Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Cell Physiol ; 237(6): 2713-2723, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35621037

RESUMO

TMEM67 (mecklin or MKS3) locates in the transition zone of cilia. Dysfunction of TMEM67 disrupts cilia-related signaling and leads to developmental defects of multiple organs in humans. Typical autosomal recessive TMEM67 defects cause partial overlapping phenotypes, including abnormalities in the brain, eyes, liver, kidneys, bones, and so forth. However, emerging reports of isolated nephronophthisis suggest the possibility of a broader phenotype spectrum. In this study, we analyzed the genetic data of cholestasis patients with no obvious extrahepatic involvement but with an unexplained high level of gamma-glutamyl transpeptidase (GGT). We identified five Han Chinese patients from three unrelated families with biallelic nonnull low-frequency TMEM67 variants. All variants were predicted pathogenic in silico, of which p. Arg820Ile and p. Leu144del were previously unreported. In vitro studies revealed that the protein levels of the TMEM67 variants were significantly decreased; however, their interaction with MKS1 remained unaffected. All the patients, aged 7-39 years old, had silently progressive cholestasis with elevated GGT but had normal bilirubin levels. Histological studies of liver biopsy of patients 1, 3, and 5 showed the presence of congenital hepatic fibrosis. We conclude that variants in TMEM67 are associated with a mild phenotype of unexplained, persistent, anicteric, and high GGT cholestasis without typical symptoms of TMEM67 defects; this possibility should be considered by physicians in gastroenterology and hepatology.


Assuntos
Colestase , gama-Glutamiltransferase , Colestase/genética , Doenças Genéticas Inatas , Humanos , Cirrose Hepática/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fenótipo , gama-Glutamiltransferase/genética
2.
J Med Genet ; 58(8): 514-525, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-32737136

RESUMO

BACKGROUND: For many children with intrahepatic cholestasis and high-serum gamma-glutamyl transferase (GGT) activity, a genetic aetiology of hepatobiliary disease remains undefined. We sought to identify novel genes mutated in children with idiopathic high-GGT intrahepatic cholestasis, with clinical, histopathological and functional correlations. METHODS: We assembled a cohort of 25 children with undiagnosed high-GGT cholestasis and without clinical features of biliary-tract infection or radiological features of choledochal malformation, sclerosing cholangitis or cholelithiasis. Mutations were identified through whole-exome sequencing and targeted Sanger sequencing. We reviewed histopathological findings and assessed phenotypical effects of ZFYVE19 deficiency in cultured cells by immunofluorescence microscopy. RESULTS: Nine Han Chinese children harboured biallelic, predictedly complete loss-of-function pathogenic mutations in ZFYVE19 (c.314C>G, p.S105X; c.379C>T, p.Q127X; c.514C>T, p.R172X; c.547C>T, p.R183X; c.226A>G, p.M76V). All had portal hypertension and, at liver biopsy, histopathological features of the ductal plate malformation (DPM)/congenital hepatic fibrosis (CHF). Four children required liver transplantation for recurrent gastrointestinal haemorrhage. DPM/CHF was confirmed at hepatectomy, with sclerosing small-duct cholangitis. Immunostaining for two primary-cilium axonemal proteins found expression that was deficient intraluminally and ectopic within cholangiocyte cytoplasm. ZFYVE19 depletion in cultured cells yielded abnormalities of centriole and axoneme. CONCLUSION: Biallelic ZFYVE19 mutations can lead to high-GGT cholestasis and DPM/CHF in vivo. In vitro, they can lead to centriolar and axonemal abnormalities. These observations indicate that mutation in ZFYVE19 results, through as yet undefined mechanisms, in a ciliopathy.


Assuntos
Colangite Esclerosante/genética , Colestase Intra-Hepática/genética , Mutação/genética , Proteínas Oncogênicas/genética , Alelos , Sequência de Aminoácidos , Linhagem Celular Tumoral , Doenças Genéticas Inatas , Células HeLa , Humanos , Cirrose Hepática , Sequenciamento do Exoma/métodos
3.
Hum Mutat ; 41(2): 502-511, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31696999

RESUMO

To assess the spectrum of pediatric clinical phenotypes in TJP2 disease, we reviewed records of our seven patients in whom intrahepatic cholestasis was associated with biallelic TJP2 variants (13; 12 novel) and correlated clinical manifestations with mutation type. The effect of a splicing variant was analyzed with a minigene assay. The effects of three missense variants were analyzed with protein expression in vitro. Our patients had both remitting and persistent cholestasis. Three exhibited growth retardation. Six responded to treatment with cholestyramine, ursodeoxycholic acid, or both. Two had cholecystolithiasis. None required liver transplantation or developed hepatocellular or cholangiocellular malignancy. None manifested extrahepatic disease not attributable to effects of cholestasis. The variant c.2180-5T>G resulted in exon 15 skipping with in-frame deletion of 32 amino acid residues in TJP2. The three missense variants decreased but did not abolish TJP2 expression. Patients with truncating or canonical splice-site variants had clinically more severe disease. TJP2 disease in children includes a full clinical spectrum of severity, with mild or intermittent forms as well as the severe and minimal forms hitherto described. Biallelic TJP2 variants must be considered in children with clinically intermittent or resolved intrahepatic cholestasis.


Assuntos
Colestase Intra-Hepática/diagnóstico , Colestase Intra-Hepática/genética , Predisposição Genética para Doença , Variação Genética , Proteína da Zônula de Oclusão-2/genética , Idade de Início , Alelos , Substituição de Aminoácidos , Biópsia , Biologia Computacional/métodos , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Mutação , Linhagem , Splicing de RNA , Sequenciamento do Exoma
4.
BMC Med Genet ; 19(1): 139, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30086703

RESUMO

BACKGROUND: Branchio-oto-renal (BOR) syndrome is a dominant autosomal disorder characterized by phenotypes such as hearing loss, branchial fistulae, preauricular pits, and renal abnormalities. EYA1, the human homolog of the Drosophila "eye absent" gene on chromosome 8q13.3, is recognized as one of the most important genes associated with BOR syndrome. METHODS: The proposita in this study was a 5-year-old Chinese girl with hearing loss, bilateral otitis media with effusion, microtia, facial hypoplasia, palatoschisis, and bilateral branchial cleft fistulae. The girl's family members, except two who were deceased, agreed to undergo clinical examination. We collected blood samples from 10 family members, including six who were affected by the syndrome. Genomic DNA was extracted and subjected to Sanger sequencing. A minigene assay was performed to confirm whether splicing signals were altered. In addition, we performed western blotting to determine alterations in protein levels of the wild-type and mutant gene. RESULTS: Clinical tests showed that some of the family members met the criteria for BOR syndrome. The affected members harbored a novel heterozygous nonsense variation in exon 11 of EYA1, whereas no unaffected member carried the mutation at this position. Functional experiments did not detect abnormal splicing at the RNA level; however, western blotting showed that the mutated protein was truncated. CONCLUSIONS: This study reports a novel mutation associated with BOR syndrome in a Chinese family. We highlight the usefulness of genetic testing in the diagnosis of BOR syndrome. Thus, we believe that this report would benefit clinicians in this field.


Assuntos
Síndrome Brânquio-Otorrenal/genética , Códon sem Sentido/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Nucleares/genética , Proteínas Tirosina Fosfatases/genética , Povo Asiático , Pré-Escolar , DNA/genética , Éxons/genética , Feminino , Perda Auditiva/genética , Humanos , Masculino , Otite Média/genética , Linhagem , Fenótipo
5.
Hepatology ; 65(5): 1655-1669, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28027573

RESUMO

Hereditary cholestasis in childhood and infancy with normal serum gamma-glutamyltransferase (GGT) activity is linked to several genes. Many patients, however, remain genetically undiagnosed. Defects in myosin VB (MYO5B; encoded by MYO5B) cause microvillus inclusion disease (MVID; MIM251850) with recurrent watery diarrhea. Cholestasis, reported as an atypical presentation in MVID, has been considered a side effect of parenteral alimentation. Here, however, we report on 10 patients who experienced cholestasis associated with biallelic, or suspected biallelic, mutations in MYO5B and who had neither recurrent diarrhea nor received parenteral alimentation. Seven of them are from two study cohorts, together comprising 31 undiagnosed low-GGT cholestasis patients; 3 are sporadic. Cholestasis in 2 patients was progressive, in 3 recurrent, in 2 transient, and in 3 uncategorized because of insufficient follow-up. Liver biopsy specimens revealed giant-cell change of hepatocytes and intralobular cholestasis with abnormal distribution of bile salt export pump (BSEP) at canaliculi, as well as coarse granular dislocation of MYO5B. Mass spectrometry of plasma demonstrated increased total bile acids, primary bile acids, and conjugated bile acids, with decreased free bile acids, similar to changes in BSEP-deficient patients. Literature review revealed that patients with biallelic mutations predicted to eliminate MYO5B expression were more frequent in typical MVID than in isolated-cholestasis patients (11 of 38 vs. 0 of 13). CONCLUSION: MYO5B deficiency may underlie 20% of previously undiagnosed low-GGT cholestasis. MYO5B deficiency appears to impair targeting of BSEP to the canalicular membrane with hampered bile acid excretion, resulting in a spectrum of cholestasis without diarrhea. (Hepatology 2017;65:1655-1669).


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/deficiência , Colestase Intra-Hepática/genética , Cadeias Pesadas de Miosina/genética , Miosina Tipo V/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/sangue , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Ácidos e Sais Biliares/sangue , Colestase Intra-Hepática/sangue , Colestase Intra-Hepática/patologia , Análise Mutacional de DNA , Exoma , Feminino , Humanos , Lactente , Recém-Nascido , Fígado/metabolismo , Fígado/patologia , Masculino , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Estudos Retrospectivos
6.
Ann Vasc Surg ; 28(8): 1909-12, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25110237

RESUMO

BACKGROUND: This study aimed at exploring the causative gene and summarizing the clinical characteristics in a Chinese thoracic aortic aneurysm and dissection (TAAD) family. METHODS: Family members were examined for features of syndromic genetic diseases by clinician and geneticist. Genomic DNA was extracted from 2 distantly related members with definite TAAD for exome sequencing. RESULTS: A pathogenic mutation (rs111426349, c.1459C >T) of transforming growth factor ß receptor 1 (TGFBR1) was confirmed, which result in the amino acid substitution p.R487W. Fourteen TGFBR1 mutation carriers were detected among 39 tested members in this family. The average age at diagnosis of aortic root dilatation or aneurysm was 23.2 ± 12.6 years (range 3-37 years). Early onset of aortic root dilatation was significant in this family without reported phenotypes. The David procedure was performed prophylactically in 3 carriers of this family. CONCLUSIONS: Familial TAAD caused by TGFBR1 mutation (c.1459C >T) was confirmed in a large Chinese Han ethnic family using exome sequencing. Aggressively prophylactic David procedure may be not necessary at a smaller aortic size in familial TAAD patients with TGFBR1 mutation and further observation is warranted.


Assuntos
Aneurisma da Aorta Torácica/genética , Dissecção Aórtica/genética , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Adolescente , Adulto , Dissecção Aórtica/diagnóstico , Dissecção Aórtica/cirurgia , Aneurisma da Aorta Torácica/diagnóstico , Aneurisma da Aorta Torácica/cirurgia , Pré-Escolar , China , Genótipo , Humanos , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Receptor do Fator de Crescimento Transformador beta Tipo I , Resultado do Tratamento
7.
Pharmacogenomics ; 13(10): 1193-201, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22909208

RESUMO

AIM: Allopurinol is widely used as an effective urate-lowering drug and is one of the most frequent causes of cutaneous adverse drug reactions (cADRs). Recently, a strong association of HLA-B*58:01 with allopurinol-induced severe cADRs was identified. This study investigated the predisposition to different types of allopurinol-cADRs conferred by HLA-B*5801 in a Han population from mainland China. PATIENTS & METHODS: HLA-B genotyping was performed on 38 Chinese patients with different types of allopurinol-cADRs from 2008 to 2011. RESULTS: All the allopurinol-cADR patients carried HLA-B*58:01, in contrast with only 11.11% (7/63) in the allopurinol-tolerant patients (odds ratio [OR] = 580.07; p < 0.0001) and 13.99% (80/572) in a Han Chinese population from the human MHC database (dbMHC; OR: 471.09; p < 0.0001) carried the genotype. Each type of allopurinol cADRs revealed a statistically significant association with HLA-B*58:01. In particular, the risk of allopurinol-induced maculopapular eruption was significantly higher in patients with HLA-B*58:01 (OR: 339.00; p < 0.0001). CONCLUSION: The strong association of both the mild and severe types of allopurinol cADRs with the HLA-B*58:01 allele were observed. The results indicated that the prospective use of a genetic test of HLA-B*58:01 might reduce the prevalence of allopurinol-induced cADRs. Original submitted 7 March 2012; Revision submitted 21 May 2012.


Assuntos
Alopurinol , Antígenos HLA-B/genética , Síndrome de Stevens-Johnson/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Alopurinol/administração & dosagem , Alopurinol/toxicidade , China , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/genética , Exantema/induzido quimicamente , Exantema/genética , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Risco , Síndrome de Stevens-Johnson/induzido quimicamente
8.
Cancer Sci ; 99(4): 770-80, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18307539

RESUMO

China has the largest numbers of hereditary non-polyposis colorectal cancer (HNPCC) patients based on its population of 1.4 billion. However, the clinical data and mismatch repair (MMR) gene analyses have been limited. Here we performed microsatellite instability (MSI) and immunohistochemistry (IHC) analyses on a series of patients with a high-risk for HNPCC: 61 patients with family histories fulfilling Amsterdam criteria II (ACII-HNPCC) or suspected HNPCC criteria (S-HNPCC), and 106 early onset colorectal cancer (CRC) patients. Sixty late-onset CRC patients were used as control. Methylation of the hMLH1 promoter was analyzed on tumors lacking hMLH1 expression. MMR germ-line mutations were screened on patients with tumors classified as MSI-H/L or negative for IHC. We identified 27 germ-line MMR variants in the 167 patients with a high-risk for HNPCC while only one germ-line mutation in hMSH6 was found in the late-onset CRC group. Of those, 23 were pathogenic mutations. The high incidence of gastric and hepatobiliary cancers coupled with the increasing number of small families in China reduces the sensitivity (43.5%, 30.4%) and positive predictive value (PPV) (45.5%, 17.9%) of the ACII- or S-HNPCC criteria. MSI or IHC testing are highly sensitive in detecting pathogenic mutations (sensitivities = 91.3% and 95.6%, respectively), but the PPVs are quite low (25.6% and 27.8%, respectively). Considering that all 12 tumors with pathogenic mutations in hMLH1 also showed promoter unmethylation, the sensitivity of IHC in conjunction with hMLH1 promoter methylation analysis is not reduced, but the PPV was increased from 27.8% to 61.1%, and the total cost was greatly reduced.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Povo Asiático/genética , Neoplasias Colorretais Hereditárias sem Polipose/diagnóstico , Neoplasias Colorretais Hereditárias sem Polipose/genética , Reparo de Erro de Pareamento de DNA , Testes Genéticos/métodos , Proteínas Nucleares/genética , Adulto , China , Análise Custo-Benefício , Metilação de DNA , Proteínas de Ligação a DNA/genética , Feminino , Testes Genéticos/economia , Mutação em Linhagem Germinativa , Humanos , Imuno-Histoquímica , Masculino , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS/genética , Regiões Promotoras Genéticas
9.
J Clin Psychiatry ; 68(9): 1358-67, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17915974

RESUMO

BACKGROUND: A number of studies have pointed to the involvement of AKT signaling pathways in the etiology of schizophrenia. The purpose of this study was to determine whether the AKT1 gene is involved in the etiology of schizophrenia and whether it affects therapeutic outcomes in the Chinese population. METHOD: Five single nucleotide polymorphisms (SNPs) were genotyped among 384 schizophrenic patients (DSM-IV criteria) and 384 healthy controls from the Chinese population. We systematically analyzed the association of the AKT1 gene with schizophrenia on the basis of sex, age at onset, therapeutic response to typical antipsychotics and atypical antipsychotics, and presence or absence of extrapyramidal syndrome. The study was conducted from May 2004 to June 2006. RESULTS: We found a positive association of the G allele of the SNP marker rs3803300 with schizophrenia (p = .003), both in early-onset and late-onset subjects, and that a haplotype A-G-C-G-A constructed by the 5 SNPs showed significant association (p = .00004886). However, we found no relationship between any of the 5 SNP markers and therapeutic response to typical and atypical antipsychotics and chlorpromazine-induced extrapyramidal syndrome. CONCLUSIONS: Our study suggests that AKT1 is a susceptibility gene for schizophrenia in the Chinese population and that the AKT1 gene may play no major role in the therapeutic response to antipsychotics or in chlorpromazine-induced extrapyramidal syndrome.


Assuntos
Antipsicóticos/uso terapêutico , Povo Asiático/genética , Clorpromazina/uso terapêutico , Expressão Gênica/genética , Polimorfismo Genético/genética , Proteínas Proto-Oncogênicas c-akt/genética , Esquizofrenia , Adulto , Idade de Início , Antipsicóticos/efeitos adversos , Doenças dos Gânglios da Base/induzido quimicamente , Doenças dos Gânglios da Base/etnologia , China , Clorpromazina/efeitos adversos , Cromossomos Humanos Par 14/genética , Primers do DNA/genética , Feminino , Frequência do Gene , Genótipo , Haplótipos/genética , Humanos , Masculino , Plasticidade Neuronal/fisiologia , Esquizofrenia/tratamento farmacológico , Esquizofrenia/etnologia , Esquizofrenia/genética , Transdução de Sinais/genética
10.
Am J Hum Genet ; 73(2): 377-82, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12815562

RESUMO

Dyschromatosis symmetrica hereditaria (DSH) is a hereditary skin disease characterized by the presence of hyperpigmented and hypopigmented macules on extremities and face. The gene, or even its chromosomal location, for DSH has not yet been identified. In this study, two Chinese families with DSH were identified and subjected to a genomewide screen for linkage analysis. Two-point linkage analysis for pedigree A (maximum LOD score [Z(max)] = 7.28 at recombination fraction [theta] = 0.00) and pedigree B (Z(max) = 2.41 at theta = 0.00) mapped the locus for DSH in the two families to chromosome 6q. Subsequent multipoint analysis of the two families also provided additional support for the DSH gene being located within the region 6q24.2-q25.2, with Z(max) = 10.64. Haplotype analysis confined the locus within an interval of 10.2 Mbp, flanked by markers D6S1703 and D6S1708. The two families had no identical haplotype within the defined region, which suggests that the two families were different in origin. Further work on identification of the gene for DSH will open new avenues to exploration of the genetics of pigmentation.


Assuntos
Cromossomos Humanos Par 6/genética , Transtornos da Pigmentação/genética , China , Mapeamento Cromossômico , Feminino , Marcadores Genéticos , Haplótipos , Humanos , Escore Lod , Masculino , Neurofibromatose 1/genética , Linhagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA