Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 26(4): 831-43, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25145932

RESUMEN

FSGS is a clinical disorder characterized by focal scarring of the glomerular capillary tuft, podocyte injury, and nephrotic syndrome. Although idiopathic forms of FSGS predominate, recent insights into the molecular and genetic causes of FSGS have enhanced our understanding of disease pathogenesis. Here, we report a novel missense mutation of the transcriptional regulator Wilms' Tumor 1 (WT1) as the cause of nonsyndromic, autosomal dominant FSGS in two Northern European kindreds from the United States. We performed sequential genome-wide linkage analysis and whole-exome sequencing to evaluate participants from family DUK6524. Subsequently, whole-exome sequencing and direct sequencing were performed on proband DNA from family DUK6975. We identified multiple suggestive loci on chromosomes 6, 11, and 13 in family DUK6524 and identified a segregating missense mutation (R458Q) in WT1 isoform D as the cause of FSGS in this family. The identical mutation was found in family DUK6975. The R458Q mutation was not found in 1600 control chromosomes and was predicted as damaging by in silico simulation. We depleted wt1a in zebrafish embryos and observed glomerular injury and filtration defects, both of which were rescued with wild-type but not mutant human WT1D mRNA. Finally, we explored the subcellular mechanism of the mutation in vitro. WT1(R458Q) overexpression significantly downregulated nephrin and synaptopodin expression, promoted apoptosis in HEK293 cells and impaired focal contact formation in podocytes. Taken together, these data suggest that the WT1(R458Q) mutation alters the regulation of podocyte homeostasis and causes nonsyndromic FSGS.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas de Microfilamentos/metabolismo , Proteínas WT1/genética , Adolescente , Adulto , Animales , Movimiento Celular , Supervivencia Celular , Exoma , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Células HEK293 , Humanos , Masculino , Mutación Missense , Nefrosis/etiología , Nefrosis/metabolismo , Podocitos/fisiología , Análisis de Secuencia de ADN , Proteínas WT1/deficiencia , Adulto Joven , Pez Cebra , Proteínas de Pez Cebra/deficiencia
2.
J Am Soc Nephrol ; 26(7): 1701-10, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25349203

RESUMEN

Steroid-sensitive nephrotic syndrome (SSNS) accounts for >80% of cases of nephrotic syndrome in childhood. However, the etiology and pathogenesis of SSNS remain obscure. Hypothesizing that coding variation may underlie SSNS risk, we conducted an exome array association study of SSNS. We enrolled a discovery set of 363 persons (214 South Asian children with SSNS and 149 controls) and genotyped them using the Illumina HumanExome Beadchip. Four common single nucleotide polymorphisms (SNPs) in HLA-DQA1 and HLA-DQB1 (rs1129740, rs9273349, rs1071630, and rs1140343) were significantly associated with SSNS at or near the Bonferroni-adjusted P value for the number of single variants that were tested (odds ratio, 2.11; 95% confidence interval, 1.56 to 2.86; P=1.68×10(-6) (Fisher exact test). Two of these SNPs-the missense variants C34Y (rs1129740) and F41S (rs1071630) in HLA-DQA1-were replicated in an independent cohort of children of white European ancestry with SSNS (100 cases and ≤589 controls; P=1.42×10(-17)). In the rare variant gene set-based analysis, the best signal was found in PLCG2 (P=7.825×10(-5)). In conclusion, this exome array study identified HLA-DQA1 and PLCG2 missense coding variants as candidate loci for SSNS. The finding of a MHC class II locus underlying SSNS risk suggests a major role for immune response in the pathogenesis of SSNS.


Asunto(s)
Predisposición Genética a la Enfermedad/epidemiología , Cadenas alfa de HLA-DQ/genética , Síndrome Nefrótico/epidemiología , Síndrome Nefrótico/genética , Fosfolipasa C gamma/genética , Esteroides/uso terapéutico , Distribución por Edad , Edad de Inicio , Alelos , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Femenino , Genotipo , Humanos , Incidencia , Masculino , Mutación Missense , Síndrome Nefrótico/tratamiento farmacológico , Distribución por Sexo , Sri Lanka/epidemiología
3.
J Am Soc Nephrol ; 25(9): 1991-2002, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24676636

RESUMEN

FSGS is characterized by segmental scarring of the glomerulus and is a leading cause of kidney failure. Identification of genes causing FSGS has improved our understanding of disease mechanisms and points to defects in the glomerular epithelial cell, the podocyte, as a major factor in disease pathogenesis. Using a combination of genome-wide linkage studies and whole-exome sequencing in a kindred with familial FSGS, we identified a missense mutation R431C in anillin (ANLN), an F-actin binding cell cycle gene, as a cause of FSGS. We screened 250 additional families with FSGS and found another variant, G618C, that segregates with disease in a second family with FSGS. We demonstrate upregulation of anillin in podocytes in kidney biopsy specimens from individuals with FSGS and kidney samples from a murine model of HIV-1-associated nephropathy. Overexpression of R431C mutant ANLN in immortalized human podocytes results in enhanced podocyte motility. The mutant anillin displays reduced binding to the slit diaphragm-associated scaffold protein CD2AP. Knockdown of the ANLN gene in zebrafish morphants caused a loss of glomerular filtration barrier integrity, podocyte foot process effacement, and an edematous phenotype. Collectively, these findings suggest that anillin is important in maintaining the integrity of the podocyte actin cytoskeleton.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas de Microfilamentos/genética , Mutación , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Movimiento Celular/genética , Secuencia Conservada , Proteínas Contráctiles/genética , Proteínas del Citoesqueleto/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exoma , Femenino , Técnicas de Silenciamiento del Gen , Barrera de Filtración Glomerular/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Linaje , Podocitos/metabolismo , Homología de Secuencia de Aminoácido , Regulación hacia Arriba , Pez Cebra , Proteínas de Pez Cebra/genética
4.
Am J Physiol Renal Physiol ; 306(12): F1442-50, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24740790

RESUMEN

The emerging role of the transient receptor potential cation channel isotype 6 (TRPC6) as a central contributor to various pathological processes affecting podocytes has generated interest in the development of therapeutics to modulate its function. Recent insights into the regulation of TRPC6 have revealed PKG as a potent negative modulator of TRPC6 conductance and associated signaling via its phosphorylation at two highly conserved amino acid residues: Thr(69)/Thr(70) (Thr(69) in mice and Thr(70) in humans) and Ser(321)/Ser(322) (Ser(321) in mice and Ser(322) in humans). Here, we tested the role of PKG in modulating TRPC6-dependent responses in primary and conditionally immortalized mouse podocytes. TRPC6 was phosphorylated at Thr(69) in nonstimulated podocytes, but this declined upon ANG II stimulation or overexpression of constitutively active calcineurin phosphatase. ANG II induced podocyte motility in an in vitro wound assay, and this was reduced 30-60% in cells overexpressing a phosphomimetic mutant TRPC6 (TRPC6T70E/S322E) or activated PKG (P < 0.05). Pretreatment of podocytes with the PKG agonists S-nitroso-N-acetyl-dl-penicillamine (nitric oxide donor), 8-bromo-cGMP, Bay 41-2772 (soluble guanylate cyclase activator), or phosphodiesterase 5 (PDE5) inhibitor 4-{[3',4'-(methylenedioxy)benzyl]amino}[7]-6-methoxyquinazoline attenuated ANG II-induced Thr(69) dephosphorylation and also inhibited TRPC6-dependent podocyte motility by 30-60%. These data reveal that PKG activation strategies, including PDE5 inhibition, ameliorate ANG II-induced podocyte dysmotility by targeting TRPC6 in podocytes, highlighting the potential therapeutic utility of these approaches to treat hyperactive TRPC6-dependent glomerular disease.


Asunto(s)
Angiotensina II/farmacología , Movimiento Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Podocitos/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Células Cultivadas , Regulación hacia Abajo/fisiología , Células HEK293 , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos , Modelos Animales , Factores de Transcripción NFATC/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Podocitos/citología , Podocitos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Canal Catiónico TRPC6
5.
Kidney Int ; 86(6): 1253-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25229338

RESUMEN

Focal segmental glomerulosclerosis (FSGS) is a histological lesion with many causes, including inherited genetic defects, with significant proteinuria being the predominant clinical finding at presentation. Mutations in COL4A3 and COL4A4 are known to cause Alport syndrome (AS), thin basement membrane nephropathy, and to result in pathognomonic glomerular basement membrane (GBM) findings. Secondary FSGS is known to develop in classic AS at later stages of the disease. Here, we present seven families with rare or novel variants in COL4A3 or COL4A4 (six with single and one with two heterozygous variants) from a cohort of 70 families with a diagnosis of hereditary FSGS. The predominant clinical finding at diagnosis was proteinuria associated with hematuria. In all seven families, there were individuals with nephrotic-range proteinuria with histologic features of FSGS by light microscopy. In one family, electron microscopy showed thin GBM, but four other families had variable findings inconsistent with classical Alport nephritis. There was no recurrence of disease after kidney transplantation. Families with COL4A3 and COL4A4 variants that segregated with disease represent 10% of our cohort. Thus, COL4A3 and COL4A4 variants should be considered in the interpretation of next-generation sequencing data from such patients. Furthermore, this study illustrates the power of molecular genetic diagnostics in the clarification of renal phenotypes.


Asunto(s)
Autoantígenos/genética , Colágeno Tipo IV/genética , Glomeruloesclerosis Focal y Segmentaria/genética , Adolescente , Adulto , Niño , Análisis Mutacional de ADN , Exoma , Femenino , Pruebas Genéticas , Genotipo , Membrana Basal Glomerular/ultraestructura , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Glomeruloesclerosis Focal y Segmentaria/patología , Pérdida Auditiva/genética , Hematuria/etiología , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Fenotipo , Podocitos/ultraestructura , Proteinuria/etiología , Adulto Joven
6.
J Am Soc Nephrol ; 24(8): 1313-22, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23620400

RESUMEN

Primary vesicoureteral reflux (VUR) is the most common congenital anomaly of the kidney and the urinary tract, and it is a major risk factor for pyelonephritic scarring and CKD in children. Although twin studies support the heritability of VUR, specific genetic causes remain elusive. We performed a sequential genome-wide linkage study and whole-exome sequencing in a family with hereditary VUR. We obtained a significant multipoint parametric logarithm of odds score of 3.3 on chromosome 6p, and whole-exome sequencing identified a deleterious heterozygous mutation (T3257I) in the gene encoding tenascin XB (TNXB in 6p21.3). This mutation segregated with disease in the affected family as well as with a pathogenic G1331R change in another family. Fibroblast cell lines carrying the T3257I mutation exhibited a reduction in both cell motility and phosphorylated focal adhesion kinase expression, suggesting a defect in the focal adhesions that link the cell cytoplasm to the extracellular matrix. Immunohistochemical studies revealed that the human uroepithelial lining of the ureterovesical junction expresses TNXB, suggesting that TNXB may be important for generating tensile forces that close the ureterovesical junction during voiding. Taken together, these results suggest that mutations in TNXB can cause hereditary VUR.


Asunto(s)
Adhesión Celular/genética , Movimiento Celular/genética , Riñón/patología , Tenascina/genética , Sistema Urinario/anomalías , Reflujo Vesicoureteral/genética , Femenino , Estudio de Asociación del Genoma Completo , Heterocigoto , Humanos , Riñón/metabolismo , Masculino , Mutación , Linaje , Análisis de Secuencia de ADN , Tenascina/metabolismo , Sistema Urinario/metabolismo , Sistema Urinario/patología , Reflujo Vesicoureteral/metabolismo , Reflujo Vesicoureteral/patología
7.
J Am Soc Nephrol ; 23(7): 1155-60, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22626820

RESUMEN

Controlled activation of the complement system, a key component of innate immunity, enables destruction of pathogens with minimal damage to host tissue. Complement factor H (CFH), which inhibits complement activation, and five CFH-related proteins (CFHR1-5) compose a family of structurally related molecules. Combined deletion of CFHR3 and CFHR1 is common and confers a protective effect in IgA nephropathy. Here, we report an autosomal dominant complement-mediated GN associated with abnormal increases in copy number across the CFHR3 and CFHR1 loci. In addition to normal copies of these genes, affected individuals carry a unique hybrid CFHR3-1 gene. In addition to identifying an association between these genetic observations and complement-mediated kidney disease, these results provide insight into the protective role of the combined deletion of CFHR3 and CFHR1 in IgA nephropathy.


Asunto(s)
Proteínas Sanguíneas/genética , Quimera/genética , Complemento C3/metabolismo , Proteínas Inactivadoras del Complemento C3b/genética , Glomerulonefritis por IGA/genética , Glomerulonefritis por IGA/metabolismo , Adolescente , Adulto , Biopsia , Preescolar , Femenino , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Riñón/metabolismo , Riñón/patología , Masculino , Persona de Mediana Edad , Linaje
8.
Kidney Int ; 81(1): 94-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21866090

RESUMEN

Focal and segmental glomerulosclerosis (FSGS) is a major cause of end-stage kidney disease. Recent advances in molecular genetics show that defects in the podocyte play a major role in its pathogenesis and mutations in inverted formin 2 (INF2) cause autosomal dominant FSGS. In order to delineate the role of INF2 mutations in familial and sporadic FSGS, we sought to identify variants in a large cohort of patients with FSGS. A secondary objective was to define an approach for genetic screening in families with autosomal dominant disease. A total of 248 individuals were identified with FSGS, of whom 31 had idiopathic disease. The remaining patients clustered into 64 families encompassing 15 from autosomal recessive and 49 from autosomal dominant kindreds. There were missense mutations in 8 of the 49 families with autosomal dominant disease. Three of the detected variants were novel and all mutations were confined to exon 4 of INF2, a regulatory region responsible for 90% of all changes reported in FSGS due to INF2 mutations. Thus, in our series, INF2 mutations were responsible for 16% of all cases of autosomal dominant FSGS, with these mutations clustered in exon 4. Hence, screening for these mutations may represent a rapid, non-invasive and cost-effective method for the diagnosis of autosomal dominant FSGS.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas de Microfilamentos/genética , Mutación , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Niño , Preescolar , Exones , Femenino , Forminas , Genes Dominantes , Genes Recesivos , Pruebas Genéticas , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Lactante , Masculino , Proteínas de Microfilamentos/química , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación Missense , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Adulto Joven
9.
Mol Ther ; 19(11): 1961-70, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21730973

RESUMEN

Glycogen storage disease type Ia (GSD-Ia) is caused by the deficiency of glucose-6-phosphatase (G6Pase). Long-term complications of GSD-Ia include life-threatening hypoglycemia and proteinuria progressing to renal failure. A double-stranded (ds) adeno-associated virus serotype 2 (AAV2) vector encoding human G6Pase was pseudotyped with four serotypes, AAV2, AAV7, AAV8, and AAV9, and we evaluated efficacy in 12-day-old G6pase (-/-) mice. Hypoglycemia during fasting (plasma glucose <100 mg/dl) was prevented for >6 months by the dsAAV2/7, dsAAV2/8, and dsAAV2/9 vectors. Prolonged fasting for 8 hours revealed normalization of blood glucose following dsAAV2/9 vector administration at the higher dose. The glycogen content of kidney was reduced by >65% with both the dsAAV2/7 and dsAAV2/9 vectors, and renal glycogen content was stably reduced between 7 and 12 months of age for the dsAAV2/9 vector-treated mice. Every vector-treated group had significantly reduced glycogen content in the liver, in comparison with untreated G6pase (-/-) mice. G6Pase was expressed in many renal epithelial cells of with the dsAAV2/9 vector for up to 12 months. Albuminuria and renal fibrosis were reduced by the dsAAV2/9 vector. Hepatorenal correction in G6pase (-/-) mice demonstrates the potential of AAV vectors for the correction of inherited diseases of metabolism.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Animales , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Vectores Genéticos/administración & dosificación , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/mortalidad , Humanos , Hipoglucemia/genética , Hipoglucemia/terapia , Estimación de Kaplan-Meier , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados
10.
J Am Soc Nephrol ; 22(3): 526-35, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21258036

RESUMEN

Mutations in the canonical transient receptor potential cation channel 6 (TRPC6) are responsible for familial forms of adult onset focal segmental glomerulosclerosis (FSGS). The mechanisms by which TRPC6 mutations cause kidney disease are not well understood. We used TRPC6-deficient mice to examine the function of TRPC6 in the kidney. We found that adult TRPC6-deficient mice had BP and albumin excretion rates similar to wild-type animals. Glomerular histomorphology revealed no abnormalities on both light and electron microscopy. To determine whether the absence of TRPC6 would alter susceptibility to hypertension and renal injury, we infused mice with angiotensin II continuously for 28 days. Although both groups developed similar levels of hypertension, TRPC6-deficient mice had significantly less albuminuria, especially during the early phase of the infusion; this suggested that TRPC6 adversely influences the glomerular filter. We used whole-cell patch-clamp recording to measure cell-membrane currents in primary cultures of podocytes from both wild-type and TRPC6-deficient mice. In podocytes from wild-type mice, angiotensin II and a direct activator of TRPC6 both augmented cell-membrane currents; TRPC6 deficiency abrogated these increases in current magnitude. Our findings suggest that TRPC6 promotes albuminuria, perhaps by promoting angiotensin II-dependent increases in Ca(2+), suggesting that TRPC6 blockade may be therapeutically beneficial in proteinuric kidney disease.


Asunto(s)
Albuminuria/metabolismo , Angiotensina II/efectos adversos , Riñón/metabolismo , Canales Catiónicos TRPC/metabolismo , Albuminuria/etiología , Albuminuria/fisiopatología , Angiotensina II/administración & dosificación , Angiotensina II/farmacología , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Hipertensión/fisiopatología , Inyecciones Subcutáneas , Riñón/efectos de los fármacos , Riñón/fisiopatología , Masculino , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Podocitos/patología , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6
11.
J Am Soc Nephrol ; 22(3): 579-85, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21355056

RESUMEN

Steroid-resistant focal segmental glomerulosclerosis (FSGS) often recurs after renal transplantation. In this international survey, we sought to identify genotype-phenotype correlations of recurrent FSGS. We surveyed 83 patients with childhood-onset primary FSGS who received at least one renal allograft and analyzed 53 of these patients for NPHS2 mutations. The mean age at diagnosis was 6.7 years, and the mean age at first renal transplantation was 13 years. FSGS recurred in 30 patients (36%) after a median of 13 days (range, 1.5 to 152 days). Twenty-three patients received a second kidney transplant, and FSGS recurred in 11 (48%) after a median of 16 days (range, 2.7 to 66 days). None of the 11 patients with homozygous or compound heterozygous NPHS2 mutations developed recurrent FSGS compared with 45% of patients without mutations. These data suggest that genetic testing for pathogenic mutations may be important for prognosis and treatment of FSGS both before and after transplantation.


Asunto(s)
Pruebas Genéticas , Glomeruloesclerosis Focal y Segmentaria/epidemiología , Glomeruloesclerosis Focal y Segmentaria/cirugía , Péptidos y Proteínas de Señalización Intracelular/genética , Trasplante de Riñón , Proteínas de la Membrana/genética , Mutación/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Glomeruloesclerosis Focal y Segmentaria/genética , Supervivencia de Injerto , Heterocigoto , Homocigoto , Humanos , Lactante , Masculino , Recurrencia , Estudios Retrospectivos , Adulto Joven
12.
J Am Soc Nephrol ; 21(8): 1390-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20616172

RESUMEN

FSGS is a clinicopathologic entity characterized by nephrotic syndrome and progression to ESRD. Although the pathogenesis is unknown, the podocyte seems to play a central role in this disorder. Here, we present six kindreds with hereditary FSGS that did not associate with mutations in known causal genes, and we report a new locus for the disease on chromosome 2p15 in one kindred. We performed genome-wide linkage analysis and refined the linkage area with microsatellite markers and haplotype analysis to define the minimal candidate region. Genome-wide linkage analysis yielded a maximum two-point logarithm of odds (LOD) score of 3.6 for the six families on chromosome 2p. One family contributed the largest proportion of the additive score (LOD 2.02) at this locus. Multipoint parametric LOD score calculation in this family yielded a significant LOD score of 3.1 at markers D2S393 and D2S337, and fine mapping of this region with microsatellite markers defined a minimal candidate region of 0.9 Mb with observed recombinations at markers D2S2332 and RS1919481. We excluded the remaining five families from linkage to this region by haplotype analysis. These data support a new gene locus for familial FSGS on chromosome 2p15. Identification of the mutated gene at this locus may provide further insight into the disease mechanisms of FSGS.


Asunto(s)
Fallo Renal Crónico/etiología , Fallo Renal Crónico/genética , Síndrome Nefrótico/complicaciones , Síndrome Nefrótico/genética , Adolescente , Adulto , Niño , Preescolar , Cromosomas Humanos/genética , Progresión de la Enfermedad , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Escala de Lod , Masculino , Linaje , Adulto Joven
13.
J Am Soc Nephrol ; 19(6): 1071-5, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18434567

RESUMEN

FSGS is a pathologic lesion that frequently causes the nephrotic syndrome and ensuing renal failure. The cause remains unknown in the majority of individuals; however, in the past two decades, rare familial forms have been identified. It has been suggested that known genetic causes of the hereditary form of this disease account for upwards of 18% of cases. Mutations in five genes have been found to cause inherited nephrotic syndromes and FSGS. In this article, I discuss the phenotypic characteristics of hereditary FSGS and the transient receptor potential cation channel 6 (TRPC6) protein, which is the genetic impetus for an autosomal dominant form of FSGS. The TRP channels have been implicated in varied biologic functions such as mechanosensation, ion homeostasis, cell growth, and phospholipase C-dependent calcium entry into cells. The mutated ion channel causes an increase in calcium transients. Current evidence also suggests that blocking TRPC6 channels may be of therapeutic benefit in idiopathic FSGS, a disease with a generally poor prognosis. Preliminary experiments reveal that the commonly used immunosuppressive agent FK-506 can inhibit TRPC6 activity in vivo. This creates the intriguing possibility that blocking TRPC6 channels within the podocyte may translate into long-lasting clinical benefits in patients with FSGS.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Mutación , Canales Catiónicos TRPC/genética , Distinciones y Premios , Humanos , Linaje , Canal Catiónico TRPC6
14.
Biochim Biophys Acta ; 1772(8): 859-68, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17459670

RESUMEN

Focal and segmental glomerulosclerosis (FSGS) is a common cause of nephrotic syndrome in children and adults throughout the world. In the past 50 years, significant advances have been made in the identification and characterization of familial forms of nephrotic syndrome and FSGS. Resultant to these pursuits, several podocyte structural proteins such as nephrin, podocin, alpha-actinin 4 (ACTN4), and CD2-associated protein (CD2AP) have emerged to provide critical insight into the pathogenesis of hereditary nephrotic syndromes. The latest advance in familial FSGS has been the discovery of a mutant form of canonical transient receptor potential cation channel 6 (TRPC6), which causes an increase in calcium transients and essentially a gain of function in this cation channel located on the podocyte cell membrane. The TRP ion channel family is a diverse group of cation channels united by a common primary structure which contains six membrane-spanning domains, with both carboxy and amino termini located intracellularly. TRP channels are unique in their ability to activate independently of membrane depolarization. TRPC6 channels have been shown to be activated via phospholipase C stimulation. The mechanisms by which mutant TRPC6 causes an increase in intracellular calcium and leads to glomerulosclerosis are unknown. Mutant TRPC6 may affect critical interactions with the aforementioned podocyte structural proteins, leading to abnormalities in the slit diaphragm or podocyte foot processes. Mutant TRPC6 may also amplify injurious signals mediated by Ang II, a common final pathway of podocyte apoptosis in various mammalian species. Current evidence also suggests that blocking TRPC6 channels may be of therapeutic benefit in idiopathic FSGS, a disease with a generally poor prognosis. Preliminary experiments reveal the commonly used immunosuppressive agent FK-506 can inhibit TRPC6 activity in vivo. This creates the exciting possibility that blocking TRPC6 channels within the podocyte may translate into long-lasting clinical benefits in patients with FSGS.


Asunto(s)
Canalopatías/genética , Glomeruloesclerosis Focal y Segmentaria/genética , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/fisiología , Actinina/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas del Citoesqueleto/genética , Enfermedades Genéticas Congénitas/genética , Glomeruloesclerosis Focal y Segmentaria/patología , Glomeruloesclerosis Focal y Segmentaria/terapia , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Modelos Biológicos , Mutación , Síndrome Nefrótico/genética , Podocitos/patología , Canales Catiónicos TRPC/antagonistas & inhibidores , Canal Catiónico TRPC6
16.
Semin Nephrol ; 26(2): 89-94, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16530601

RESUMEN

Focal and segmental glomerulosclerosis (FSGS) is a pathologic entity that is a common and increasing cause of end-stage renal disease. Typical manifestations include proteinuria, hypertension, worsening renal insufficiency, and, frequently, renal failure. The etiology, however, remains unknown in a majority of patients. There is an estimated recurrence rate of 30% to 40% in renal transplant patients, suggesting that the pathogenesis is not solely a result of intrinsic kidney disease. Although some of its characteristics have been reported, the precise identification of a circulating factor associated with FSGS has not been made. Remarkable progress has been made in recent years regarding biologic mechanisms surrounding FSGS and proteinuria. Insight into the pathogenesis of FSGS has been gained through the study of hereditary forms of FSGS and nephrotic syndromes. Mutations in cytoskeletal proteins that affect podocyte structure have been the target until recently. Here we review the current understanding of this glomerular disease and areas for future concentration.


Asunto(s)
Biomarcadores/sangre , Glomeruloesclerosis Focal y Segmentaria , Proteínas Adaptadoras Transductoras de Señales , Proteínas del Citoesqueleto/sangre , Progresión de la Enfermedad , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Fallo Renal Crónico/etiología , Fallo Renal Crónico/patología , Proteínas de la Membrana/sangre , Factores de Riesgo , Canales Catiónicos TRPC/sangre , Canal Catiónico TRPC6 , Dominios Homologos src
17.
Am J Kidney Dis ; 45(3): 519-30, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15754274

RESUMEN

BACKGROUND: Post-cardiac surgery renal dysfunction is a common, serious, multifactorial disorder, with interpatient variability predicted poorly by preoperative clinical, procedural, and biological markers. Therefore, we tested the hypothesis that selected gene variants are associated with acute renal injury, reflected by a serum creatinine level increase after cardiac surgery. METHODS: One thousand six hundred seventy-one patients undergoing aortocoronary surgery were studied. Clinical covariates were recorded. DNA was isolated from preoperative blood; mass spectrometry was used for genotype analysis. A model was developed relating clinical and genetic factors to postoperative acute renal injury. RESULTS: A race effect was found; therefore, Caucasians and African Americans were analyzed separately. Overall, clinical factors alone account poorly for postoperative renal injury, although more so in African Americans than Caucasians. When 12 candidate polymorphisms were assessed, 2 alleles (interleukin 6 -572C and angiotensinogen 842C) showed a strong association with renal injury in Caucasians (P < 0.0001; >50% decrease in renal filtration when they present together). Using less stringent criteria for significance (0.01 > P > 0.001), 4 additional polymorphisms are identified (apolipoproteinE 448C [4], angiotensin receptor1 1166C, and endothelial nitric oxide synthase [eNOS] 894T in Caucasians; eNOS 894T and angiotensin-converting enzyme deletion and insertion in African Americans). Adding genetic to clinical factors resulted in the best model, with overall ability to explain renal injury increasing approximately 4-fold in Caucasians and doubling in African Americans (P < 0.0005). CONCLUSION: In this study, we identify genetic polymorphisms that collectively provide 2- to 4-fold improvement over preoperative clinical factors alone in explaining post-cardiac surgery renal dysfunction. From a mechanistic perspective, most identified genetic variants are associated with increased renal inflammatory and/or vasoconstrictor responses.


Asunto(s)
Lesión Renal Aguda/epidemiología , Puente de Arteria Coronaria , Polimorfismo Genético , Complicaciones Posoperatorias/epidemiología , Lesión Renal Aguda/sangre , Lesión Renal Aguda/etnología , Lesión Renal Aguda/genética , Negro o Afroamericano/genética , Anciano , Alelos , Angiotensinas/genética , Apolipoproteínas E/genética , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/cirugía , Creatinina/sangre , Citocinas/genética , Análisis Mutacional de ADN , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo III , North Carolina/epidemiología , Estudios Prospectivos , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Receptor de Angiotensina Tipo 1/genética , Riesgo , Población Blanca/genética
18.
J Clin Invest ; 125(5): 1913-26, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25844902

RESUMEN

Familial forms of focal segmental glomerulosclerosis (FSGS) have been linked to gain-of-function mutations in the gene encoding the transient receptor potential channel C6 (TRPC6). GPCRs coupled to Gq signaling activate TRPC6, suggesting that Gq-dependent TRPC6 activation underlies glomerular diseases. Here, we developed a murine model in which a constitutively active Gq α subunit (Gq(Q209L), referred to herein as GqQ>L) is specifically expressed in podocytes and examined the effects of this mutation in response to puromycin aminonucleoside (PAN) nephrosis. We found that compared with control animals, animals expressing GqQ>L exhibited robust albuminuria, structural features of FSGS, and reduced numbers of glomerular podocytes. Gq activation stimulated calcineurin (CN) activity, resulting in CN-dependent upregulation of TRPC6 in murine kidneys. Deletion of TRPC6 in GqQ>L-expressing mice prevented FSGS development and inhibited both tubular damage and podocyte loss induced by PAN nephrosis. Similarly, administration of the CN inhibitor FK506 reduced proteinuria and tubular injury but had more modest effects on glomerular pathology and podocyte numbers in animals with constitutive Gq activation. Moreover, these Gq-dependent effects on podocyte injury were generalizable to diabetic kidney disease, as expression of GqQ>L promoted albuminuria, mesangial expansion, and increased glomerular basement membrane width in diabetic mice. Together, these results suggest that targeting Gq/TRPC6 signaling may have therapeutic benefits for the treatment of glomerular diseases.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Glomeruloesclerosis Focal y Segmentaria/genética , Canales Catiónicos TRPC/fisiología , Albuminuria/inducido químicamente , Animales , Calcineurina/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Eliminación de Gen , Genes Reporteros , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Glomeruloesclerosis Focal y Segmentaria/patología , Células HEK293 , Humanos , Glomérulos Renales/patología , Túbulos Renales/patología , Ratones , Ratones Mutantes , Ratones Transgénicos , Factores de Transcripción NFATC/metabolismo , Podocitos/metabolismo , Mutación Puntual , Puromicina Aminonucleósido/toxicidad , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Canales Catiónicos TRPC/biosíntesis , Canales Catiónicos TRPC/deficiencia , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6 , Tacrolimus/farmacología
19.
Clin Kidney J ; 8(5): 538-42, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26413278

RESUMEN

BACKGROUND: Mutations in podocin (NPHS2) are the most common cause of childhood onset autosomal recessive steroid-resistant nephrotic syndrome (SRNS). The disease is characterized by early-onset proteinuria, resistance to immunosuppressive therapy and rapid progression to end-stage renal disease. Compound heterozygous changes involving the podocin variant R229Q combined with another pathogenic mutation have been associated with a mild phenotype with disease onset often in adulthood. METHODS: We screened 19 families with early-onset SRNS for mutations in NPHS2 and WT1 and identified four disease-causing mutations (three in NPHS2 and one in WT1) prior to planned whole-exome sequencing. RESULTS: We describe two families with three individuals presenting in childhood who are compound heterozygous for R229Q and one other pathogenic NPHS2 mutation, either L327F or A297V. One child presented at age 4 years (A297V plus R229Q) and the other two at age 13 (L327F plus R229Q), one with steadily deteriorating renal function. CONCLUSIONS: These cases highlight the phenotypic variability associated with the NPHS2 R229Q variant plus pathogenic mutation. Individuals may present with early aggressive disease.

20.
Nat Rev Nephrol ; 9(3): 179-84, 2013 03.
Artículo en Inglés | MEDLINE | ID: mdl-23321566

RESUMEN

Monogenic nephrotic syndrome (nephrotic syndrome caused by a single gene defect) is responsible for only a small percentage of cases of nephrotic syndrome, but information from studies of the unique cohort of patients with this form of the disease has dramatically improved our understanding of the disease pathogenesis. The use of genetic testing in the management of children and adults with nephrotic syndrome poses unique challenges for clinicians in terms of who to test and how to use the information obtained from testing in the clinical setting. In our view, not enough data exist at present to justify the routine genetic testing of all patients with nephrotic syndrome. Testing is warranted, however, in patients with congenital nephrotic syndrome (onset at 0-3 months), infantile nephrotic syndrome (onset at 3-12 months), a family history of nephrotic syndrome, and those in whom nephrotic syndrome is associated with other congenital malformations. The family and/or the patient should be given complete and unbiased information on the potential benefits and risks associated with therapy, including the reported outcomes of treatment in patients with similar mutations. Based on the data available in the literature so far, intensive immunosuppressive treatment is probably not indicated in monogenic nephrotic syndrome if complete or partial remission has not been achieved within 6 weeks of starting treatment. We advocate that family members of individuals with genetic forms of nephrotic syndrome undergo routine genetic testing prior to living-related kidney transplantation. Prospective, multicentre studies are needed to more completely determine the burden of disease caused by monogenic nephrotic syndrome, and randomized controlled trials are needed to clarify the presence or absence of clinical responses of monogenic nephrotic syndrome to available therapies.


Asunto(s)
Pruebas Genéticas , Síndrome Nefrótico/genética , Adulto , Niño , Pruebas Genéticas/economía , Pruebas Genéticas/métodos , Humanos , Cobertura del Seguro , Trasplante de Riñón , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA