Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
3.
Ann Oncol ; 28(1): 83-89, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28177435

RESUMEN

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.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Mutación , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Estudios de Cohortes , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Inmunofenotipificación , Neoplasias Pulmonares/patología , Estadificación de Neoplasias , Medicina de Precisión , Secuenciación del Exoma
4.
Ann Oncol ; 28(1): 75-82, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27687306

RESUMEN

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.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Adenocarcinoma/mortalidad , Adenocarcinoma del Pulmón , Biomarcadores de Tumor/análisis , Análisis Mutacional de ADN , Supervivencia sin Enfermedad , Exoma , Femenino , Estudios de Seguimiento , Estudio de Asociación del Genoma Completo , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Masculino , Mutación , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales
5.
Mol Psychiatry ; 20(11): 1350-65, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25385366

RESUMEN

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.


Asunto(s)
Trastorno Autístico/patología , Neuronas/patología , Canales Catiónicos TRPC/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Carboplatino/metabolismo , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/genética , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Embrión de Mamíferos , Etopósido/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas/fisiología , Potenciales Postsinápticos Inhibidores/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitoxantrona/metabolismo , Mutación/genética , Neuronas/metabolismo , Prednisolona/metabolismo , Transducción de Señal/genética , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6
7.
Genes Immun ; 14(5): 310-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23615072

RESUMEN

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.


Asunto(s)
Enfermedad de Crohn/genética , Judíos/genética , Mutación Missense , FN-kappa B/genética , Proteínas/genética , Transducción de Señal/genética , Proteínas Cromosómicas no Histona/genética , Cromosomas Humanos Par 16/genética , Exones/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Genotipo , Células HEK293 , Haplotipos , Humanos , Modelos Logísticos , Proteína Adaptadora de Señalización NOD2/genética , Polimorfismo de Nucleótido Simple , Interferencia de ARN , Análisis de Secuencia de ADN
9.
Kidney Int ; 70(10): 1706-16, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16985514

RESUMEN

Final urinary acidification is achieved by electrogenic vacuolar H(+)-ATPases expressed in acid-secretory intercalated cells (ICs) in the connecting tubule (CNT) and the cortical (CCD) and initial medullary collecting duct (MCD), respectively. Electrogenic Na(+) reabsorption via epithelial Na(+) channels (ENaCs) in the apical membrane of the segment-specific CNT and collecting duct cells may promote H(+)-ATPases-mediated proton secretion by creating a more lumen-negative voltage. The exact localization where this supposed functional interaction takes place is unknown. We used several mouse models performing renal clearance experiments and assessed the furosemide-induced urinary acidification. Increasing Na(+) delivery to the CNT and CCD by blocking Na(+) reabsorption in the thick ascending limb with furosemide enhanced urinary acidification and net acid excretion. This effect of furosemide was abolished with amiloride or benzamil blocking ENaC action. In mice deficient for the IC-specific B1 subunit of the vacuolar H(+)-ATPase, furosemide led to only a small urinary acidification. In contrast, in mice with a kidney-specific inactivation of the alpha subunit of ENaC in the CCD and MCD, but not in the CNT, furosemide alone and in combination with hydrochlorothiazide induced normal urinary acidification. These results suggest that the B1 vacuolar H(+)-ATPase subunit is necessary for the furosemide-induced acute urinary acidification. Loss of ENaC channels in the CCD and MCD does not affect this acidification. Thus, functional expression of ENaC channels in the CNT is sufficient for furosemide-stimulated urinary acidification and identifies the CNT as a major segment in electrogenic urinary acidification.


Asunto(s)
Equilibrio Ácido-Base/efectos de los fármacos , Diuréticos/farmacología , Furosemida/farmacología , Túbulos Renales Distales/efectos de los fármacos , ATPasas de Translocación de Protón/metabolismo , Equilibrio Ácido-Base/fisiología , Amilorida/farmacocinética , Amilorida/farmacología , Animales , Diuréticos/farmacocinética , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Furosemida/farmacocinética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Hidroclorotiazida/farmacocinética , Hidroclorotiazida/farmacología , Concentración de Iones de Hidrógeno , Túbulos Renales Colectores/metabolismo , Túbulos Renales Distales/metabolismo , Tasa de Depuración Metabólica/efectos de los fármacos , Tasa de Depuración Metabólica/fisiología , Ratones , Ratones Noqueados , Nefronas/efectos de los fármacos , Nefronas/fisiología , ATPasas de Translocación de Protón/genética , Equilibrio Hidroelectrolítico/fisiología
11.
J Biol Chem ; 276(45): 42382-8, 2001 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-11495928

RESUMEN

The 116-kDa a-subunit of the vacuolar proton pump (H(+)-ATPase) exists as several isoforms encoded by different genes and with different patterns of tissue expression. Its function within the multisubunit pump complex has yet to be elucidated. To date, three isoforms have been identified in mouse (designated a1-a3). We now report the cloning and characterization of Atp6n1b, encoding a novel fourth murine isoform (a4). Murine a4 has 833 residues and shows 85% amino acid identity to the human kidney-specific ATP6N1B protein in which loss-of-function alterations cause autosomal recessive distal renal tubular acidosis. The human and murine genes have similar genomic organization; furthermore, Atp6n1b maps to a region of mouse chromosome 6 that is syntenic with the segment of human 7q33-34 containing ATP6N1B. Together these findings establish the two genes as orthologs. The mouse a4 protein is 61, 52, and 47% identical to a1, a2, and a3, respectively. Phylogenetic analysis confirms that among vertebrates there are four a-subunit families, with a4 most resembling a1. Northern blot analysis of Atp6n1b reveals a 3.7-kilobase a4 transcript in kidney but not other major organs, and a reverse transcription polymerase chain reaction in 12 mouse tissues detects expression in kidney alone. Immunofluorescence studies in murine kidney demonstrate high intensity a4 staining at the surface of intercalated cells, with additional expression in the proximal tubule (not previously reported in human kidney). Similar apical a4 immunostaining is also present in male genital tissue. Identification of this novel murine kidney-enriched 116-kDa a-subunit provides a molecular tool for investigation of the currently unknown role of this protein, which is essential for proper function of the apical renal vacuolar H(+)-ATPase in man.


Asunto(s)
Proteínas Gestacionales , Bombas de Protones/genética , ATPasas de Translocación de Protón , Factores Supresores Inmunológicos , Secuencia de Aminoácidos , Animales , Acuaporina 2 , Acuaporina 6 , Acuaporinas/análisis , Mapeo Cromosómico , Clonación Molecular , Ratones , Datos de Secuencia Molecular , Filogenia , Polimorfismo Genético , Subunidades de Proteína , Bombas de Protones/análisis , Bombas de Protones/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ATPasas de Translocación de Protón Vacuolares
12.
Science ; 293(5532): 1107-12, 2001 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-11498583

RESUMEN

Hypertension is a major public health problem of largely unknown cause. Here, we identify two genes causing pseudohypoaldosteronism type II, a Mendelian trait featuring hypertension, increased renal salt reabsorption, and impaired K+ and H+ excretion. Both genes encode members of the WNK family of serine-threonine kinases. Disease-causing mutations in WNK1 are large intronic deletions that increase WNK1 expression. The mutations in WNK4 are missense, which cluster in a short, highly conserved segment of the encoded protein. Both proteins localize to the distal nephron, a kidney segment involved in salt, K+, and pH homeostasis. WNK1 is cytoplasmic, whereas WNK4 localizes to tight junctions. The WNK kinases and their associated signaling pathway(s) may offer new targets for the development of antihypertensive drugs.


Asunto(s)
Hipertensión/genética , Mutación , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/genética , Secuencia de Aminoácidos , Secuencia de Bases , Mapeo Cromosómico , Cromosomas Humanos Par 12/genética , Cromosomas Humanos Par 17/genética , Citoplasma/enzimología , Femenino , Regulación Enzimológica de la Expresión Génica , Ligamiento Genético , Humanos , Hipertensión/enzimología , Hipertensión/fisiopatología , Uniones Intercelulares/enzimología , Péptidos y Proteínas de Señalización Intracelular , Intrones , Túbulos Renales Colectores/enzimología , Túbulos Renales Colectores/ultraestructura , Túbulos Renales Distales/enzimología , Túbulos Renales Distales/ultraestructura , Masculino , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Mutación Missense , Linaje , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/enzimología , Seudohipoaldosteronismo/fisiopatología , Eliminación de Secuencia , Transducción de Señal , Proteína Quinasa Deficiente en Lisina WNK 1 , Proteína de la Zonula Occludens-1
13.
Hypertension ; 37(6): 1458-64, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11408395

RESUMEN

The relationship between salt homeostasis and blood pressure has remained difficult to establish from epidemiological studies of the general population. Recently, mendelian forms of hypertension have demonstrated that mutations that increase renal salt balance lead to higher blood pressure, suggesting that mutations that decrease the net salt balance might have the converse effect. Gitelman's syndrome, caused by loss of function mutations in the Na-Cl cotransporter of the distal convoluted tubule (NCCT), features inherited hypokalemic alkalosis with so-called "normal" blood pressure. We hypothesized that the mild salt wasting of Gitelman's syndrome results in reduced blood pressure and protection from hypertension. We have formally addressed this question through the study of 199 members of a large Amish kindred with Gitelman's syndrome. Through genetic testing, family members were identified as inheriting 0 (n=60), 1 (n=113), or 2 (n=26) mutations in NCCT, permitting an unbiased assessment of the clinical consequences of inheriting these mutations by comparison of the phenotypes of relatives with contrasting genotypes. The results demonstrate high penetrance of hypokalemic alkalosis, hypomagnesemia, and hypocalciuria in patients inheriting 2 mutant NCCT alleles. In addition, the NCCT genotype was a significant predictor of blood pressure, with homozygous mutant family members having significantly lower age- and gender-adjusted systolic and diastolic blood pressures than those of their wild-type relatives. Moreover, both homozygote and heterozygote subjects had significantly higher 24-hour urinary Na(+) than did wild-type subjects, reflecting a self-selected higher salt intake. Finally, heterozygous children, but not adults, had significantly lower blood pressures than those of the wild-type relatives. These findings provide formal demonstration that inherited mutations that impair renal salt handling lower blood pressure in humans.


Asunto(s)
Presión Sanguínea/genética , Proteínas Portadoras/genética , Mutación , Sodio/metabolismo , Desequilibrio Hidroelectrolítico/genética , Alcalosis/genética , Bicarbonatos/sangre , Calcio/orina , Efecto Fundador , Genotipo , Heterocigoto , Humanos , Hiperpotasemia/genética , Hipertensión/genética , Magnesio/orina , Linaje , Sodio/orina , Simportadores de Cloruro de Sodio-Potasio , Síndrome
15.
Kidney Int ; 59(2): 710-7, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11168953

RESUMEN

BACKGROUND: Gitelman's syndrome (GS), also called Gitelman's variant of Bartter's syndrome, is an autosomal recessive renal disorder characterized by hypokalemia, hypomagnesemia, metabolic alkalosis, and hypocalciuria. GS is caused by inactivating mutations in the thiazide-sensitive sodium chloride cotransporter gene (NCCT). It is also known as the "milder" form of Bartter's syndrome, as patients with GS are usually diagnosed in adulthood during routine investigation. Symptoms reported in the literature range from asymptomatic, to mild symptoms of cramps and fatigue, to severe manifestations such as tetany, paralysis, and rhabdomyolysis. This is the first systematic evaluation of a large group of patients with genetically defined GS. METHODS: We evaluated the symptoms and quality of life (QOL) in 50 adult GS patients with confirmed mutations in NCCT, using a standardized questionnaire. This cohort was compared with 25 age- and sex-matched controls. RESULTS: GS patients were significantly more symptomatic than controls. The most common symptoms were salt craving, with musculoskeletal symptoms such as cramps, muscle weakness, and aches and constitutional symptoms such as fatigue, generalized weakness and dizziness, and nocturia and polydipsia. Forty-five percent of GS patients consider their symptoms a moderate to big problem. Measures of health-related QOL were significantly lower in GS patients compared with controls, particularly in terms of role limitations caused by physical health, emotion, level of energy, and general health perception. CONCLUSIONS: This descriptive study indicates that GS is not an asymptomatic disease and adversely affects QOL in these patients. Further studies are needed to assess the impact of therapy on symptoms and QOL.


Asunto(s)
Síndrome de Bartter/fisiopatología , Estado de Salud , Calidad de Vida , Adolescente , Adulto , Anciano , Síndrome de Bartter/complicaciones , Femenino , Humanos , Enfermedades Renales/etiología , Masculino , Persona de Mediana Edad , Enfermedades Musculoesqueléticas/etiología
16.
Nat Genet ; 26(3): 354-7, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11062479

RESUMEN

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.


Asunto(s)
Cromosomas Humanos Par 6/genética , Glomerulonefritis por IGA/genética , Mapeo Cromosómico , Femenino , Genes Dominantes , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Italia , Escala de Lod , Masculino , Linaje , Estados Unidos
17.
Hypertension ; 36(4): 477-83, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11040222

RESUMEN

Hypertension is a leading cause of morbidity and mortality. Efforts to identify hypertension genes have focused on 3 approaches: mendelian disorders, candidate genes, and genome-wide scans. Thus far, these efforts have not identified genes that contribute substantively to overall blood pressure (BP) variation in the community. A 10-centiMorgan (cM) density genome-wide scan was performed in the largest families from 2 generations of Framingham Heart Study participants. Heritability and linkage for long-term mean systolic and diastolic BP phenotypes were analyzed by use of SOLAR software. Heritability estimates were based on BP measurements in 1593 families. Genotyping was performed on 1702 subjects from 332 large families, and BP data were available for 1585 (93%) genotyped subjects who contributed 12 588 longitudinal BP observations. The mean age was 47 years, and mean BP was 127/80 (systolic/diastolic) mm Hg. Long-term systolic and diastolic BP phenotypes had high heritability estimates, 0.57 and 0.56, respectively. For systolic BP, multipoint log-of-the-odds (LOD) scores >2.0 were located on chromosome 17 at 67 cM (LOD 4.7, P=0.0000016) and 94 cM (LOD 2.2). For diastolic BP, LOD scores >2.0 were identified on chromosome 17 (74 cM, LOD 2.1) and chromosome 18 (7 cM, LOD 2.1). Using a genome-wide scan, we found strong evidence for a BP quantitative trait locus on chromosome 17. Follow-up studies are warranted to identify the gene or genes in this quantitative trait locus that influence BP. Such knowledge could extend our understanding of the genetic basis of essential hypertension and have implications for the evaluation and treatment of patients with high BP.


Asunto(s)
Presión Sanguínea/genética , Cromosomas Humanos Par 17/genética , Genes , Ligamiento Genético/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estatura , Peso Corporal , Cromosomas Humanos Par 18/genética , Estudios de Cohortes , Diástole , Femenino , Genotipo , Humanos , Escala de Lod , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Fenotipo , Estudios Prospectivos , Carácter Cuantitativo Heredable , Análisis de Regresión , Sístole
18.
Pediatr Nephrol ; 14(10-11): 970-2, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10975308

RESUMEN

We describe a patient with signs and symptoms of classic Bartter syndrome. The patient tested negative for all known genetic abnormalities associated with this tubular disorder. Proteinuria was found within 1 year after the diagnosis of Bartter syndrome. A renal biopsy performed 6 months later, when her kidney function was normal, revealed focal segmental glomerulosclerosis (FSGS). We propose a link between stimulation of the renin-angiotensin system and sclerotic changes in the glomerulus. This lesion may explain previous reports of kidney failure in patients with Bartter syndrome.


Asunto(s)
Síndrome de Bartter/complicaciones , Glomeruloesclerosis Focal y Segmentaria/etiología , Adolescente , Síndrome de Bartter/patología , Síndrome de Bartter/orina , Biopsia , Femenino , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Riñón/patología , Proteinuria/etiología
19.
Nat Genet ; 26(1): 71-5, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10973252

RESUMEN

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.


Asunto(s)
Acidosis Tubular Renal/genética , Audición/genética , Mutación , Proteínas Gestacionales , Bombas de Protones/química , Bombas de Protones/genética , ATPasas de Translocación de Protón , Factores Supresores Inmunológicos , Acidosis Tubular Renal/metabolismo , Acidosis Tubular Renal/orina , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adolescente , Adulto , Secuencia de Aminoácidos , Audiometría , Northern Blotting , Encéfalo/metabolismo , Niño , Preescolar , Cromosomas Humanos Par 7 , Mapeo Contig , ADN Complementario/metabolismo , Exones , Femenino , Eliminación de Gen , Genes Recesivos , Ligamiento Genético , Marcadores Genéticos , Audición/fisiología , Homocigoto , Humanos , Riñón/metabolismo , Riñón/patología , Corteza Renal/metabolismo , Masculino , Microscopía Fluorescente , ATPasas de Translocación de Protón Mitocondriales , Modelos Genéticos , Datos de Secuencia Molecular , Linaje , Mapeo Físico de Cromosoma , Polimorfismo Genético , Polimorfismo Conformacional Retorcido-Simple , Biosíntesis de Proteínas , Isoformas de Proteínas , Bombas de Protones/biosíntesis , Empalme del ARN , Recombinación Genética , Homología de Secuencia de Aminoácido , Distribución Tisular , ATPasas de Translocación de Protón Vacuolares
20.
Science ; 289(5476): 119-23, 2000 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-10884226

RESUMEN

Hypertension and pregnancy-related hypertension are major public health problems of largely unknown causes. We describe a mutation in the mineralocorticoid receptor (MR), S810L, that causes early-onset hypertension that is markedly exacerbated in pregnancy. This mutation results in constitutive MR activity and alters receptor specificity, with progesterone and other steroids lacking 21-hydroxyl groups, normally MR antagonists, becoming potent agonists. Structural and biochemical studies indicate that the mutation results in the gain of a van der Waals interaction between helix 5 and helix 3 that substitutes for interaction of the steroid 21-hydroxyl group with helix 3 in the wild-type receptor. This helix 5-helix 3 interaction is highly conserved among diverse nuclear hormone receptors, suggesting its general role in receptor activation.


Asunto(s)
Aldosterona/metabolismo , Hipertensión/genética , Complicaciones Cardiovasculares del Embarazo , Progesterona/metabolismo , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Adolescente , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Unión Competitiva , Dimerización , Femenino , Heterocigoto , Humanos , Hipertensión/etiología , Hipertensión/metabolismo , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Linaje , Mutación Puntual , Embarazo , Complicaciones Cardiovasculares del Embarazo/etiología , Complicaciones Cardiovasculares del Embarazo/metabolismo , Conformación Proteica , Estructura Secundaria de Proteína , Receptores de Mineralocorticoides/química , Receptores de Esteroides/química , Receptores de Esteroides/metabolismo , Esteroides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...