced cardiac hypertrophy in s100beta transgenic mice.
We have recently reported that the Ca2+-binding protein S100beta was induced in rat heart after infarction and forced expression of S100beta in neonatal rat cardiac myocyte cultures inhibited alpha1-adrenergic induction of beta myosin heavy chain (MHC) and skeletal alpha-actin (skACT). We now extend this work by showing that S100beta is induced in hearts of human subjects after myocardial infarction. Furthermore, to determine whether overexpression of S100beta was sufficient to inhibit in vivo hypertrophy, transgenic mice containing multiple copies of the human gene under the control of its own promoter, and CD1 control mice were treated with norepinephrine (NE) (1.5 mg/kg) or vehicle, intraperitoneally twice daily for 15 d. In CD1, NE produced an increase in left ventricular/body weight ratio, ventricular wall thickness, induction of skACT, atrial natriuretic factor, betaMHC, and downregulation of alphaMHC. In transgenic mice, NE induced S100beta transgene mRNA and protein, but provoked neither hypertrophy nor regulated cardiac-specific gene expression. NE induced hypertrophy in cultured CD1 but not S100beta transgenic myocytes, confirming that the effects of S100beta on cardiac mass reflected myocyte-specific responses. These transgenic studies complement in vitro data and support the hypothesis that S100beta acts as an intrinsic negative regulator of the myocardial hypertrophic response.
[ { "begin_idx": "37", "end_idx": "56", "entity_id": "D006332", "entity_type": "Disease", "text_name": "cardiac hypertrophy" }, { "begin_idx": "585", "end_idx": "596", "entity_id": "D006984", "entity_type": "Disease", "text_name": "hypertrophy" }, { "begin_idx": "1097", "end_idx": "1108", "entity_id": "D006984", "entity_type": "Disease", "text_name": "hypertrophy" }, { "begin_idx": "1168", "end_idx": "1179", "entity_id": "D006984", "entity_type": "Disease", "text_name": "hypertrophy" }, { "begin_idx": "1477", "end_idx": "1489", "entity_id": "D006984", "entity_type": "Disease", "text_name": "hypertrophic" }, { "begin_idx": "182", "end_idx": "192", "entity_id": "D007238", "entity_type": "Disease", "text_name": "infarction" }, { "begin_idx": "467", "end_idx": "488", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "824", "end_idx": "827", "entity_id": "910", "entity_type": "Gene", "text_name": "CD1" }, { "begin_idx": "1192", "end_idx": "1195", "entity_id": "910", "entity_type": "Gene", "text_name": "CD1" }, { "begin_idx": "309", "end_idx": "332", "entity_id": "4625", "entity_type": "Gene", "text_name": "beta myosin heavy chain" }, { "begin_idx": "60", "end_idx": "68", "entity_id": "6285", "entity_type": "Gene", "text_name": "s100beta" }, { "begin_idx": "142", "end_idx": "150", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" }, { "begin_idx": "218", "end_idx": "226", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" }, { "begin_idx": "413", "end_idx": "421", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" }, { "begin_idx": "542", "end_idx": "550", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" }, { "begin_idx": "1039", "end_idx": "1047", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" }, { "begin_idx": "1204", "end_idx": "1212", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" }, { "begin_idx": "1265", "end_idx": "1273", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" }, { "begin_idx": "1410", "end_idx": "1418", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" }, { "begin_idx": "702", "end_idx": "705", "entity_id": "910", "entity_type": "Gene", "text_name": "CD1" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "60", "end_idx": "68", "entity_id": "6285", "entity_type": "Gene", "text_name": "s100beta" }, { "begin_idx": "142", "end_idx": "150", "entity_id": "6285", "entity_type": "Gene", "text_name": "S100beta" } ]
[ { "begin_idx": "467", "end_idx": "488", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "182", "end_idx": "192", "entity_id": "D007238", "entity_type": "Disease", "text_name": "infarction" } ]
[ "s100beta", "S100beta" ]
[ "myocardial infarction", "infarction" ]
9792406
Ten novel mutations found in Aniridia.
Aniridia (AN) is a sight-threatening congenital ocular disorder characterized by iris hypoplasia, corneal pannus, foveal and optic nerve hypoplasia, cataract formation, and glaucoma. In two-thirds of the patients, AN is inherited in an autosomal dominant fashion with almost complete penetrance but variable expression. The remaining cases are sporadic. Aniridia has been shown to be associated with mutations in the PAX6 gene, located on chromosome 11p13, telomeric to the Wilms' tumor predisposition gene (WT1). This paper describes 14 mutations in the PAX6 gene in patients with AN. Among these 14 mutations, 10 have been unpublished until now. They result most probably in haploinsufficiency and consequently in a reduced protein level of functional PAX6 protein. The mutations reported here are scattered all over the gene, including the paired-box, the glycine-rich region, the homeobox, and the proline-serine-threonine (PST)-rich region.
[ { "begin_idx": "188", "end_idx": "196", "entity_id": "D002386", "entity_type": "Disease", "text_name": "cataract" }, { "begin_idx": "137", "end_idx": "151", "entity_id": "D003316", "entity_type": "Disease", "text_name": "corneal pannus" }, { "begin_idx": "212", "end_idx": "220", "entity_id": "D005901", "entity_type": "Disease", "text_name": "glaucoma" }, { "begin_idx": "120", "end_idx": "135", "entity_id": "D007499", "entity_type": "Disease", "text_name": "iris hypoplasia" }, { "begin_idx": "496", "end_idx": "525", "entity_id": "D009396", "entity_type": "Disease", "text_name": "telomeric to the Wilms' tumor" }, { "begin_idx": "164", "end_idx": "186", "entity_id": "D009901", "entity_type": "Disease", "text_name": "optic nerve hypoplasia" }, { "begin_idx": "29", "end_idx": "37", "entity_id": "D015783", "entity_type": "Disease", "text_name": "Aniridia" }, { "begin_idx": "39", "end_idx": "47", "entity_id": "D015783", "entity_type": "Disease", "text_name": "Aniridia" }, { "begin_idx": "49", "end_idx": "51", "entity_id": "D015783", "entity_type": "Disease", "text_name": "AN" }, { "begin_idx": "253", "end_idx": "255", "entity_id": "D015783", "entity_type": "Disease", "text_name": "AN" }, { "begin_idx": "393", "end_idx": "401", "entity_id": "D015783", "entity_type": "Disease", "text_name": "Aniridia" }, { "begin_idx": "621", "end_idx": "623", "entity_id": "D015783", "entity_type": "Disease", "text_name": "AN" }, { "begin_idx": "76", "end_idx": "102", "entity_id": "D015785", "entity_type": "Disease", "text_name": "congenital ocular disorder" }, { "begin_idx": "456", "end_idx": "460", "entity_id": "5080", "entity_type": "Gene", "text_name": "PAX6" }, { "begin_idx": "594", "end_idx": "598", "entity_id": "5080", "entity_type": "Gene", "text_name": "PAX6" }, { "begin_idx": "793", "end_idx": "797", "entity_id": "5080", "entity_type": "Gene", "text_name": "PAX6" }, { "begin_idx": "547", "end_idx": "550", "entity_id": "7490", "entity_type": "Gene", "text_name": "WT1" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "456", "end_idx": "460", "entity_id": "5080", "entity_type": "Gene", "text_name": "PAX6" }, { "begin_idx": "456", "end_idx": "460", "entity_id": "5080", "entity_type": "Gene", "text_name": "PAX6" } ]
[ { "begin_idx": "29", "end_idx": "37", "entity_id": "D015783", "entity_type": "Disease", "text_name": "Aniridia" }, { "begin_idx": "496", "end_idx": "525", "entity_id": "D009396", "entity_type": "Disease", "text_name": "telomeric to the Wilms' tumor" } ]
[ "PAX6", "PAX6" ]
[ "Aniridia", "telomeric to the Wilms' tumor" ]
9794433
Nicotinamide-adenine dinucleotide phosphate oxidase assembly and activation in EBV-transformed B lymphoblastoid cell lines of normal and chronic granulomatous disease patients.
This paper deals with the mechanisms of activation of NADPH oxidase investigated using EBV-transformed human B lymphoblastoid cell lines (B cells) from normal subjects and from patients affected by X-linked chronic granulomatous disease (CGD). The results reported are as follows. 1) In normal B cells, the NADPH oxidase components p67phox, p40phox, p22phox, and gp91phox were less expressed than in polymorphonuclear neutrophils. 2) In normal B cells stimulated with PMA, p47phox, p67phox, and p40phox translocated to the membranes as occurs in polymorphonuclear neutrophils. 3) In CGD, B cells expressing p22phox in the absence of gp91phox, p47phox, p67phox, and p40phox did not translocate to the membranes after stimulation with PMA. 4) In PMA-stimulated B cells from an X91+ CGD patient in which p22phox was normally expressed and gp91phox was present but lacked five amino acids, translocation of p47phox to the membranes was unaffected, but p67phox and p40phox were poorly translocated, and the production of O2- was greatly reduced with respect to that by normal B cells. Taken together, these findings indicate that 1) a low expression of some NADPH oxidase components may represent the molecular basis of the low production of O2- in B lymphocytes; 2) the cytosolic components of NADPH oxidase cannot bind to p22phox on the membranes in the absence of gp91phox; 3) p47phox can translocate to the membranes independently of p67phox and p40phox; and 4) gp91phox may have a role in mediating and/or stabilizing the binding of p67phox and p40phox to the membranes of activated cells.
[ { "begin_idx": "137", "end_idx": "166", "entity_id": "D006105", "entity_type": "Disease", "text_name": "chronic granulomatous disease" }, { "begin_idx": "375", "end_idx": "413", "entity_id": "D006105", "entity_type": "Disease", "text_name": "X-linked chronic granulomatous disease" }, { "begin_idx": "415", "end_idx": "418", "entity_id": "D006105", "entity_type": "Disease", "text_name": "CGD" }, { "begin_idx": "760", "end_idx": "763", "entity_id": "D006105", "entity_type": "Disease", "text_name": "CGD" }, { "begin_idx": "957", "end_idx": "960", "entity_id": "D006105", "entity_type": "Disease", "text_name": "CGD" }, { "begin_idx": "527", "end_idx": "534", "entity_id": "1535", "entity_type": "Gene", "text_name": "p22phox" }, { "begin_idx": "784", "end_idx": "791", "entity_id": "1535", "entity_type": "Gene", "text_name": "p22phox" }, { "begin_idx": "978", "end_idx": "985", "entity_id": "1535", "entity_type": "Gene", "text_name": "p22phox" }, { "begin_idx": "1496", "end_idx": "1503", "entity_id": "1535", "entity_type": "Gene", "text_name": "p22phox" }, { "begin_idx": "540", "end_idx": "548", "entity_id": "1536", "entity_type": "Gene", "text_name": "gp91phox" }, { "begin_idx": "810", "end_idx": "818", "entity_id": "1536", "entity_type": "Gene", "text_name": "gp91phox" }, { "begin_idx": "1013", "end_idx": "1021", "entity_id": "1536", "entity_type": "Gene", "text_name": "gp91phox" }, { "begin_idx": "1539", "end_idx": "1547", "entity_id": "1536", "entity_type": "Gene", "text_name": "gp91phox" }, { "begin_idx": "1638", "end_idx": "1646", "entity_id": "1536", "entity_type": "Gene", "text_name": "gp91phox" }, { "begin_idx": "1622", "end_idx": "1636", "entity_id": "201780", "entity_type": "Gene", "text_name": "p40phox; and 4" }, { "begin_idx": "509", "end_idx": "516", "entity_id": "4688", "entity_type": "Gene", "text_name": "p67phox" }, { "begin_idx": "659", "end_idx": "666", "entity_id": "4688", "entity_type": "Gene", "text_name": "p67phox" }, { "begin_idx": "829", "end_idx": "836", "entity_id": "4688", "entity_type": "Gene", "text_name": "p67phox" }, { "begin_idx": "1125", "end_idx": "1132", "entity_id": "4688", "entity_type": "Gene", "text_name": "p67phox" }, { "begin_idx": "1610", "end_idx": "1617", "entity_id": "4688", "entity_type": "Gene", "text_name": "p67phox" }, { "begin_idx": "1710", "end_idx": "1717", "entity_id": "4688", "entity_type": "Gene", "text_name": "p67phox" }, { "begin_idx": "518", "end_idx": "525", "entity_id": "4689", "entity_type": "Gene", "text_name": "p40phox" }, { "begin_idx": "672", "end_idx": "679", "entity_id": "4689", "entity_type": "Gene", "text_name": "p40phox" }, { "begin_idx": "842", "end_idx": "849", "entity_id": "4689", "entity_type": "Gene", "text_name": "p40phox" }, { "begin_idx": "1137", "end_idx": "1144", "entity_id": "4689", "entity_type": "Gene", "text_name": "p40phox" }, { "begin_idx": "1622", "end_idx": "1636", "entity_id": "4689", "entity_type": "Gene", "text_name": "p40phox; and 4" }, { "begin_idx": "1722", "end_idx": "1729", "entity_id": "4689", "entity_type": "Gene", "text_name": "p40phox" }, { "begin_idx": "0", "end_idx": "51", "entity_id": "50506", "entity_type": "Gene", "text_name": "Nicotinamide-adenine dinucleotide phosphate oxidase" }, { "begin_idx": "650", "end_idx": "657", "entity_id": "653361", "entity_type": "Gene", "text_name": "p47phox" }, { "begin_idx": "820", "end_idx": "827", "entity_id": "653361", "entity_type": "Gene", "text_name": "p47phox" }, { "begin_idx": "1080", "end_idx": "1087", "entity_id": "653361", "entity_type": "Gene", "text_name": "p47phox" }, { "begin_idx": "1552", "end_idx": "1559", "entity_id": "653361", "entity_type": "Gene", "text_name": "p47phox" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "540", "end_idx": "548", "entity_id": "1536", "entity_type": "Gene", "text_name": "gp91phox" }, { "begin_idx": "1722", "end_idx": "1729", "entity_id": "4689", "entity_type": "Gene", "text_name": "p40phox" } ]
[ { "begin_idx": "375", "end_idx": "413", "entity_id": "D006105", "entity_type": "Disease", "text_name": "X-linked chronic granulomatous disease" }, { "begin_idx": "415", "end_idx": "418", "entity_id": "D006105", "entity_type": "Disease", "text_name": "CGD" } ]
[ "gp91phox", "p40phox" ]
[ "X-linked chronic granulomatous disease", "CGD" ]
9799084
The human FE65 gene: genomic structure and an intronic biallelic polymorphism associated with sporadic dementia of the Alzheimer type.
The FE65 protein binds to the intracellular domain of the beta-amyloid precursor protein (betaPP) and may modulate the internalization of betaPP. This gene is highly expressed in regions of the brain specifically affected in dementia of the Alzheimer type (DAT). As a prelude to further investigations of the role of FE65 in the metabolism of betaPP and in the pathogenesis of DAT, we have determined the entire genomic structure and sequence of human FE65 and have discovered several polymorphisms in this gene. Human FE65 contains 14 exons ranging in size from 6 to 735 bp. All splice sites conform to consensus sequences except for the donor site of intron 10. The 5' end of FE65 mRNA was identified by rapid amplification of the cDNA 5' end and is 31 bp longer than the previously published cDNA sequence. The 5'-flanking region of this gene is TATA-less and is very GC-rich with at least five putative Sp1 binding sites. In comparison to the genomic rat FE65 sequence, the human FE65 5'-untranslated region is 134 bp longer and has an extra exon (exon 1, 86 bp). To identify mutations/polymorphisms of the coding regions of this gene, we performed blinded analysis of 457 Caucasian case-control samples from a large epidemiological study of sporadic DAT. Screening was conducted by single-strand conformation polymorphism. Four minor variants were found within the coding region, with frequencies between 0.002 and 0.015; two of the four result in amino acid substitutions. The more informative biallelic polymorphism (a trinucleotide deletion and a single base substitution) was found within intron 13 (84 bp), which interrupts two exons encoding the betaPP binding site. The frequency of the minor allele in this intron was 0.097 in DAT cases and 0.161 in controls (chi2=7.78, P=0.0054). Having at least one copy of the minor allele was associated with a decreased risk for DAT (chi2=9.20, P<0.005, odds ratio=0.49, 95% CI 0.31-0.77). Multivariate analysis showed that this association was independent of the APOE genotype. These results suggest that either FE65 itself or a closely linked gene influences the pathogenesis of sporadic DAT. The interaction of FE65 with betaPP and the association of a FE65 polymorphism with DAT lend credence to the hypothesis that the metabolism of betaPP is central to the pathogenesis of common sporadic forms of DAT.
[ { "begin_idx": "119", "end_idx": "133", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer type" }, { "begin_idx": "376", "end_idx": "390", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer type" }, { "begin_idx": "94", "end_idx": "111", "entity_id": "D003704", "entity_type": "Disease", "text_name": "sporadic dementia" }, { "begin_idx": "360", "end_idx": "368", "entity_id": "D003704", "entity_type": "Disease", "text_name": "dementia" }, { "begin_idx": "10", "end_idx": "14", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "139", "end_idx": "143", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "452", "end_idx": "456", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "587", "end_idx": "591", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "654", "end_idx": "658", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "813", "end_idx": "817", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "1094", "end_idx": "1098", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "1119", "end_idx": "1123", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "2200", "end_idx": "2204", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "2301", "end_idx": "2305", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "2343", "end_idx": "2347", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "2151", "end_idx": "2155", "entity_id": "348", "entity_type": "Gene", "text_name": "APOE" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "10", "end_idx": "14", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" }, { "begin_idx": "2301", "end_idx": "2305", "entity_id": "322", "entity_type": "Gene", "text_name": "FE65" } ]
[ { "begin_idx": "119", "end_idx": "133", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer type" }, { "begin_idx": "94", "end_idx": "111", "entity_id": "D003704", "entity_type": "Disease", "text_name": "sporadic dementia" } ]
[ "FE65", "FE65" ]
[ "Alzheimer type", "sporadic dementia" ]
9804344
Genomic organization and fine mapping of the keratin 2e gene (KRT2E): K2e V1 domain polymorphism and novel mutations in ichthyosis bullosa of Siemens.
We and others have previously shown that ichthyosis bullosa of Siemens, an autosomal dominant disorder characterized by epidermal thickening and blistering, is caused by mutations in the late-differentiation keratin K2e. Here, we have determined the genomic organization and complete sequence of the KRT2E gene, which consists of nine exons, spanning 7634 bp of DNA. The gene was mapped by high-resolution radiation-hybrid mapping to the interval between microsatellite markers D12S368 and CHLC.GATA11B02.1112. Several intragenic polymorphisms were detected, including an 18 bp duplication in exon 1, corresponding to the V1 domain of the K2e polypeptide. Genomic polymerase chain reaction conditions were optimized for all exons, and two novel mutations, N192Y in the 1A domain and E482K in the 2B domain of K2e, were found in ichthyosis bullosa of Siemens families. Mutations were excluded from 50 normal unrelated individuals by restriction analysis. These results emphasize that mutations in K2e underlie ichthyosis bullosa of Siemens and provide a comprehensive mutation detection strategy for ongoing studies of keratinizing disorders.
[ { "begin_idx": "226", "end_idx": "253", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal dominant disorder" }, { "begin_idx": "120", "end_idx": "149", "entity_id": "D053560", "entity_type": "Disease", "text_name": "ichthyosis bullosa of Siemens" }, { "begin_idx": "192", "end_idx": "221", "entity_id": "D053560", "entity_type": "Disease", "text_name": "ichthyosis bullosa of Siemens" }, { "begin_idx": "979", "end_idx": "1008", "entity_id": "D053560", "entity_type": "Disease", "text_name": "ichthyosis bullosa of Siemens" }, { "begin_idx": "1160", "end_idx": "1189", "entity_id": "D053560", "entity_type": "Disease", "text_name": "ichthyosis bullosa of Siemens" }, { "begin_idx": "45", "end_idx": "55", "entity_id": "3849", "entity_type": "Gene", "text_name": "keratin 2e" }, { "begin_idx": "62", "end_idx": "67", "entity_id": "3849", "entity_type": "Gene", "text_name": "KRT2E" }, { "begin_idx": "70", "end_idx": "73", "entity_id": "3849", "entity_type": "Gene", "text_name": "K2e" }, { "begin_idx": "367", "end_idx": "370", "entity_id": "3849", "entity_type": "Gene", "text_name": "K2e" }, { "begin_idx": "451", "end_idx": "456", "entity_id": "3849", "entity_type": "Gene", "text_name": "KRT2E" }, { "begin_idx": "790", "end_idx": "793", "entity_id": "3849", "entity_type": "Gene", "text_name": "K2e" }, { "begin_idx": "960", "end_idx": "963", "entity_id": "3849", "entity_type": "Gene", "text_name": "K2e" }, { "begin_idx": "1147", "end_idx": "1150", "entity_id": "3849", "entity_type": "Gene", "text_name": "K2e" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "45", "end_idx": "55", "entity_id": "3849", "entity_type": "Gene", "text_name": "keratin 2e" }, { "begin_idx": "790", "end_idx": "793", "entity_id": "3849", "entity_type": "Gene", "text_name": "K2e" } ]
[ { "begin_idx": "120", "end_idx": "149", "entity_id": "D053560", "entity_type": "Disease", "text_name": "ichthyosis bullosa of Siemens" }, { "begin_idx": "226", "end_idx": "253", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal dominant disorder" } ]
[ "keratin 2e", "K2e" ]
[ "ichthyosis bullosa of Siemens", "autosomal dominant disorder" ]
9805126
Heterozygous glycine substitution in the COL11A2 gene in the original patient with the Weissenbacher-Zweym ller syndrome demonstrates its identity with heterozygous OSMED (nonocular Stickler syndrome).
The original patient with the Weissenbacher-Zweym ller syndrome was analyzed for mutations in two candidate genes expressed in cartilage (COL2A1 and COL11A2). No mutations were found in the COL2A1 gene but the COL11A2 gene contained a single-base mutation that converted a codon for an obligate glycine to a codon for glutamate at position alpha 2-955 (G955E). The results here and those published previously indicate that the Weissenbacher-Zweym ller syndrome (heterozygous OSMED), nonocular Stickler syndrome, and homozygous OSMED are all caused by mutations in the COL11A2 gene.
[ { "begin_idx": "87", "end_idx": "121", "entity_id": "C535776", "entity_type": "Disease", "text_name": "Weissenbacher-Zweym ller syndrome" }, { "begin_idx": "233", "end_idx": "267", "entity_id": "C535776", "entity_type": "Disease", "text_name": "Weissenbacher-Zweym ller syndrome" }, { "begin_idx": "631", "end_idx": "665", "entity_id": "C535776", "entity_type": "Disease", "text_name": "Weissenbacher-Zweym ller syndrome" }, { "begin_idx": "173", "end_idx": "200", "entity_id": "C537494", "entity_type": "Disease", "text_name": "nonocular Stickler syndrome" }, { "begin_idx": "688", "end_idx": "715", "entity_id": "C537494", "entity_type": "Disease", "text_name": "nonocular Stickler syndrome" }, { "begin_idx": "342", "end_idx": "348", "entity_id": "1280", "entity_type": "Gene", "text_name": "COL2A1" }, { "begin_idx": "394", "end_idx": "400", "entity_id": "1280", "entity_type": "Gene", "text_name": "COL2A1" }, { "begin_idx": "41", "end_idx": "48", "entity_id": "1302", "entity_type": "Gene", "text_name": "COL11A2" }, { "begin_idx": "353", "end_idx": "360", "entity_id": "1302", "entity_type": "Gene", "text_name": "COL11A2" }, { "begin_idx": "414", "end_idx": "421", "entity_id": "1302", "entity_type": "Gene", "text_name": "COL11A2" }, { "begin_idx": "773", "end_idx": "780", "entity_id": "1302", "entity_type": "Gene", "text_name": "COL11A2" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "41", "end_idx": "48", "entity_id": "1302", "entity_type": "Gene", "text_name": "COL11A2" }, { "begin_idx": "394", "end_idx": "400", "entity_id": "1280", "entity_type": "Gene", "text_name": "COL2A1" } ]
[ { "begin_idx": "87", "end_idx": "121", "entity_id": "C535776", "entity_type": "Disease", "text_name": "Weissenbacher-Zweym ller syndrome" }, { "begin_idx": "233", "end_idx": "267", "entity_id": "C535776", "entity_type": "Disease", "text_name": "Weissenbacher-Zweym ller syndrome" } ]
[ "COL11A2", "COL2A1" ]
[ "Weissenbacher-Zweym ller syndrome", "Weissenbacher-Zweym ller syndrome" ]
9811940
Genetic association of an alpha2-macroglobulin (Val1000lle) polymorphism and Alzheimer's disease.
alpha2-Macroglobulin (A2M) is a proteinase inhibitor found in association with senile plaques (SP) in Alzheimer's disease (AD). A2M has been implicated biochemically in binding and degradation of the amyloid beta (Abeta) protein which accumulates in SP. We studied the relationship between Alzheimer's disease and a common A2M polymorphism, Val1000 (GTC)/Ile1000 (ATC), which occurs near the thiolester active site of the molecule. In an initial exploratory data set (90 controls and 171 Alzheimer's disease) we noted an increased frequency of the G/G genotype from 0.07 to 0.12. We therefore tested the hypothesis that the G/G genotype is over-represented in Alzheimer's disease in an additional independent data set: a group of 359 controls and 566 Alzheimer's disease patients. In the hypothesis testing cohort, the G/G genotype increased from 0.07 in controls to 0.12 in Alzheimer's disease (P < 0.05, Fisher's exact test). The odds ratio for Alzheimer's disease associated with the G/G genotype was 1.77 (1.16-2.70, P < 0.01) and in combination with APOE4 was 9.68 (95% CI 3.91-24.0, P < 0.001). The presence of the G allele was associated with an increase in Abeta burden in a small series. The A2M receptor, A2M-r/LRP, is a multifunctional receptor whose ligands include apolipoprotein E and the amyloid precursor protein. These four proteins have each been genetically linked to Alzheimer's disease, suggesting that they may participate in a common disease pathway.
[ { "begin_idx": "77", "end_idx": "96", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "200", "end_idx": "219", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "221", "end_idx": "223", "entity_id": "D000544", "entity_type": "Disease", "text_name": "AD" }, { "begin_idx": "388", "end_idx": "407", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "586", "end_idx": "605", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "758", "end_idx": "777", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "849", "end_idx": "868", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "973", "end_idx": "992", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "1045", "end_idx": "1064", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "1485", "end_idx": "1504", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "177", "end_idx": "191", "entity_id": "D058225", "entity_type": "Disease", "text_name": "senile plaques" }, { "begin_idx": "193", "end_idx": "195", "entity_id": "D058225", "entity_type": "Disease", "text_name": "SP" }, { "begin_idx": "348", "end_idx": "350", "entity_id": "D058225", "entity_type": "Disease", "text_name": "SP" }, { "begin_idx": "26", "end_idx": "46", "entity_id": "2", "entity_type": "Gene", "text_name": "alpha2-macroglobulin" }, { "begin_idx": "98", "end_idx": "118", "entity_id": "2", "entity_type": "Gene", "text_name": "alpha2-Macroglobulin" }, { "begin_idx": "1153", "end_idx": "1158", "entity_id": "348", "entity_type": "Gene", "text_name": "APOE4" }, { "begin_idx": "1376", "end_idx": "1392", "entity_id": "348", "entity_type": "Gene", "text_name": "apolipoprotein E" }, { "begin_idx": "1313", "end_idx": "1318", "entity_id": "4035", "entity_type": "Gene", "text_name": "A2M-r" }, { "begin_idx": "1319", "end_idx": "1322", "entity_id": "4035", "entity_type": "Gene", "text_name": "LRP" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "26", "end_idx": "46", "entity_id": "2", "entity_type": "Gene", "text_name": "alpha2-macroglobulin" }, { "begin_idx": "1319", "end_idx": "1322", "entity_id": "4035", "entity_type": "Gene", "text_name": "LRP" } ]
[ { "begin_idx": "77", "end_idx": "96", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" }, { "begin_idx": "1045", "end_idx": "1064", "entity_id": "D000544", "entity_type": "Disease", "text_name": "Alzheimer's disease" } ]
[ "alpha2-macroglobulin", "LRP" ]
[ "Alzheimer's disease", "Alzheimer's disease" ]
9825838
Predisposition towards urolithiasis associated with the NQO1 null-allele.
The distribution of two alleles of the NQO1 gene encoding NADP(H):quinone oxidoreductase was studied in 140 urolithiasis patients and 271 control individuals. The minor allele encoding a protein lacking quinone reductase activity was significantly more frequent (q = 0.214) among these patients than in control individuals (P = 0.135) indicating an increased risk for kidney stone formation among heterozygotes (odds ratio 1.83, confidence interval 1.17-2.86) and homozygotes for the null-allele (odds ratio 2.97, confidence interval 0.78-11.33). Since NADP(H):quinone oxidoreductase is thought to participate in activation of vitamin K for protein gamma-carboxylation, decreased activity of the enzyme in heterozygotes or in null-allele homozygotes may disturb the post-translational modification of urinary calcium-binding proteins protective against kidney stone formation. The NQO1 null-allele might therefore be a determinant in enhanced risk of urolithiasis.
[ { "begin_idx": "442", "end_idx": "454", "entity_id": "D007669", "entity_type": "Disease", "text_name": "kidney stone" }, { "begin_idx": "927", "end_idx": "939", "entity_id": "D007669", "entity_type": "Disease", "text_name": "kidney stone" }, { "begin_idx": "23", "end_idx": "35", "entity_id": "D052878", "entity_type": "Disease", "text_name": "urolithiasis" }, { "begin_idx": "182", "end_idx": "194", "entity_id": "D052878", "entity_type": "Disease", "text_name": "urolithiasis" }, { "begin_idx": "1025", "end_idx": "1037", "entity_id": "D052878", "entity_type": "Disease", "text_name": "urolithiasis" }, { "begin_idx": "132", "end_idx": "139", "entity_id": "1666", "entity_type": "Gene", "text_name": "NADP(H)" }, { "begin_idx": "627", "end_idx": "634", "entity_id": "1666", "entity_type": "Gene", "text_name": "NADP(H)" }, { "begin_idx": "56", "end_idx": "60", "entity_id": "1728", "entity_type": "Gene", "text_name": "NQO1" }, { "begin_idx": "113", "end_idx": "117", "entity_id": "1728", "entity_type": "Gene", "text_name": "NQO1" }, { "begin_idx": "955", "end_idx": "959", "entity_id": "1728", "entity_type": "Gene", "text_name": "NQO1" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "56", "end_idx": "60", "entity_id": "1728", "entity_type": "Gene", "text_name": "NQO1" }, { "begin_idx": "627", "end_idx": "634", "entity_id": "1666", "entity_type": "Gene", "text_name": "NADP(H)" } ]
[ { "begin_idx": "23", "end_idx": "35", "entity_id": "D052878", "entity_type": "Disease", "text_name": "urolithiasis" }, { "begin_idx": "23", "end_idx": "35", "entity_id": "D052878", "entity_type": "Disease", "text_name": "urolithiasis" } ]
[ "NQO1", "NADP(H)" ]
[ "urolithiasis", "urolithiasis" ]
9827908
Molecular characterization of the PK-LR gene in pyruvate kinase deficient Spanish patients. Red Cell Pathology Group of the Spanish Society of Haematology (AEHH).
The PK-LR gene has been studied in 12 unrelated patients with red cell pyruvate kinase deficiency and hereditary nonspherocytic haemolytic anaemia (CNSHA). The entire codifying region of the R-type PK gene and the flanking intronic regions were analysed by single-stranded conformation polymorphism (SSCP) followed by direct sequencing of abnormal DNA. 10 different mutations were identified in 22/24 alleles at risk. Eight of these were missense mutations that caused the following single amino acid changes: G514C (172Glu-Gln), G1010A (337Arg-Gln), G1015C (339Asp-Gln), T1070C (357Ile-Thr), C1223T (408Thr-Ile), G1291A (431Ala-Thr), C1456T (486Arg-Trp) and G1595A (532Arg-Gln). Two were nonsense mutations: G721T (241Glu-Stop) and C1675T (559Arg-Stop). 7/22 alleles demonstrated the same C1456 --> T mutation. The study of the polymorphic site at nucleotide (nt) 1705 performed in all cases disclosed a 1705 C/C mutation in 10 and a 1705 A/C mutation in three. This is the first report on the presence of several different L-type PK gene mutations within Spanish population. Furthermore, from the PK gene mutations found, six were unique and not previously described (1015C, 1070C, 1223T, 1291A, 1595A and 1675T) and one (C1456T) seems to be predominant in Spain. Interestingly, no case with the 1529A mutation commonly found in Northern European populations was present here.
[ { "begin_idx": "234", "end_idx": "260", "entity_id": "C564858", "entity_type": "Disease", "text_name": "pyruvate kinase deficiency" }, { "begin_idx": "265", "end_idx": "309", "entity_id": "D000745", "entity_type": "Disease", "text_name": "hereditary nonspherocytic haemolytic anaemia" }, { "begin_idx": "34", "end_idx": "39", "entity_id": "5313", "entity_type": "Gene", "text_name": "PK-LR" }, { "begin_idx": "167", "end_idx": "172", "entity_id": "5313", "entity_type": "Gene", "text_name": "PK-LR" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "34", "end_idx": "39", "entity_id": "5313", "entity_type": "Gene", "text_name": "PK-LR" }, { "begin_idx": "167", "end_idx": "172", "entity_id": "5313", "entity_type": "Gene", "text_name": "PK-LR" } ]
[ { "begin_idx": "234", "end_idx": "260", "entity_id": "C564858", "entity_type": "Disease", "text_name": "pyruvate kinase deficiency" }, { "begin_idx": "265", "end_idx": "309", "entity_id": "D000745", "entity_type": "Disease", "text_name": "hereditary nonspherocytic haemolytic anaemia" } ]
[ "PK-LR", "PK-LR" ]
[ "pyruvate kinase deficiency", "hereditary nonspherocytic haemolytic anaemia" ]
9829908
Use of denaturing gradient gel blots to screen for point mutations in the factor VIII gene.
Denaturing gradient gel electrophoresis (DGGE) is commonly used to search for point mutations in DNA fragments amplified in vitro by the polymerase chain reaction (PCR). For the complete detection of mutations in large genes with many exons, the DGGE-PCR approach, or any other PCR-based method, requires many primer sets and amplification reactions to scan the entire protein-coding sequence. We previously demonstrated that DGGE analysis using DNA blots detects mutations in Drosophila genes and sequence polymorphisms in human genes without prior PCR amplification. To determine if human point mutations could be detected using denaturing gradient gels (DGG blots), genomic DNA samples from hemophilia A families were analyzed for mutations in the factor VIII (FVIII) gene. Restriction enzyme digested DNA samples were subjected to DGGE and transferred to nylon blots. Hybridization of the DGG blots with FVIII cDNA probes revealed mutant and polymorphic DNA sequence differences. Among 26 affected families that were not carriers of intron 22 inversion mutations, 18 family-specific DNA fragment polymorphisms and one multiexon deletion were mapped. DNA sequencing of eight patient-specific polymorphic DNA fragments revealed six single base change mutations, one 4 bp deletion, and one 13 bp duplication.
[ { "begin_idx": "786", "end_idx": "798", "entity_id": "D006467", "entity_type": "Disease", "text_name": "hemophilia A" }, { "begin_idx": "74", "end_idx": "85", "entity_id": "2157", "entity_type": "Gene", "text_name": "factor VIII" }, { "begin_idx": "843", "end_idx": "854", "entity_id": "2157", "entity_type": "Gene", "text_name": "factor VIII" }, { "begin_idx": "856", "end_idx": "861", "entity_id": "2157", "entity_type": "Gene", "text_name": "FVIII" }, { "begin_idx": "1000", "end_idx": "1005", "entity_id": "2157", "entity_type": "Gene", "text_name": "FVIII" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "74", "end_idx": "85", "entity_id": "2157", "entity_type": "Gene", "text_name": "factor VIII" } ]
[ { "begin_idx": "786", "end_idx": "798", "entity_id": "D006467", "entity_type": "Disease", "text_name": "hemophilia A" } ]
[ "factor VIII" ]
[ "hemophilia A" ]
9834209
Congenital erythropoietic porphyria successfully treated by allogeneic bone marrow transplantation.
The long-term biochemical and clinical effectiveness of allogenic bone marrow transplantation (BMT) was shown in a severely affected, transfusion-dependent 18-month-old female with congenital erythropoietic porphyria (CEP), an autosomal recessive inborn error of heme biosynthesis resulting from mutations in the uroporphyrinogen III synthase (URO-synthase) gene. Three years post-BMT, the recipient had normal hemoglobin, markedly reduced urinary porphyrin excretion, and no cutaneous lesions with unlimited exposure to sunlight. The patient was homoallelic for a novel URO-synthase missense mutation, G188R, that expressed less than 5% of mean normal activity in Escherichia coli, consistent with her transfusion dependency. Because the clinical severity of CEP is highly variable, ranging from nonimmune hydrops fetalis to milder, later onset forms with only cutaneous lesions, the importance of genotyping newly diagnosed infants to select severely affected patients for BMT is emphasized. In addition, the long-term effectiveness of BMT in this patient provides the rationale for future hematopoietic stem cell gene therapy in severely affected patients with CEP.
[ { "begin_idx": "0", "end_idx": "35", "entity_id": "D017092", "entity_type": "Disease", "text_name": "Congenital erythropoietic porphyria" }, { "begin_idx": "281", "end_idx": "316", "entity_id": "D017092", "entity_type": "Disease", "text_name": "congenital erythropoietic porphyria" }, { "begin_idx": "318", "end_idx": "321", "entity_id": "D017092", "entity_type": "Disease", "text_name": "CEP" }, { "begin_idx": "860", "end_idx": "863", "entity_id": "D017092", "entity_type": "Disease", "text_name": "CEP" }, { "begin_idx": "1264", "end_idx": "1267", "entity_id": "D017092", "entity_type": "Disease", "text_name": "CEP" }, { "begin_idx": "327", "end_idx": "359", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive inborn error" }, { "begin_idx": "413", "end_idx": "442", "entity_id": "7390", "entity_type": "Gene", "text_name": "uroporphyrinogen III synthase" } ]
[ "Yes", "No" ]
[ true, true ]
[ { "begin_idx": "413", "end_idx": "442", "entity_id": "7390", "entity_type": "Gene", "text_name": "uroporphyrinogen III synthase" }, { "begin_idx": "413", "end_idx": "442", "entity_id": "7390", "entity_type": "Gene", "text_name": "uroporphyrinogen III synthase" } ]
[ { "begin_idx": "0", "end_idx": "35", "entity_id": "D017092", "entity_type": "Disease", "text_name": "Congenital erythropoietic porphyria" }, { "begin_idx": "327", "end_idx": "359", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive inborn error" } ]
[ "uroporphyrinogen III synthase", "uroporphyrinogen III synthase" ]
[ "Congenital erythropoietic porphyria", "autosomal recessive inborn error" ]
9856504
Functional characterisation of mutations in the ligand-binding domain of the androgen receptor gene in patients with androgen insensitivity syndrome.
Five mutations in the ligand-binding domain of the androgen receptor gene were identified in patients with complete (A765T, C784Y, R831X and M895T) or partial (R840G) androgen insensitivity. A765T and R831X have been reported previously whereas the other three mutations are novel. Receptors carrying these mutations were transiently expressed in COS-1 cells, and androgen binding and capacity to transactivate an androgen-responsive reporter gene were assayed. C784Y led to abolished androgen binding and transactivating capacity, R840G and M895T showed reduced specific binding and partial transactivation. The in vitro functions of the R840G and M895T mutants were improved with supraphysiological concentrations of steroid.
[ { "begin_idx": "117", "end_idx": "148", "entity_id": "D013734", "entity_type": "Disease", "text_name": "androgen insensitivity syndrome" }, { "begin_idx": "77", "end_idx": "94", "entity_id": "367", "entity_type": "Gene", "text_name": "androgen receptor" }, { "begin_idx": "201", "end_idx": "218", "entity_id": "367", "entity_type": "Gene", "text_name": "androgen receptor" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "77", "end_idx": "94", "entity_id": "367", "entity_type": "Gene", "text_name": "androgen receptor" } ]
[ { "begin_idx": "117", "end_idx": "148", "entity_id": "D013734", "entity_type": "Disease", "text_name": "androgen insensitivity syndrome" } ]
[ "androgen receptor" ]
[ "androgen insensitivity syndrome" ]
9887333
Characterization of ATM gene mutations in 66 ataxia telangiectasia families.
Ataxia telangiectasia (AT) is an autosomal recessive disease characterized by neurological and immunological symptoms, radiosensitivity and cancer predisposition. The gene mutated in AT, designated the ATM gene, encodes a large protein kinase with a PI-3 kinase-related domain. In this study, we investigated the mutational spectrum of the ATM gene in a cohort of AT patients living in Germany. We amplified and sequenced all 66 exons and the flanking untranslated regions from genomic DNA of 66 unrelated AT patients. We identified 46 different ATM mutations and 26 sequence polymorphisms and variants scattered throughout the gene. A total of 34 mutations have not been described in other populations. Seven mutations occurred in more than one family, but none of these accounted for more than five alleles in our patient group. The majority of the mutations were truncating, confirming that the absence of full-length ATM protein is the most common molecular basis of AT. Transcript analyses demonstrated single exon skipping as the consequence of most splice site substitutions, but a more complex pattern was observed for two mutations. Immunoblot studies of cell lines carrying ATM missense substitutions or in-frame deletions detected residual ATM protein in four cases. One of these mutations, a valine deletion proximal to the kinase domain, resulted in ATM protein levels >20% of normal in an AT lymphoblastoid cell line. In summary, our results survey and characterize a plethora of variations in the ATM gene identified by exon scanning sequencing and indicate a high diversity of mutations giving rise to AT in a non-isolated population.
[ { "begin_idx": "45", "end_idx": "66", "entity_id": "D001260", "entity_type": "Disease", "text_name": "ataxia telangiectasia" }, { "begin_idx": "77", "end_idx": "98", "entity_id": "D001260", "entity_type": "Disease", "text_name": "Ataxia telangiectasia" }, { "begin_idx": "100", "end_idx": "102", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" }, { "begin_idx": "260", "end_idx": "262", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" }, { "begin_idx": "441", "end_idx": "443", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" }, { "begin_idx": "583", "end_idx": "585", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" }, { "begin_idx": "1048", "end_idx": "1050", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" }, { "begin_idx": "1480", "end_idx": "1482", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" }, { "begin_idx": "1695", "end_idx": "1697", "entity_id": "D001260", "entity_type": "Disease", "text_name": "AT" }, { "begin_idx": "217", "end_idx": "223", "entity_id": "D009369", "entity_type": "Disease", "text_name": "cancer" }, { "begin_idx": "155", "end_idx": "194", "entity_id": "D009422", "entity_type": "Disease", "text_name": "neurological and immunological symptoms" }, { "begin_idx": "110", "end_idx": "137", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive disease" }, { "begin_idx": "20", "end_idx": "23", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "279", "end_idx": "282", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "417", "end_idx": "420", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "623", "end_idx": "626", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "998", "end_idx": "1001", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "1261", "end_idx": "1264", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "1328", "end_idx": "1331", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "1440", "end_idx": "1443", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "1589", "end_idx": "1592", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "20", "end_idx": "23", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" }, { "begin_idx": "1328", "end_idx": "1331", "entity_id": "472", "entity_type": "Gene", "text_name": "ATM" } ]
[ { "begin_idx": "45", "end_idx": "66", "entity_id": "D001260", "entity_type": "Disease", "text_name": "ataxia telangiectasia" }, { "begin_idx": "217", "end_idx": "223", "entity_id": "D009369", "entity_type": "Disease", "text_name": "cancer" } ]
[ "ATM", "ATM" ]
[ "ataxia telangiectasia", "cancer" ]
9888857
Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice.
The function of the central cannabinoid receptor (CB1) was investigated by invalidating its gene. Mutant mice did not respond to cannabinoid drugs, demonstrating the exclusive role of the CB1 receptor in mediating analgesia, reinforcement, hypothermia, hypolocomotion, and hypotension. The acute effects of opiates were unaffected, but the reinforcing properties of morphine and the severity of the withdrawal syndrome were strongly reduced. These observations suggest that the CB1 receptor is involved in the motivational properties of opiates and in the development of physical dependence and extend the concept of an interconnected role of CB1 and opiate receptors in the brain areas mediating addictive behavior.
[ { "begin_idx": "378", "end_idx": "389", "entity_id": "D007022", "entity_type": "Disease", "text_name": "hypotension" }, { "begin_idx": "345", "end_idx": "356", "entity_id": "D007035", "entity_type": "Disease", "text_name": "hypothermia" }, { "begin_idx": "504", "end_idx": "523", "entity_id": "D013375", "entity_type": "Disease", "text_name": "withdrawal syndrome" }, { "begin_idx": "685", "end_idx": "695", "entity_id": "D019966", "entity_type": "Disease", "text_name": "dependence" }, { "begin_idx": "77", "end_idx": "80", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "155", "end_idx": "158", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "293", "end_idx": "296", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "583", "end_idx": "586", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "748", "end_idx": "751", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" } ]
[ "Yes", "Yes", "Yes", "No", "No", "No" ]
[ true, true, true, false, false, false ]
[ { "begin_idx": "77", "end_idx": "80", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "77", "end_idx": "80", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "77", "end_idx": "80", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "583", "end_idx": "586", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "77", "end_idx": "80", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" }, { "begin_idx": "293", "end_idx": "296", "entity_id": "1268", "entity_type": "Gene", "text_name": "CB1" } ]
[ { "begin_idx": "685", "end_idx": "695", "entity_id": "D019966", "entity_type": "Disease", "text_name": "dependence" }, { "begin_idx": "378", "end_idx": "389", "entity_id": "D007022", "entity_type": "Disease", "text_name": "hypotension" }, { "begin_idx": "345", "end_idx": "356", "entity_id": "D007035", "entity_type": "Disease", "text_name": "hypothermia" }, { "begin_idx": "504", "end_idx": "523", "entity_id": "D013375", "entity_type": "Disease", "text_name": "withdrawal syndrome" }, { "begin_idx": "504", "end_idx": "523", "entity_id": "D013375", "entity_type": "Disease", "text_name": "withdrawal syndrome" }, { "begin_idx": "504", "end_idx": "523", "entity_id": "D013375", "entity_type": "Disease", "text_name": "withdrawal syndrome" } ]
[ "CB1", "CB1", "CB1", "CB1", "CB1", "CB1" ]
[ "dependence", "hypotension", "hypothermia", "withdrawal syndrome", "withdrawal syndrome", "withdrawal syndrome" ]
9920104
Phenocopies for deafness and goiter development in a large inbred Brazilian kindred with Pendred's syndrome associated with a novel mutation in the PDS gene.
Pendred's syndrome is an autosomal recessive disease characterized by goiter, impaired iodide organification, and congenital sensorineural deafness. The gene mutated in Pendred's syndrome, PDS (Pendred's syndrome gene), was cloned very recently and encodes the putative sulfate transporter pendrin. Pendred's syndrome may account for up to 10% of the cases with hereditary hearing loss, and pendrin mutations have also been found in a kindred with non-syndromic deafness. In this study, 41 individuals from a large, highly inbred pedigree from Northeastern Brazil were examined for features of Pendred's syndrome. Linkage studies and sequence analysis of the coding region of the PDS gene were performed with DNA from 36 individuals. The index patient, with the classical triad of deafness, positive perchlorate test, and goiter, was found to be homozygous for a deletion of thymidine 279 in exon 3, resulting in a frameshift and a premature stop codon at amino acid 96. This alteration resulted in truncation of the protein in the first transmembrane domain. Two other patients with deafness were found to be homozygous for this mutation; 19 were heterozygous and 14 were homozygous for the wild type allele. Surprisingly, 6 deaf individuals in this kindred were not homozygous for the PDS gene mutation; 3 were heterozygous and 3 were homozygous for the wild type allele, suggesting a probable distinct genetic cause for their deafness. All 3 homozygous individuals for the PDS mutation had goiters. However, goiters were also found in 10 heterozygous individuals and in 6 individuals without the PDS mutation and are most likely caused by iodine deficiency. In conclusion, we identified a novel mutation in the PDS gene causing Pendred's syndrome. The comparison of phenotype and genotype reveals, however, that phenocopies generated by distinct environmental and/or genetic causes are present in this kindred and that the diagnosis of Pendred's syndrome may be difficult without molecular analysis.
[ { "begin_idx": "89", "end_idx": "107", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome" }, { "begin_idx": "148", "end_idx": "151", "entity_id": "C536648", "entity_type": "Disease", "text_name": "PDS" }, { "begin_idx": "158", "end_idx": "176", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome" }, { "begin_idx": "327", "end_idx": "345", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome" }, { "begin_idx": "347", "end_idx": "350", "entity_id": "C536648", "entity_type": "Disease", "text_name": "PDS" }, { "begin_idx": "352", "end_idx": "375", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome gene" }, { "begin_idx": "457", "end_idx": "475", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome" }, { "begin_idx": "752", "end_idx": "770", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome" }, { "begin_idx": "838", "end_idx": "841", "entity_id": "C536648", "entity_type": "Disease", "text_name": "PDS" }, { "begin_idx": "1445", "end_idx": "1448", "entity_id": "C536648", "entity_type": "Disease", "text_name": "PDS" }, { "begin_idx": "1634", "end_idx": "1637", "entity_id": "C536648", "entity_type": "Disease", "text_name": "PDS" }, { "begin_idx": "1757", "end_idx": "1760", "entity_id": "C536648", "entity_type": "Disease", "text_name": "PDS" }, { "begin_idx": "1872", "end_idx": "1875", "entity_id": "C536648", "entity_type": "Disease", "text_name": "PDS" }, { "begin_idx": "1889", "end_idx": "1907", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome" }, { "begin_idx": "2097", "end_idx": "2115", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome" }, { "begin_idx": "1800", "end_idx": "1817", "entity_id": "D003409", "entity_type": "Disease", "text_name": "iodine deficiency" }, { "begin_idx": "16", "end_idx": "24", "entity_id": "D003638", "entity_type": "Disease", "text_name": "deafness" }, { "begin_idx": "606", "end_idx": "628", "entity_id": "D003638", "entity_type": "Disease", "text_name": "non-syndromic deafness" }, { "begin_idx": "939", "end_idx": "947", "entity_id": "D003638", "entity_type": "Disease", "text_name": "deafness" }, { "begin_idx": "1242", "end_idx": "1250", "entity_id": "D003638", "entity_type": "Disease", "text_name": "deafness" }, { "begin_idx": "1587", "end_idx": "1595", "entity_id": "D003638", "entity_type": "Disease", "text_name": "deafness" }, { "begin_idx": "29", "end_idx": "35", "entity_id": "D006042", "entity_type": "Disease", "text_name": "goiter" }, { "begin_idx": "228", "end_idx": "234", "entity_id": "D006042", "entity_type": "Disease", "text_name": "goiter" }, { "begin_idx": "980", "end_idx": "986", "entity_id": "D006042", "entity_type": "Disease", "text_name": "goiter" }, { "begin_idx": "1651", "end_idx": "1658", "entity_id": "D006042", "entity_type": "Disease", "text_name": "goiters" }, { "begin_idx": "1669", "end_idx": "1676", "entity_id": "D006042", "entity_type": "Disease", "text_name": "goiters" }, { "begin_idx": "283", "end_idx": "305", "entity_id": "D006319", "entity_type": "Disease", "text_name": "sensorineural deafness" }, { "begin_idx": "183", "end_idx": "210", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive disease" }, { "begin_idx": "520", "end_idx": "543", "entity_id": "D034381", "entity_type": "Disease", "text_name": "hereditary hearing loss" }, { "begin_idx": "148", "end_idx": "151", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" }, { "begin_idx": "347", "end_idx": "350", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" }, { "begin_idx": "352", "end_idx": "375", "entity_id": "5172", "entity_type": "Gene", "text_name": "Pendred's syndrome gene" }, { "begin_idx": "838", "end_idx": "841", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" }, { "begin_idx": "1445", "end_idx": "1448", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" }, { "begin_idx": "1634", "end_idx": "1637", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" }, { "begin_idx": "1757", "end_idx": "1760", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" }, { "begin_idx": "1872", "end_idx": "1875", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "352", "end_idx": "375", "entity_id": "5172", "entity_type": "Gene", "text_name": "Pendred's syndrome gene" }, { "begin_idx": "838", "end_idx": "841", "entity_id": "5172", "entity_type": "Gene", "text_name": "PDS" } ]
[ { "begin_idx": "352", "end_idx": "375", "entity_id": "C536648", "entity_type": "Disease", "text_name": "Pendred's syndrome gene" }, { "begin_idx": "183", "end_idx": "210", "entity_id": "D030342", "entity_type": "Disease", "text_name": "autosomal recessive disease" } ]
[ "Pendred's syndrome gene", "PDS" ]
[ "Pendred's syndrome gene", "autosomal recessive disease" ]
9921913
Mutational spectrum of the iduronate-2-sulfatase (IDS) gene in 36 unrelated Russian MPS II patients.
We present a mutational analysis of the iduronate-2-sulfatase (IDS) gene of 36 Russian patients with Hunter syndrome. Among 29 mutant alleles, there were 19 missense mutations, 1 nonsense mutation, 6 mutations affecting splice sites, and 3 major structural alterations resulting in deletions. Of the 25 different mutations, 15 are novel and unique. Most of the missense mutations result in intermediate or severe phenotypes.
[ { "begin_idx": "84", "end_idx": "90", "entity_id": "D016532", "entity_type": "Disease", "text_name": "MPS II" }, { "begin_idx": "202", "end_idx": "217", "entity_id": "D016532", "entity_type": "Disease", "text_name": "Hunter syndrome" }, { "begin_idx": "27", "end_idx": "48", "entity_id": "3423", "entity_type": "Gene", "text_name": "iduronate-2-sulfatase" }, { "begin_idx": "50", "end_idx": "53", "entity_id": "3423", "entity_type": "Gene", "text_name": "IDS" }, { "begin_idx": "141", "end_idx": "162", "entity_id": "3423", "entity_type": "Gene", "text_name": "iduronate-2-sulfatase" }, { "begin_idx": "164", "end_idx": "167", "entity_id": "3423", "entity_type": "Gene", "text_name": "IDS" } ]
[ "Yes" ]
[ true ]
[ { "begin_idx": "27", "end_idx": "48", "entity_id": "3423", "entity_type": "Gene", "text_name": "iduronate-2-sulfatase" } ]
[ { "begin_idx": "202", "end_idx": "217", "entity_id": "D016532", "entity_type": "Disease", "text_name": "Hunter syndrome" } ]
[ "iduronate-2-sulfatase" ]
[ "Hunter syndrome" ]
9927496
The molecular basis for apoptotic defects in patients with CD95 (Fas/Apo-1) mutations.
Heterozygous mutations of the receptor CD95 (Fas/Apo-1) are associated with defective lymphocyte apoptosis and a clinical disease characterized by lymphadenopathy, splenomegaly, and systemic autoimmunity. From our cohort of 11 families, we studied eight patients to define the mechanisms responsible for defective CD95-mediated apoptosis. Mutations in and around the death domain of CD95 had a dominant-negative effect that was explained by interference with the recruitment of the signal adapter protein, FADD, to the death domain. The intracellular domain (ICD) mutations were associated with a highly penetrant Canale-Smith syndrome (CSS) phenotype and an autosomal dominant inheritance pattern. In contrast, mutations affecting the CD95 extracellular domain (ECD) resulted in failure of extracellular expression of the mutant protein or impaired binding to CD95 ligand. They did not have a dominant-negative effect. In each of the families with an ECD mutation, only a single individual was affected. These observations were consistent with differing mechanisms of action and modes of inheritance of ICD and ECD mutations, suggesting that individuals with an ECD mutation may require additional defect(s) for expression of CSS.
[ { "begin_idx": "828", "end_idx": "848", "entity_id": "C535509", "entity_type": "Disease", "text_name": "extracellular domain" }, { "begin_idx": "850", "end_idx": "853", "entity_id": "C535509", "entity_type": "Disease", "text_name": "ECD" }, { "begin_idx": "1039", "end_idx": "1042", "entity_id": "C535509", "entity_type": "Disease", "text_name": "ECD" }, { "begin_idx": "1199", "end_idx": "1202", "entity_id": "C535509", "entity_type": "Disease", "text_name": "ECD" }, { "begin_idx": "1250", "end_idx": "1253", "entity_id": "C535509", "entity_type": "Disease", "text_name": "ECD" }, { "begin_idx": "278", "end_idx": "290", "entity_id": "D001327", "entity_type": "Disease", "text_name": "autoimmunity" }, { "begin_idx": "163", "end_idx": "193", "entity_id": "D004194", "entity_type": "Disease", "text_name": "defective lymphocyte apoptosis" }, { "begin_idx": "251", "end_idx": "263", "entity_id": "D013163", "entity_type": "Disease", "text_name": "splenomegaly" }, { "begin_idx": "624", "end_idx": "644", "entity_id": "D015270", "entity_type": "Disease", "text_name": "intracellular domain" }, { "begin_idx": "646", "end_idx": "649", "entity_id": "D015270", "entity_type": "Disease", "text_name": "ICD" }, { "begin_idx": "1191", "end_idx": "1194", "entity_id": "D015270", "entity_type": "Disease", "text_name": "ICD" }, { "begin_idx": "701", "end_idx": "722", "entity_id": "D056735", "entity_type": "Disease", "text_name": "Canale-Smith syndrome" }, { "begin_idx": "724", "end_idx": "727", "entity_id": "D056735", "entity_type": "Disease", "text_name": "CSS" }, { "begin_idx": "1314", "end_idx": "1317", "entity_id": "D056735", "entity_type": "Disease", "text_name": "CSS" }, { "begin_idx": "59", "end_idx": "63", "entity_id": "355", "entity_type": "Gene", "text_name": "CD95" }, { "begin_idx": "69", "end_idx": "74", "entity_id": "355", "entity_type": "Gene", "text_name": "Apo-1" }, { "begin_idx": "126", "end_idx": "130", "entity_id": "355", "entity_type": "Gene", "text_name": "CD95" }, { "begin_idx": "136", "end_idx": "141", "entity_id": "355", "entity_type": "Gene", "text_name": "Apo-1" }, { "begin_idx": "401", "end_idx": "405", "entity_id": "355", "entity_type": "Gene", "text_name": "CD95" }, { "begin_idx": "470", "end_idx": "474", "entity_id": "355", "entity_type": "Gene", "text_name": "CD95" }, { "begin_idx": "823", "end_idx": "827", "entity_id": "355", "entity_type": "Gene", "text_name": "CD95" }, { "begin_idx": "948", "end_idx": "959", "entity_id": "356", "entity_type": "Gene", "text_name": "CD95 ligand" }, { "begin_idx": "593", "end_idx": "597", "entity_id": "8772", "entity_type": "Gene", "text_name": "FADD" } ]
[ "Yes", "No" ]
[ false, false ]
[ { "begin_idx": "69", "end_idx": "74", "entity_id": "355", "entity_type": "Gene", "text_name": "Apo-1" }, { "begin_idx": "401", "end_idx": "405", "entity_id": "355", "entity_type": "Gene", "text_name": "CD95" } ]
[ { "begin_idx": "701", "end_idx": "722", "entity_id": "D056735", "entity_type": "Disease", "text_name": "Canale-Smith syndrome" }, { "begin_idx": "646", "end_idx": "649", "entity_id": "D015270", "entity_type": "Disease", "text_name": "ICD" } ]
[ "Apo-1", "CD95" ]
[ "Canale-Smith syndrome", "ICD" ]
9949172
Muc-1 core protein is expressed on multiple myeloma cells and is induced by dexamethasone.
Monoclonal antibodies (MoAbs) that selectively identify Muc-1 core protein (MoAbs DF3-P, VU-4H5) determinants were used to identify the Muc-1 glycoform present on 7 multiple myeloma (MM) cell lines, 5 MM patient plasma cells, 12 MM patient B cells, as well as 32 non-MM cell lines and normal hematopoietic cells. Flow cytometry studies demonstrated that all MM cell lines, MM patient plasma cells, and MM patient B cells expressed Muc-1 core protein epitopes. Circulating B cells from 4 normal donors also expressed Muc-1 core protein. In contrast, Muc-1 core protein was absent on 28 of 32 non-MM neoplastic cell lines, 17 of which expressed Muc-1. Splenic and tonsillar B cells, CD34(+) stem cells, resting T cells, and bone marrow plasma cells obtained from normal donors both lacked Muc-1 glycoforms. We next studied the effects of estrogen, progesterone, and glucocorticoid receptor agonists and antagonists on Muc-1 expression, because consensus sequences for the response elements of these steroids are present on the Muc-1 gene promoter. These studies showed that dexamethasone (Dex) induced Muc-1 expression on MM cell lines, as determined by both flow cytometry and Western blot analyses. Dex also induced upregulation of Muc-1 on prostate and ovarian cancer cell lines. Time and dose-response studies demonstrated that Dex induced maximal cell surface Muc-1 expression by 24 hours at concentrations of 10(-8) mol/L. Dex induced Muc-1 upregulation could be blocked with a 10-fold excess of the glucocorticoid receptor antagonist RU486, confirming that Dex was acting via the glucocorticoid receptor. No changes in Muc-1 expression were observed on MM cells treated with estrogen and progesterone receptor agonists and antagonists or with RU486. These studies provide the framework for targeting Muc-1 core protein in vaccination and serotherapy trials in MM. In addition, the finding that Muc-1 expression on MM cells can be augmented by Dex at pharmacologically achievable levels suggests their potential utility in enhancing treatments targeting Muc-1 in MM.
[ { "begin_idx": "35", "end_idx": "51", "entity_id": "D009101", "entity_type": "Disease", "text_name": "multiple myeloma" }, { "begin_idx": "256", "end_idx": "272", "entity_id": "D009101", "entity_type": "Disease", "text_name": "multiple myeloma" }, { "begin_idx": "274", "end_idx": "276", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "292", "end_idx": "294", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "320", "end_idx": "322", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "358", "end_idx": "360", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "449", "end_idx": "451", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "464", "end_idx": "466", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "493", "end_idx": "495", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "686", "end_idx": "688", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "1211", "end_idx": "1213", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "1749", "end_idx": "1751", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "1956", "end_idx": "1958", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "2010", "end_idx": "2012", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "2158", "end_idx": "2160", "entity_id": "D009101", "entity_type": "Disease", "text_name": "MM" }, { "begin_idx": "1345", "end_idx": "1359", "entity_id": "D010051", "entity_type": "Disease", "text_name": "ovarian cancer" }, { "begin_idx": "955", "end_idx": "978", "entity_id": "2908", "entity_type": "Gene", "text_name": "glucocorticoid receptor" }, { "begin_idx": "1595", "end_idx": "1618", "entity_id": "2908", "entity_type": "Gene", "text_name": "glucocorticoid receptor" }, { "begin_idx": "1676", "end_idx": "1699", "entity_id": "2908", "entity_type": "Gene", "text_name": "glucocorticoid receptor" }, { "begin_idx": "0", "end_idx": "5", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "147", "end_idx": "152", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "227", "end_idx": "232", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "522", "end_idx": "527", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "607", "end_idx": "612", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "640", "end_idx": "645", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "734", "end_idx": "739", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "878", "end_idx": "883", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1007", "end_idx": "1012", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1116", "end_idx": "1121", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1191", "end_idx": "1196", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1323", "end_idx": "1328", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1454", "end_idx": "1459", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1530", "end_idx": "1535", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1715", "end_idx": "1720", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1896", "end_idx": "1901", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1990", "end_idx": "1995", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "2149", "end_idx": "2154", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "1784", "end_idx": "1805", "entity_id": "5241", "entity_type": "Gene", "text_name": "progesterone receptor" }, { "begin_idx": "772", "end_idx": "776", "entity_id": "947", "entity_type": "Gene", "text_name": "CD34" } ]
[ "Yes", "No" ]
[ true, false ]
[ { "begin_idx": "0", "end_idx": "5", "entity_id": "4582", "entity_type": "Gene", "text_name": "Muc-1" }, { "begin_idx": "772", "end_idx": "776", "entity_id": "947", "entity_type": "Gene", "text_name": "CD34" } ]
[ { "begin_idx": "35", "end_idx": "51", "entity_id": "D009101", "entity_type": "Disease", "text_name": "multiple myeloma" }, { "begin_idx": "1345", "end_idx": "1359", "entity_id": "D010051", "entity_type": "Disease", "text_name": "ovarian cancer" } ]
[ "Muc-1", "CD34" ]
[ "multiple myeloma", "ovarian cancer" ]
9950439
Association of two silent polymorphisms of platelet glycoprotein Ia/IIa receptor with risk of myocardial infarction: a case-control study.
BACKGROUND: The platelet membrane glycoprotein Ia/IIa plays a major part in platelet function as a primary receptor for collagen. A previous report showed a variation of glycoprotein Ia/IIa receptor density and function associated with two silent and linked polymorphisms (807C/T and 873G/A) within the glycoprotein Ia gene. Because platelet thrombus formation is implicated in the pathogenesis of acute myocardial infarction, we investigated these polymorphisms among patients who had had a myocardial infarction. METHODS: We did a 2/1 case-control study including 177 patients (median age 57.0 [range 32-72] years) and 89 controls with same age and sex. Distributions of the 807C/T and 873G/A polymorphisms were investigated by genotyping DNA by PCR, single-strand conformation polymorphism analysis, and sequencing. FINDINGS: The prevalence of the homozygous 807T/873A genotype was 2.9 times higher among patients with myocardial infarction than among controls (16.4% vs 5.6%, p=0.022). There was an association between patients homozygous for the 807T/873A allele and myocardial infarction (odds ratio 3.3 [95% CI 1.2-8.8]), which was strongest in a subgroup of smokers. The homozygous 807T/873A genotype remained an independent risk factor for myocardial infarction (p=0.005) when age, sex, smoking, hypertension, diabetes, body-mass index, LDL-cholesterol and HDL-cholesterol, and fibrinogen were adjusted for by logistic regression. INTERPRETATION: The 807T/873A homozygosity of the platelet glycoprotein Ia/IIa gene polymorphism, associated with differences in surface collagen receptor density and activity, appears to be an independent risk factor for acute myocardial infarction. Our findings need to be confirmed in a larger, prospective study that includes patients from different populations and cardiovascular risk groups.
[ { "begin_idx": "472", "end_idx": "489", "entity_id": "D001791", "entity_type": "Disease", "text_name": "platelet thrombus" }, { "begin_idx": "1458", "end_idx": "1466", "entity_id": "D003920", "entity_type": "Disease", "text_name": "diabetes" }, { "begin_idx": "1444", "end_idx": "1456", "entity_id": "D006973", "entity_type": "Disease", "text_name": "hypertension" }, { "begin_idx": "94", "end_idx": "115", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "537", "end_idx": "564", "entity_id": "D009203", "entity_type": "Disease", "text_name": "acute myocardial infarction" }, { "begin_idx": "631", "end_idx": "652", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "1061", "end_idx": "1082", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "1211", "end_idx": "1232", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "1388", "end_idx": "1409", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" }, { "begin_idx": "1801", "end_idx": "1828", "entity_id": "D009203", "entity_type": "Disease", "text_name": "acute myocardial infarction" }, { "begin_idx": "52", "end_idx": "67", "entity_id": "22915", "entity_type": "Gene", "text_name": "glycoprotein Ia" }, { "begin_idx": "309", "end_idx": "324", "entity_id": "22915", "entity_type": "Gene", "text_name": "glycoprotein Ia" }, { "begin_idx": "442", "end_idx": "457", "entity_id": "22915", "entity_type": "Gene", "text_name": "glycoprotein Ia" }, { "begin_idx": "1638", "end_idx": "1653", "entity_id": "22915", "entity_type": "Gene", "text_name": "glycoprotein Ia" }, { "begin_idx": "155", "end_idx": "188", "entity_id": "3673", "entity_type": "Gene", "text_name": "platelet membrane glycoprotein Ia" } ]
[ "Yes", "No" ]
[ false, true ]
[ { "begin_idx": "155", "end_idx": "188", "entity_id": "3673", "entity_type": "Gene", "text_name": "platelet membrane glycoprotein Ia" }, { "begin_idx": "52", "end_idx": "67", "entity_id": "22915", "entity_type": "Gene", "text_name": "glycoprotein Ia" } ]
[ { "begin_idx": "537", "end_idx": "564", "entity_id": "D009203", "entity_type": "Disease", "text_name": "acute myocardial infarction" }, { "begin_idx": "94", "end_idx": "115", "entity_id": "D009203", "entity_type": "Disease", "text_name": "myocardial infarction" } ]
[ "platelet membrane glycoprotein Ia", "glycoprotein Ia" ]
[ "acute myocardial infarction", "myocardial infarction" ]