Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Ann Oncol ; 28(1): 75-82, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27687306

RESUMO

Background: Lung adenocarcinomas (LUADs) lead to the majority of deaths attributable to lung cancer. We performed whole-exome sequencing (WES) and immune profiling analyses of a unique set of clinically annotated early-stage LUADs to better understand the pathogenesis of this disease and identify clinically relevant molecular markers. Methods: We performed WES of 108 paired stage I-III LUADs and normal lung tissues using the Illumina HiSeq 2000 platform. Ten immune markers (PD-L1, PD-1, CD3, CD4, CD8, CD45ro, CD57, CD68, FOXP3 and Granzyme B) were profiled by imaging-based immunohistochemistry (IHC) in a subset of LUADs (n = 92). Associations among mutations, immune markers and clinicopathological variables were analyzed using ANOVA and Fisher's exact test. Cox proportional hazards regression models were used for multivariate analysis of clinical outcome. Results: LUADs in this cohort exhibited an average of 243 coding mutations. We identified 28 genes with significant enrichment for mutation. SETD2-mutated LUADs exhibited relatively poor recurrence- free survival (RFS) and mutations in STK11 and ATM were associated with poor RFS among KRAS-mutant tumors. EGFR, KEAP1 and PIK3CA mutations were predictive of poor response to adjuvant therapy. Immune marker analysis revealed that LUADs in smokers and with relatively high mutation burdens exhibited increased levels of immune markers. Analysis of immunophenotypes revealed that LUADs with STK11 mutations exhibited relatively low levels of infiltrating CD4+/CD8+ T-cells indicative of a muted immune response. Tumoral PD-L1 was significantly elevated in TP53 mutant LUADs whereas PIK3CA mutant LUADs exhibited markedly down-regulated PD-L1 expression. LUADs with TP53 or KEAP1 mutations displayed relatively increased CD57 and Granzyme B levels indicative of augmented natural killer (NK) cell infiltration. Conclusion(s): Our study highlights molecular and immune phenotypes that warrant further analysis for their roles in clinical outcomes and personalized immune-based therapy of LUAD.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/imunologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Adenocarcinoma/mortalidade , Adenocarcinoma de Pulmão , Biomarcadores Tumorais/análise , Análise Mutacional de DNA , Intervalo Livre de Doença , Exoma , Feminino , Seguimentos , Estudo de Associação Genômica Ampla , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/mortalidade , Masculino , Mutação , Estadiamento de Neoplasias , Prognóstico , Modelos de Riscos Proporcionais
2.
Ann Oncol ; 28(1): 83-89, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28177435

RESUMO

Background: Lung squamous cell carcinoma (LUSC) accounts for 20­30% of non-small cell lung cancers (NSCLCs). There are limited treatment strategies for LUSC in part due to our inadequate understanding of the molecular underpinnings of the disease. We performed whole-exome sequencing (WES) and comprehensive immune profiling of a unique set of clinically annotated early-stage LUSCs to increase our understanding of the pathobiology of this malignancy. Methods: Matched pairs of surgically resected stage I-III LUSCs and normal lung tissues (n = 108) were analyzed by WES. Immunohistochemistry and image analysis-based profiling of 10 immune markers were done on a subset of LUSCs (n = 91). Associations among mutations, immune markers and clinicopathological variables were statistically examined using analysis of variance and Fisher's exact test. Cox proportional hazards regression models were used for statistical analysis of clinical outcome. Results: This early-stage LUSC cohort displayed an average of 209 exonic mutations per tumor. Fourteen genes exhibited significant enrichment for somatic mutation: TP53, MLL2, PIK3CA, NFE2L2, CDH8, KEAP1, PTEN, ADCY8, PTPRT, CALCR, GRM8, FBXW7, RB1 and CDKN2A. Among mutated genes associated with poor recurrence-free survival, MLL2 mutations predicted poor prognosis in both TP53 mutant and wild-type LUSCs. We also found that in treated patients, FBXW7 and KEAP1 mutations were associated with poor response to adjuvant therapy, particularly in TP53-mutant tumors. Analysis of mutations with immune markers revealed that ADCY8 and PIK3CA mutations were associated with markedly decreased tumoral PD-L1 expression, LUSCs with PIK3CA mutations exhibited elevated CD45ro levels and CDKN2A-mutant tumors displayed an up-regulated immune response. Conclusion(s): Our findings pinpoint mutated genes that may impact clinical outcome as well as personalized strategies for targeted immunotherapies in early-stage LUSC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Mutação , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/terapia , Estudos de Coortes , Seguimentos , Humanos , Imuno-Histoquímica , Imunofenotipagem , Neoplasias Pulmonares/patologia , Estadiamento de Neoplasias , Medicina de Precisão , Sequenciamento do Exoma
3.
Mol Psychiatry ; 20(11): 1350-65, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25385366

RESUMO

An increasing number of genetic variants have been implicated in autism spectrum disorders (ASDs), and the functional study of such variants will be critical for the elucidation of autism pathophysiology. Here, we report a de novo balanced translocation disruption of TRPC6, a cation channel, in a non-syndromic autistic individual. Using multiple models, such as dental pulp cells, induced pluripotent stem cell (iPSC)-derived neuronal cells and mouse models, we demonstrate that TRPC6 reduction or haploinsufficiency leads to altered neuronal development, morphology and function. The observed neuronal phenotypes could then be rescued by TRPC6 complementation and by treatment with insulin-like growth factor-1 or hyperforin, a TRPC6-specific agonist, suggesting that ASD individuals with alterations in this pathway may benefit from these drugs. We also demonstrate that methyl CpG binding protein-2 (MeCP2) levels affect TRPC6 expression. Mutations in MeCP2 cause Rett syndrome, revealing common pathways among ASDs. Genetic sequencing of TRPC6 in 1041 ASD individuals and 2872 controls revealed significantly more nonsynonymous mutations in the ASD population, and identified loss-of-function mutations with incomplete penetrance in two patients. Taken together, these findings suggest that TRPC6 is a novel predisposing gene for ASD that may act in a multiple-hit model. This is the first study to use iPSC-derived human neurons to model non-syndromic ASD and illustrate the potential of modeling genetically complex sporadic diseases using such cells.


Assuntos
Transtorno Autístico/patologia , Neurônios/patologia , Canais de Cátion TRPC/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Transtorno Autístico/genética , Transtorno Autístico/fisiopatologia , Carboplatina/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/genética , Células Cultivadas , Criança , Modelos Animais de Doenças , Embrião de Mamíferos , Etoposídeo/metabolismo , Regulação da Expressão Gênica/genética , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/fisiologia , Potenciais Pós-Sinápticos Inibidores/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitoxantrona/metabolismo , Mutação/genética , Neurônios/metabolismo , Prednisolona/metabolismo , Transdução de Sinais/genética , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6
4.
Genes Immun ; 14(5): 310-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23615072

RESUMO

The Ashkenazi Jewish population has a several-fold higher prevalence of Crohn's disease (CD) compared with non-Jewish European ancestry populations and has a unique genetic history. Haplotype association is critical to CD etiology in this population, most notably at NOD2, in which three causal, uncommon and conditionally independent NOD2 variants reside on a shared background haplotype. We present an analysis of extended haplotypes that showed significantly greater association to CD in the Ashkenazi Jewish population compared with a non-Jewish population (145 haplotypes and no haplotypes with P-value <10(-3), respectively). Two haplotype regions, one each on chromosomes 16 and 21, conferred increased disease risk within established CD loci. We performed exome sequencing of 55 Ashkenazi Jewish individuals and follow-up genotyping focused on variants in these two regions. We observed Ashkenazi Jewish-specific nominal association at R755C in TRPM2 on chromosome 21. Within the chromosome 16 region, R642S of HEATR3 and rs9922362 of BRD7 showed genome-wide significance. Expression studies of HEATR3 demonstrated a positive role in NOD2-mediated NF-κB signaling. The BRD7 signal showed conditional dependence with only the downstream rare CD-causal variants in NOD2, but not with the background haplotype; this elaborates NOD2 as a key illustration of synthetic association.


Assuntos
Doença de Crohn/genética , Judeus/genética , Mutação de Sentido Incorreto , NF-kappa B/genética , Proteínas/genética , Transdução de Sinais/genética , Proteínas Cromossômicas não Histona/genética , Cromossomos Humanos Par 16/genética , Éxons/genética , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Genótipo , Células HEK293 , Haplótipos , Humanos , Modelos Logísticos , Proteína Adaptadora de Sinalização NOD2/genética , Polimorfismo de Nucleotídeo Único , Interferência de RNA , Análise de Sequência de DNA
6.
Nat Genet ; 1(1): 72-5, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1338766

RESUMO

The angiotensin converting enzyme (ACE) is a key component of the renin angiotensin system that contributes to the regulation of blood pressure (BP). Recent demonstration of linkage between the ACE locus and elevated BP in a rat model of hypertension has further emphasized ACE as a candidate gene in human hypertension. We report the localization of the ACE gene on the genetic map of chromosome 17, and identify an extremely polymorphic marker at the human growth hormone (hGH) locus which shows no recombination with ACE. We have found no evidence to support linkage between the ACE locus and hypertension, which suggests that mutations at the ACE locus do not commonly contribute to the pathogenesis of hypertension in our test population.


Assuntos
Ligação Genética , Hipertensão/genética , Peptidil Dipeptidase A/genética , Sequência de Bases , Mapeamento Cromossômico , Cromossomos Humanos Par 17 , DNA/genética , Feminino , Marcadores Genéticos , Hormônio do Crescimento/genética , Humanos , Hipertensão/enzimologia , Masculino , Dados de Sequência Molecular , Linhagem
7.
Nat Genet ; 11(1): 76-82, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7550319

RESUMO

Sensitivity of blood pressure to dietary salt is a common feature in subjects with hypertension. These features are exemplified by the mendelian disorder, Liddle's syndrome, previously shown to arise from constitutive activation of the renal epithelial sodium channel due to mutation in the beta subunit of this channel. We now demonstrate that this disease can also result from a mutation truncating the carboxy terminus of the gamma subunit of this channel; this truncated subunit also activates channel activity. These findings demonstrate genetic heterogeneity of Liddle's syndrome, indicate independent roles of beta and gamma subunits in the negative regulation of channel activity, and identify a new gene in which mutation causes a salt-sensitive form of human hypertension.


Assuntos
Hipertensão/genética , Ativação do Canal Iônico/genética , Canais de Sódio/genética , Sódio na Dieta/efeitos adversos , Adolescente , Adulto , Aldosterona/deficiência , Alelos , Sequência de Aminoácidos , Animais , Sequência de Bases , Códon/genética , Canais Epiteliais de Sódio , Regulação da Expressão Gênica , Genes , Genes Dominantes , Humanos , Hipertensão/induzido quimicamente , Hipertensão/classificação , Hipertensão/metabolismo , Hipopotassemia/genética , Túbulos Renais Proximais/metabolismo , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Oócitos/metabolismo , Linhagem , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Renina/deficiência , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Canais de Sódio/deficiência , Canais de Sódio/fisiologia , Síndrome , Regiões Terminadoras Genéticas , Xenopus laevis
8.
Nat Genet ; 14(2): 152-6, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8841184

RESUMO

Mutations in the Na-K-2Cl cotransporter (NKCC2), a mediator of renal salt reabsorption, cause Bartter's syndrome, featuring salt wasting, hypokalaemic alkalosis, hypercalciuria and low blood pressure. NKCC2 mutations can be excluded in some Bartter's kindreds, prompting examination of regulators of cotransporter activity. One regulator is believed to be ROMK, an ATP-sensitive K+ channel that 'recycles' reabsorbed K+ back to the tubule lumen. Examination of the ROMK gene reveals mutations that co-segregate with the disease and disrupt ROMK function in four Bartter's kindreds. Our findings establish the genetic heterogeneity of Bartter's syndrome, and demonstrate the physiologic role of ROMK in vivo.


Assuntos
Síndrome de Bartter/genética , Heterogeneidade Genética , Mutação , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/genética , Sequência de Aminoácidos , Proteínas de Transporte/genética , Membrana Celular/química , Consanguinidade , Sequência Conservada , Análise Mutacional de DNA , Feminino , Genótipo , Humanos , Masculino , Linhagem , Polimorfismo Conformacional de Fita Simples , Canais de Potássio/química , Simportadores de Cloreto de Sódio-Potássio
9.
Nat Genet ; 13(2): 183-8, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8640224

RESUMO

Inherited hypokalaemic alkalosis with low blood pressure can be divided into two groups-Gitelman's syndrome, featuring hypocalciuria, hypomagnesaemia and milder clinical manifestations, and Bartter's syndrome, featuring hypercalciuria and early presentation with severe volume depletion. Mutations in the renal Na-Cl cotransporter have been shown to cause Gitelman's syndrome. We demonstrate linkage of Bartter's syndrome to the renal Na-K-2Cl cotransporter gene NKCC2, and identify frameshift or non-conservative missense mutations for this gene that co-segregate with the disease. These findings demonstrate the molecular basis of Bartter's syndrome, provide the basis for molecular classification of patients with inherited hypokalaemic alkalosis, and suggest potential phenotypes in heterozygous carriers of NKCC2 mutations.


Assuntos
Síndrome de Bartter/genética , Proteínas de Transporte/genética , Mutação , Sequência de Aminoácidos , Síndrome de Bartter/etiologia , Sequência de Bases , Proteínas de Transporte/química , Clonagem Molecular , Consanguinidade , DNA Complementar , Feminino , Marcadores Genéticos , Homozigoto , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Análise de Sequência de DNA , Simportadores de Cloreto de Sódio-Potássio
10.
Nat Genet ; 19(3): 279-81, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9662404

RESUMO

Pseudohypoaldosteronism type I (PHA1) is characterized by neonatal renal salt wasting with dehydration, hypotension, hyperkalaemia and metabolic acidosis, despite elevated aldosterone levels. Two forms of PHA1 exist. An autosomal recessive form features severe disease with manifestations persisting into adulthood. This form is caused by loss-of-function mutations in genes encoding subunits of the amiloride-sensitive epithelial sodium channel (ENaC; refs 2,3). Autosomal dominant or sporadic PHA1 is a milder disease that remits with age. Among six dominant and seven sporadic PHA1 kindreds, we have found no ENaC gene mutations, implicating mutations in other genes. As ENaC activity in the kidney is regulated by the steroid hormone aldosterone acting through the mineralocorticoid receptor, we have screened the mineralocorticoid receptor gene (MLR) for variants and have identified heterozygous mutations in one sporadic and four dominant kindreds. These include two frameshift mutations (one a de novo mutation), two premature termination codons and one splice donor mutation. These mutations segregate with PHA1 and are not found in unaffected subjects. These findings demonstrate that heterozygous MLR mutations cause PHA1, underscore the important role of mineralocorticoid receptor function in regulation of salt and blood pressure homeostasis in humans and motivate further study of this gene for a potential role in blood pressure variation.


Assuntos
Genes Dominantes , Mutação , Pseudo-Hipoaldosteronismo/genética , Receptores de Mineralocorticoides/genética , Sequência de Bases , DNA Complementar , Feminino , Mutação da Fase de Leitura , Variação Genética , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Polimorfismo Conformacional de Fita Simples
11.
Nat Genet ; 12(1): 24-30, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8528245

RESUMO

Maintenance of fluid and electrolyte homeostasis is critical for normal neuromuscular function. Bartter's syndrome is an autosomal recessive disease characterized by diverse abnormalities in electrolyte homeostasis including hypokalaemic metabolic alkalosis; Gitelman's syndrome represents the predominant subset of Bartter's patients having hypomagnesemia and hypocalciuria. We now demonstrate complete linkage of Gitelman's syndrome to the locus encoding the renal thiazide-sensitive Na-Cl cotransporter, and identify a wide variety of non-conservative mutations, consistent with loss of function alleles, in affected subjects. These findings demonstrate the molecular basis of Gitelman's syndrome. We speculate that these mutant alleles lead to reduced sodium chloride reabsorption in the more common heterozygotes, potentially protecting against development of hypertension.


Assuntos
Síndrome de Bartter/genética , Proteínas de Transporte/genética , Cloretos/metabolismo , Receptores de Droga/genética , Sódio/metabolismo , Simportadores , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico , Cromossomos Humanos Par 16 , Clonagem Molecular , Primers do DNA/química , Repetições de Dinucleotídeos , Feminino , Linguado , Ligação Genética , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Mutação Puntual , Polimorfismo Conformacional de Fita Simples , Ratos , Alinhamento de Sequência , Simportadores de Cloreto de Sódio , Membro 3 da Família 12 de Carreador de Soluto
12.
Nat Genet ; 2(1): 66-74, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1303253

RESUMO

Patients with glucocorticoid-remediable aldosteronism (GRA) from 12 kindreds possess chimaeric gene duplications arising from unequal crossing-over, fusing regulatory sequences of steroid 11 beta-hydroxylase to coding sequences of aldosterone synthase. These chimaeric genes are specific for GRA and explain the biochemistry, physiology and genetics of this form of hypertension. Sites of crossing over range from intron 2 to intron 4. Most mutations have arisen independently from either sister or non-sister chromatid exchange between these genes, which are only 45 kilobases apart. The possibility of a susceptibility allele for GRA of Irish origin is suggested. These findings indicate the utility of a direct genetic test for this disorder.


Assuntos
Quimera/genética , Sistema Enzimático do Citocromo P-450/genética , Hipertensão/genética , Família Multigênica , Esteroide Hidroxilases/genética , Alelos , Sequência de Bases , Troca Genética , Citocromo P-450 CYP11B2 , DNA/genética , Feminino , Expressão Gênica , Haplótipos/genética , Humanos , Desequilíbrio de Ligação , Masculino , Dados de Sequência Molecular , Linhagem , Esteroide 11-beta-Hidroxilase/genética
13.
Nat Genet ; 16(2): 202-5, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9171836

RESUMO

Essential hypertension is a common multifactorial trait. The molecular basis of a number of rare diseases that after blood pressure in humans has been established, identifying pathways that may be involved in more common forms of hypertension. Pseudohypoaldosteronism type II (PHAII, also known as familial hyperkalaemia and hypertension or Gordon's syndrome; OMIM #145260), is characterized by hyperkalaemia despite normal renal glomerular filtration, hypertension and correction of physiologic abnormalities by thiazide diuretics. Mild hyperchloremia, metabolic acidosis and suppressed plasma renin activity are variable associated findings. The pathogenesis of PHAII is unknown, although clinical studies indicate an abnormality in renal ion transport. As thiazide diuretics are among the most efficacious agents in the treatment of essential hypertension, understanding the pathogenesis of PHAII may be of relevance to more common forms of hypertension. Analysis of linkage in eight PHAII families showing autosomal dominant transmission demonstrates locus heterogeneity of this trait, with a multilocus lod score of 8.1 for linkage of PHAII to chromosomes 1q31-q42 and 17p11-q21. Interestingly, the chromosome-17 locus overlaps a syntenic interval in rat that contains a blood pressure quantitative trait locus (QTL). Our findings provide a first step toward identification of the molecular basis of PHAII.


Assuntos
Cromossomos Humanos Par 17 , Cromossomos Humanos Par 1 , Ligação Genética , Hiperpotassemia/genética , Hipertensão/genética , Pseudo-Hipoaldosteronismo/genética , Animais , Mapeamento Cromossômico , Feminino , Humanos , Hiperpotassemia/complicações , Hipertensão/complicações , Masculino , Linhagem , Pseudo-Hipoaldosteronismo/complicações , Ratos
14.
Nat Genet ; 26(1): 71-5, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10973252

RESUMO

The multi-subunit H+-ATPase pump is present at particularly high density on the apical (luminal) surface of -intercalated cells of the cortical collecting duct of the distal nephron, where vectorial proton transport is required for urinary acidification. The complete subunit composition of the apical ATPase, however, has not been fully agreed upon. Functional failure of -intercalated cells results in a group of disorders, the distal renal tubular acidoses (dRTA), whose features include metabolic acidosis accompanied by disturbances of potassium balance, urinary calcium solubility, bone physiology and growth. Mutations in the gene encoding the B-subunit of the apical pump (ATP6B1) cause dRTA accompanied by deafness. We previously localized a gene for dRTA with preserved hearing to 7q33-34 (ref. 4). We report here the identification of this gene, ATP6N1B, which encodes an 840 amino acid novel kidney-specific isoform of ATP6N1A, the 116-kD non-catalytic accessory subunit of the proton pump. Northern-blot analysis demonstrated ATP6N1B expression in kidney but not other main organs. Immunofluorescence studies in human kidney cortex revealed that ATP6N1B localizes almost exclusively to the apical surface of -intercalated cells. We screened nine dRTA kindreds with normal audiometry that linked to the ATP6N1B locus, and identified different homozygous mutations in ATP6N1B in eight. These include nonsense, deletion and splice-site changes, all of which will truncate the protein. Our findings identify a new kidney-specific proton pump 116-kD accessory subunit that is highly expressed in proton-secreting cells in the distal nephron, and illustrate its essential role in normal vectorial acid transport into the urine by the kidney.


Assuntos
Acidose Tubular Renal/genética , Audição/genética , Mutação , Proteínas da Gravidez , Bombas de Próton/química , Bombas de Próton/genética , ATPases Translocadoras de Prótons , Fatores Supressores Imunológicos , Acidose Tubular Renal/metabolismo , Acidose Tubular Renal/urina , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Adolescente , Adulto , Sequência de Aminoácidos , Audiometria , Northern Blotting , Encéfalo/metabolismo , Criança , Pré-Escolar , Cromossomos Humanos Par 7 , Mapeamento de Sequências Contíguas , DNA Complementar/metabolismo , Éxons , Feminino , Deleção de Genes , Genes Recessivos , Ligação Genética , Marcadores Genéticos , Audição/fisiologia , Homozigoto , Humanos , Rim/metabolismo , Rim/patologia , Córtex Renal/metabolismo , Masculino , Microscopia de Fluorescência , ATPases Mitocondriais Próton-Translocadoras , Modelos Genéticos , Dados de Sequência Molecular , Linhagem , Mapeamento Físico do Cromossomo , Polimorfismo Genético , Polimorfismo Conformacional de Fita Simples , Biossíntese de Proteínas , Isoformas de Proteínas , Bombas de Próton/biossíntese , Splicing de RNA , Recombinação Genética , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , ATPases Vacuolares Próton-Translocadoras
15.
Nat Genet ; 12(3): 248-53, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8589714

RESUMO

Autosomal recessive pseudohypoaldosteronism type I is a rare life-threatening disease characterized by severe neonatal salt wasting, hyperkalaemia, metabolic acidosis, and unresponsiveness to mineralocorticoid hormones. Investigation of affected offspring of consanguineous union reveals mutations in either the alpha or beta subunits of the amiloride-sensitive epithelial sodium channel in five kindreds. These mutations are homozygous in affected subjects, co-segregate with the disease, and introduce frameshift, premature termination or missense mutations that result in loss of channel activity. These findings demonstrate the molecular basis and explain the pathophysiology of this disease.


Assuntos
Mutação , Pseudo-Hipoaldosteronismo/genética , Canais de Sódio/genética , Animais , Sequência de Bases , DNA , Canais Epiteliais de Sódio , Epitélio/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Dados de Sequência Molecular , Linhagem , Pseudo-Hipoaldosteronismo/classificação , Ratos , Canais de Sódio/metabolismo
16.
Nat Genet ; 26(3): 354-7, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11062479

RESUMO

End-stage renal disease (ESRD) is a major public health problem, affecting 1 in 1,000 individuals and with an annual death rate of 20% despite dialysis treatment. IgA nephropathy (IgAN) is the most common form of glomerulonephritis, a principal cause of ESRD worldwide; it affects up to 1.3% of the population and its pathogenesis is unknown. Kidneys of people with IgAN show deposits of IgA-containing immune complexes with proliferation of the glomerular mesangium (Fig. 1). Typical clinical features include onset before age 40 with haematuria and proteinuria (blood and protein in the urine), and episodes of gross haematuria following mucosal infections are common; 30% of patients develop progressive renal failure. Although not generally considered a hereditary disease, striking ethnic variation in prevalence and familial clustering, along with subclinical renal abnormalities among relatives of IgAN cases, have suggested a heretofore undefined genetic component. By genome-wide analysis of linkage in 30 multiplex IgAN kindreds, we demonstrate linkage of IgAN to 6q22-23 under a dominant model of transmission with incomplete penetrance, with a lod score of 5.6 and 60% of kindreds linked. These findings for the first time indicate the existence of a locus with large effect on development of IgAN and identify the chromosomal location of this disease gene.


Assuntos
Cromossomos Humanos Par 6/genética , Glomerulonefrite por IGA/genética , Mapeamento Cromossômico , Feminino , Genes Dominantes , Predisposição Genética para Doença , Genótipo , Humanos , Itália , Escore Lod , Masculino , Linhagem , Estados Unidos
17.
Nat Genet ; 17(2): 171-8, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9326936

RESUMO

Analysis of patients with inherited hypokalaemic alkalosis resulting from salt-wasting has proved fertile ground for identification of essential elements of renal salt homeostasis and blood-pressure regulation. We now demonstrate linkage of this phenotype to a segment of chromosome 1 containing the gene encoding a renal chloride channel, CLCNKB. Examination of this gene reveals loss-of-function mutations that impair renal chloride reabsorption in the thick ascending limb of Henle's loop. Mutations in seventeen kindreds have been identified, and they include large deletions and nonsense and missense mutations. Some of the deletions are shown to have arisen by unequal crossing over between CLCNKB and the nearby related gene, CLCNKA. Patients who harbour CLCNKB mutations are characterized by hypokalaemic alkalosis with salt-wasting, low blood pressure, normal magnesium and hyper- or normocalciuria; they define a distinct subset of patients with Bartter's syndrome in whom nephrocalcinosis is absent. These findings demonstrate the critical role of CLCNKB in renal salt reabsorption and blood-pressure homeostasis, and demonstrate the potential role of specific CLCNKB antagonists as diuretic antihypertensive agents.


Assuntos
Síndrome de Bartter/genética , Canais de Cloreto/genética , Mutação , Síndrome de Bartter/classificação , Síndrome de Bartter/metabolismo , Sequência de Bases , Canais de Cloreto/química , Canais de Cloreto/metabolismo , Cromossomos Humanos Par 1/genética , Troca Genética , Primers do DNA/genética , Éxons , Feminino , Ligação Genética , Humanos , Íntrons , Alça do Néfron/metabolismo , Masculino , Linhagem , Fenótipo , Reação em Cadeia da Polimerase , Deleção de Sequência
18.
Nat Genet ; 21(1): 84-90, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9916796

RESUMO

H+-ATPases are ubiquitous in nature; V-ATPases pump protons against an electrochemical gradient, whereas F-ATPases reverse the process, synthesizing ATP. We demonstrate here that mutations in ATP6B1, encoding the B-subunit of the apical proton pump mediating distal nephron acid secretion, cause distal renal tubular acidosis, a condition characterized by impaired renal acid secretion resulting in metabolic acidosis. Patients with ATP6B1 mutations also have sensorineural hearing loss; consistent with this finding, we demonstrate expression of ATP6B1 in cochlea and endolymphatic sac. Our data, together with the known requirement for active proton secretion to maintain proper endolymph pH, implicate ATP6B1 in endolymph pH homeostasis and in normal auditory function. ATP6B1 is the first member of the H+-ATPase gene family in which mutations are shown to cause human disease.


Assuntos
Acidose Tubular Renal/enzimologia , Cromossomos Humanos Par 2 , Perda Auditiva Neurossensorial/enzimologia , Mutação , ATPases Translocadoras de Prótons/genética , Acidose Tubular Renal/complicações , Acidose Tubular Renal/genética , Sequência de Bases , Pré-Escolar , Cóclea/metabolismo , Feminino , Genes Recessivos , Ligação Genética , Perda Auditiva Neurossensorial/complicações , Perda Auditiva Neurossensorial/genética , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Linhagem , ATPases Translocadoras de Prótons/metabolismo
20.
Curr Opin Cell Biol ; 10(4): 450-4, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9719864

RESUMO

The successful merging of modern molecular genetics with basic renal physiology is exemplified by the recent description of the molecular basis of two classic diseases of clinical nephrology; Bartter's and Gitelman's syndromes of inherited hypokalemic alkalosis. Mutations in four different genes have been identified, each of which causes hypokalemic alkalosis, salt wasting and hypotension. These genetic studies have greatly advanced our understanding of renal physiology.


Assuntos
Síndrome de Bartter/genética , Proteínas de Transporte/genética , Mutação , Humanos , Rim , Simportadores de Cloreto de Sódio-Potássio , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA