Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genet Med ; 26(2): 101013, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37924258

RESUMEN

PURPOSE: RNF213, encoding a giant E3 ubiquitin ligase, has been recognized for its role as a key susceptibility gene for moyamoya disease. Case reports have also implicated specific variants in RNF213 with an early-onset form of moyamoya disease with full penetrance. We aimed to expand the phenotypic spectrum of monogenic RNF213-related disease and to evaluate genotype-phenotype correlations. METHODS: Patients were identified through reanalysis of exome sequencing data of an unselected cohort of unsolved pediatric cases and through GeneMatcher or ClinVar. Functional characterization was done by proteomics analysis and oxidative phosphorylation enzyme activities using patient-derived fibroblasts. RESULTS: We identified 14 individuals from 13 unrelated families with (de novo) missense variants in RNF213 clustering within or around the Really Interesting New Gene (RING) domain. Individuals presented either with early-onset stroke (n = 11) or with Leigh syndrome (n = 3). No genotype-phenotype correlation could be established. Proteomics using patient-derived fibroblasts revealed no significant differences between clinical subgroups. 3D modeling revealed a clustering of missense variants in the tertiary structure of RNF213 potentially affecting zinc-binding suggesting a gain-of-function or dominant negative effect. CONCLUSION: De novo missense variants in RNF213 clustering in the E3 RING or other regions affecting zinc-binding lead to an early-onset syndrome characterized by stroke or Leigh syndrome.


Asunto(s)
Enfermedad de Leigh , Enfermedad de Moyamoya , Accidente Cerebrovascular , Humanos , Niño , Enfermedad de Moyamoya/genética , Enfermedad de Leigh/complicaciones , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Zinc , Predisposición Genética a la Enfermedad , Adenosina Trifosfatasas/genética
2.
Am J Hum Genet ; 107(4): 727-742, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32891193

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) constitute one of the most frequent birth defects and represent the most common cause of chronic kidney disease in the first three decades of life. Despite the discovery of dozens of monogenic causes of CAKUT, most pathogenic pathways remain elusive. We performed whole-exome sequencing (WES) in 551 individuals with CAKUT and identified a heterozygous de novo stop-gain variant in ZMYM2 in two different families with CAKUT. Through collaboration, we identified in total 14 different heterozygous loss-of-function mutations in ZMYM2 in 15 unrelated families. Most mutations occurred de novo, indicating possible interference with reproductive function. Human disease features are replicated in X. tropicalis larvae with morpholino knockdowns, in which expression of truncated ZMYM2 proteins, based on individual mutations, failed to rescue renal and craniofacial defects. Moreover, heterozygous Zmym2-deficient mice recapitulated features of CAKUT with high penetrance. The ZMYM2 protein is a component of a transcriptional corepressor complex recently linked to the silencing of developmentally regulated endogenous retrovirus elements. Using protein-protein interaction assays, we show that ZMYM2 interacts with additional epigenetic silencing complexes, as well as confirming that it binds to FOXP1, a transcription factor that has also been linked to CAKUT. In summary, our findings establish that loss-of-function mutations of ZMYM2, and potentially that of other proteins in its interactome, as causes of human CAKUT, offering new routes for studying the pathogenesis of the disorder.


Asunto(s)
Proteínas de Unión al ADN/genética , Epigénesis Genética , Factores de Transcripción Forkhead/genética , Mutación , Proteínas Represoras/genética , Factores de Transcripción/genética , Sistema Urinario/metabolismo , Anomalías Urogenitales/genética , Proteínas Anfibias/antagonistas & inhibidores , Proteínas Anfibias/genética , Proteínas Anfibias/metabolismo , Animales , Estudios de Casos y Controles , Niño , Preescolar , Proteínas de Unión al ADN/metabolismo , Familia , Femenino , Factores de Transcripción Forkhead/metabolismo , Heterocigoto , Humanos , Lactante , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Masculino , Ratones , Ratones Noqueados , Morfolinos/genética , Morfolinos/metabolismo , Linaje , Unión Proteica , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Sistema Urinario/anomalías , Anomalías Urogenitales/metabolismo , Anomalías Urogenitales/patología , Secuenciación del Exoma , Xenopus
3.
Mol Genet Metab ; 140(3): 107675, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37572574

RESUMEN

Recessive variants in NDUFAF3 are a known cause of complex I (CI)-related mitochondrial disorders (MDs). The seven patients reported to date exhibited severe neurologic symptoms and lactic acidosis, followed by a fatal course and death during infancy in most cases. We present a 10-year-old patient with a neurodevelopmental disorder, progressive exercise intolerance, dystonia, basal ganglia abnormalities, and elevated lactate concentration in blood. Trio-exome sequencing revealed compound-heterozygosity for a pathogenic splice-site and a likely pathogenic missense variant in NDUFAF3. Spectrophotometric analysis of fibroblast-derived mitochondria demonstrated a relatively mild reduction of CI activity. Complexome analyses revealed severely reduced NDUFAF3 as well as CI in patient fibroblasts. Accumulation of early sub-assemblies of the membrane arm of CI associated with mitochondrial complex I intermediate assembly (MCIA) complex was observed. The most striking additional findings were both the unusual occurrence of free monomeric CI holding MCIA and other assembly factors. Here we discuss our patient in context of genotype, phenotype and metabolite data from previously reported NDUFAF3 cases. With the atypical presentation of our patient, we provide further insight into the phenotypic spectrum of NDUFAF3-related MDs. Complexome analysis in our patient confirms the previously defined role of NDUFAF3 within CI biogenesis, yet adds new aspects regarding the correct timing of both the association of soluble and membrane arm modules and CI-maturation as well as respiratory supercomplex formation.


Asunto(s)
Acidosis Láctica , Enfermedades Mitocondriales , Humanos , Niño , Enfermedades Mitocondriales/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Secuenciación del Exoma , Acidosis Láctica/genética , Fenotipo , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo
4.
Am J Med Genet A ; 191(5): 1355-1359, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36694287

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are the most prevalent cause of chronic kidney disease that manifests in children. To date ~23 different monogenic causes have been implicated in isolated forms of human CAKUT, but the vast majority remains elusive. In a previous study, we identified a homozygous missense variant in E26 transformation-specific (ETS) Variant Transcription Factor 4 (ETV4) causing CAKUT via dysregulation of the transcriptional function of ETV4, and a resulting abrogation of GDNF/RET/ETV4 signaling pathway. This CAKUT family remains the only family with an ETV4 variant reported so far. Here, we describe one additional CAKUT family with a homozygous truncating variant in ETV4 (p.(Lys6*)) that was identified by exome sequencing. The variant was found in an individual with isolated CAKUT displaying posterior urethral valves and renal dysplasia. The newly identified stop variant conceptually truncates the ETS_PEA3_N and ETS domains that regulate DNA-binding transcription factor activity. The variant has never been reported homozygously in the gnomAD database. To our knowledge, we here report the first CAKUT family with a truncating variant in ETV4, potentially causing the isolated CAKUT phenotype observed in the affected individual.


Asunto(s)
Sistema Urinario , Anomalías Urogenitales , Reflujo Vesicoureteral , Niño , Humanos , Anomalías Urogenitales/genética , Riñón/anomalías , Sistema Urinario/metabolismo , Reflujo Vesicoureteral/genética , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo
5.
Am J Hum Genet ; 105(6): 1286-1293, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31708116

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of chronic kidney disease in the first three decades of life, and in utero obstruction to urine flow is a frequent cause of secondary upper urinary tract malformations. Here, using whole-exome sequencing, we identified three different biallelic mutations in CHRNA3, which encodes the α3 subunit of the nicotinic acetylcholine receptor, in five affected individuals from three unrelated families with functional lower urinary tract obstruction and secondary CAKUT. Four individuals from two families have additional dysautonomic features, including impaired pupillary light reflexes. Functional studies in vitro demonstrated that the mutant nicotinic acetylcholine receptors were unable to generate current following stimulation with acetylcholine. Moreover, the truncating mutations p.Thr337Asnfs∗81 and p.Ser340∗ led to impaired plasma membrane localization of CHRNA3. Although the importance of acetylcholine signaling in normal bladder function has been recognized, we demonstrate for the first time that mutations in CHRNA3 can cause bladder dysfunction, urinary tract malformations, and dysautonomia. These data point to a pathophysiologic sequence by which monogenic mutations in genes that regulate bladder innervation may secondarily cause CAKUT.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo/etiología , Riñón/anomalías , Mutación , Receptores Nicotínicos/genética , Sistema Urinario/anomalías , Anomalías Urogenitales/etiología , Adulto , Enfermedades del Sistema Nervioso Autónomo/genética , Enfermedades del Sistema Nervioso Autónomo/patología , Femenino , Estudios de Seguimiento , Humanos , Riñón/patología , Masculino , Linaje , Pronóstico , Sistema Urinario/patología , Anomalías Urogenitales/genética , Anomalías Urogenitales/patología , Adulto Joven
6.
J Am Soc Nephrol ; 32(3): 580-596, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33593823

RESUMEN

BACKGROUND: Galloway-Mowat syndrome (GAMOS) is characterized by neurodevelopmental defects and a progressive nephropathy, which typically manifests as steroid-resistant nephrotic syndrome. The prognosis of GAMOS is poor, and the majority of children progress to renal failure. The discovery of monogenic causes of GAMOS has uncovered molecular pathways involved in the pathogenesis of disease. METHODS: Homozygosity mapping, whole-exome sequencing, and linkage analysis were used to identify mutations in four families with a GAMOS-like phenotype, and high-throughput PCR technology was applied to 91 individuals with GAMOS and 816 individuals with isolated nephrotic syndrome. In vitro and in vivo studies determined the functional significance of the mutations identified. RESULTS: Three biallelic variants of the transcriptional regulator PRDM15 were detected in six families with proteinuric kidney disease. Four families with a variant in the protein's zinc-finger (ZNF) domain have additional GAMOS-like features, including brain anomalies, cardiac defects, and skeletal defects. All variants destabilize the PRDM15 protein, and the ZNF variant additionally interferes with transcriptional activation. Morpholino oligonucleotide-mediated knockdown of Prdm15 in Xenopus embryos disrupted pronephric development. Human wild-type PRDM15 RNA rescued the disruption, but the three PRDM15 variants did not. Finally, CRISPR-mediated knockout of PRDM15 in human podocytes led to dysregulation of several renal developmental genes. CONCLUSIONS: Variants in PRDM15 can cause either isolated nephrotic syndrome or a GAMOS-type syndrome on an allelic basis. PRDM15 regulates multiple developmental kidney genes, and is likely to play an essential role in renal development in humans.


Asunto(s)
Proteínas de Unión al ADN/genética , Hernia Hiatal/genética , Microcefalia/genética , Mutación Missense , Nefrosis/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Línea Celular , Preescolar , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/deficiencia , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Modelos Moleculares , Síndrome Nefrótico/genética , Podocitos/metabolismo , Polimorfismo de Nucleótido Simple , Pronefro/embriología , Pronefro/metabolismo , Estabilidad Proteica , Factores de Transcripción/química , Factores de Transcripción/deficiencia , Xenopus laevis/embriología , Xenopus laevis/genética , Dedos de Zinc/genética
7.
Neurogenetics ; 22(4): 263-269, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34218362

RESUMEN

ANK3 encodes multiple isoforms of ankyrin-G, resulting in variegated tissue expression and function, especially regarding its role in neuronal development. Based on the zygosity, location, and type, ANK3 variants result in different neurodevelopmental phenotypes. Autism spectrum disorder has been associated with heterozygous missense variants in ANK3, whereas a more severe neurodevelopmental phenotype is caused by isoform-dependent, autosomal-dominant, or autosomal-recessive loss-of-function variants. Here, we present four individuals affected by a variable neurodevelopmental phenotype harboring a heterozygous frameshift or nonsense variant affecting all ANK3 transcripts. Thus, we provide further evidence of an isoform-based phenotypic continuum underlying ANK3-associated pathologies and expand its phenotypic spectrum.


Asunto(s)
Ancirinas/genética , Trastorno del Espectro Autista/genética , Discapacidad Intelectual/genética , Trastornos del Neurodesarrollo/genética , Adolescente , Niño , Humanos , Pérdida de Heterocigocidad , Masculino , Mutación Missense/genética , Fenotipo , Isoformas de Proteínas/genética
8.
Genet Med ; 23(3): 543-554, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33149277

RESUMEN

PURPOSE: A few de novo missense variants in the cytoplasmic FMRP-interacting protein 2 (CYFIP2) gene have recently been described as a novel cause of severe intellectual disability, seizures, and hypotonia in 18 individuals, with p.Arg87 substitutions in the majority. METHODS: We assembled data from 19 newly identified and all 18 previously published individuals with CYFIP2 variants. By structural modeling and investigation of WAVE-regulatory complex (WRC)-mediated actin polymerization in six patient fibroblast lines we assessed the impact of CYFIP2 variants on the WRC. RESULTS: Sixteen of 19 individuals harbor two previously described and 11 novel (likely) disease-associated missense variants. We report p.Asp724 as second mutational hotspot (4/19 cases). Genotype-phenotype correlation confirms a consistently severe phenotype in p.Arg87 patients but a more variable phenotype in p.Asp724 and other substitutions. Three individuals with milder phenotypes carry putative loss-of-function variants, which remain of unclear pathogenicity. Structural modeling predicted missense variants to disturb interactions within the WRC or impair CYFIP2 stability. Consistent with its role in WRC-mediated actin polymerization we substantiate aberrant regulation of the actin cytoskeleton in patient fibroblasts. CONCLUSION: Our study expands the clinical and molecular spectrum of CYFIP2-related neurodevelopmental disorder and provides evidence for aberrant WRC-mediated actin dynamics as contributing cellular pathomechanism.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Actinas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Humanos , Discapacidad Intelectual/genética , Trastornos del Neurodesarrollo/genética , Convulsiones
9.
Clin Genet ; 100(6): 766-770, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34490615

RESUMEN

Neurological symptoms are frequent and often a leading feature of childhood-onset mitochondrial disorders (MD) but the exact incidence of MD in unselected neuropediatric patients is unknown. Their early detection is desirable due to a potentially rapid clinical decline and the availability of management options. In 491 children with neurological symptoms, a comprehensive diagnostic work-up including exome sequencing was performed. The success rate in terms of a molecular genetic diagnosis within our cohort was 51%. Disease-causing variants in a mitochondria-associated gene were detected in 12% of solved cases. In order to facilitate the clinical identification of MDs within neuropediatric cohorts, we have created an easy-to-use bedside-tool, the MDC-NP. In our cohort, the MDC-NP predicted disease conditions related to MDs with a sensitivity of 0.83, and a specificity of 0.96.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedades Mitocondriales/epidemiología , Enfermedades Mitocondriales/genética , Enfermedades del Sistema Nervioso/epidemiología , Enfermedades del Sistema Nervioso/genética , Factores de Edad , Alelos , Niño , Estudios de Cohortes , Genes Mitocondriales , Estudios de Asociación Genética , Genotipo , Humanos , Enfermedades Mitocondriales/diagnóstico , Mutación , Enfermedades del Sistema Nervioso/diagnóstico , Fenotipo , Prevalencia , Pronóstico
10.
Am J Med Genet A ; 185(12): 3784-3792, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34338422

RESUMEN

The acronym VATER/VACTERL refers to the rare nonrandom association of the following component features (CFs): vertebral defects (V), anorectal malformations (ARM) (A), cardiac anomalies (C), tracheoesophageal fistula with or without esophageal atresia (TE), renal malformations (R), and limb anomalies (L). For the clinical diagnosis, the presence of at least three CFs is required, individuals presenting with only two CFs have been categorized as VATER/VACTERL-like. The majority of VATER/VACTERL individuals displays a renal phenotype. Hitherto, variants in FGF8, FOXF1, HOXD13, LPP, TRAP1, PTEN, and ZIC3 have been associated with the VATER/VACTERL association; however, large-scale re-sequencing could only confirm TRAP1 and ZIC3 as VATER/VACTERL disease genes, both associated with a renal phenotype. In this study, we performed exome sequencing in 21 individuals and their families with a renal VATER/VACTERL or VATER/VACTERL-like phenotype to identify potentially novel genetic causes. Exome analysis identified biallelic and X-chromosomal hemizygous potentially pathogenic variants in six individuals (29%) in B9D1, FREM1, ZNF157, SP8, ACOT9, and TTLL11, respectively. The online tool GeneMatcher revealed another individual with a variant in ZNF157. Our study suggests six biallelic and X-chromosomal hemizygous VATER/VACTERL disease genes implicating all six genes in the expression of human renal malformations.


Asunto(s)
Malformaciones Anorrectales/genética , Atresia Esofágica/genética , Predisposición Genética a la Enfermedad , Cardiopatías/genética , Fístula Traqueoesofágica/genética , Malformaciones Anorrectales/complicaciones , Malformaciones Anorrectales/patología , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Atresia Esofágica/complicaciones , Atresia Esofágica/patología , Femenino , Genes Ligados a X/genética , Estudios de Asociación Genética , Proteínas HSP90 de Choque Térmico/genética , Cardiopatías/complicaciones , Cardiopatías/patología , Hemicigoto , Proteínas de Homeodominio/genética , Humanos , Riñón/anomalías , Masculino , Receptores de Interleucina/genética , Fístula Traqueoesofágica/complicaciones , Fístula Traqueoesofágica/patología , Factores de Transcripción/genética , Secuenciación del Exoma
11.
J Am Soc Nephrol ; 30(2): 201-215, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30655312

RESUMEN

BACKGROUND: Whole-exome sequencing (WES) finds a CKD-related mutation in approximately 20% of patients presenting with CKD before 25 years of age. Although provision of a molecular diagnosis could have important implications for clinical management, evidence is lacking on the diagnostic yield and clinical utility of WES for pediatric renal transplant recipients. METHODS: To determine the diagnostic yield of WES in pediatric kidney transplant recipients, we recruited 104 patients who had received a transplant at Boston Children's Hospital from 2007 through 2017, performed WES, and analyzed results for likely deleterious variants in approximately 400 genes known to cause CKD. RESULTS: By WES, we identified a genetic cause of CKD in 34 out of 104 (32.7%) transplant recipients. The likelihood of detecting a molecular genetic diagnosis was highest for patients with urinary stone disease (three out of three individuals), followed by renal cystic ciliopathies (seven out of nine individuals), steroid-resistant nephrotic syndrome (nine out of 21 individuals), congenital anomalies of the kidney and urinary tract (ten out of 55 individuals), and chronic glomerulonephritis (one out of seven individuals). WES also yielded a molecular diagnosis for four out of nine individuals with ESRD of unknown etiology. The WES-related molecular genetic diagnosis had implications for clinical care for five patients. CONCLUSIONS: Nearly one third of pediatric renal transplant recipients had a genetic cause of their kidney disease identified by WES. Knowledge of this genetic information can help guide management of both transplant patients and potential living related donors.


Asunto(s)
Secuenciación del Exoma/métodos , Trasplante de Riñón/métodos , Medicina de Precisión/métodos , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/cirugía , Adolescente , Boston , Niño , Preescolar , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Pruebas Genéticas/métodos , Rechazo de Injerto , Supervivencia de Injerto , Hospitales Pediátricos , Humanos , Trasplante de Riñón/efectos adversos , Masculino , Pronóstico , Insuficiencia Renal Crónica/fisiopatología , Estudios Retrospectivos , Medición de Riesgo , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Receptores de Trasplantes/estadística & datos numéricos , Resultado del Tratamiento
12.
Kidney Int ; 95(4): 914-928, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30773290

RESUMEN

Approximately 500 monogenic causes of chronic kidney disease (CKD) have been identified, mainly in pediatric populations. The frequency of monogenic causes among adults with CKD has been less extensively studied. To determine the likelihood of detecting monogenic causes of CKD in adults presenting to nephrology services in Ireland, we conducted whole exome sequencing (WES) in a multi-centre cohort of 114 families including 138 affected individuals with CKD. Affected adults were recruited from 78 families with a positive family history, 16 families with extra-renal features, and 20 families with neither a family history nor extra-renal features. We detected a pathogenic mutation in a known CKD gene in 42 of 114 families (37%). A monogenic cause was identified in 36% of affected families with a positive family history of CKD, 69% of those with extra-renal features, and only 15% of those without a family history or extra-renal features. There was no difference in the rate of genetic diagnosis in individuals with childhood versus adult onset CKD. Among the 42 families in whom a monogenic cause was identified, WES confirmed the clinical diagnosis in 17 (40%), corrected the clinical diagnosis in 9 (22%), and established a diagnosis for the first time in 16 families referred with CKD of unknown etiology (38%). In this multi-centre study of adults with CKD, a molecular genetic diagnosis was established in over one-third of families. In the evolving era of precision medicine, WES may be an important tool to identify the cause of CKD in adults.


Asunto(s)
Secuenciación del Exoma , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Insuficiencia Renal Crónica/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Exoma/genética , Femenino , Humanos , Irlanda , Riñón , Masculino , Anamnesis , Persona de Mediana Edad , Mutación , Linaje , Medicina de Precisión , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/terapia , Adulto Joven
13.
Nephrol Dial Transplant ; 34(3): 474-485, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30295827

RESUMEN

BACKGROUND: Alport syndrome (AS) and atypical hemolytic-uremic syndrome (aHUS) are rare forms of chronic kidney disease (CKD) that can lead to a severe decline of renal function. Steroid-resistant nephrotic syndrome (SRNS) is more common than AS and aHUS and causes 10% of childhood-onset CKD. In recent years, multiple monogenic causes of AS, aHUS and SRNS have been identified, but their relative prevalence has yet to be studied together in a typical pediatric cohort of children with proteinuria and hematuria. We hypothesized that identification of causative mutations by whole exome sequencing (WES) in known monogenic nephritis and nephrosis genes would allow distinguishing nephritis from nephrosis in a typical pediatric group of patients with both proteinuria and hematuria at any level. METHODS: We therefore conducted an exon sequencing (WES) analysis for 11 AS, aHUS and thrombotic thrombocytopenic purpura-causing genes in an international cohort of 371 patients from 362 families presenting with both proteinuria and hematuria before age 25 years. In parallel, we conducted either WES or high-throughput exon sequencing for 23 SRNS-causing genes in all patients. RESULTS: We detected pathogenic mutations in 18 of the 34 genes analyzed, leading to a molecular diagnosis in 14.1% of families (51 of 362). Disease-causing mutations were detected in 3 AS-causing genes (4.7%), 3 aHUS-causing genes (1.4%) and 12 NS-causing genes (8.0%). We observed a much higher mutation detection rate for monogenic forms of CKD in consanguineous families (35.7% versus 10.1%). CONCLUSIONS: We present the first estimate of relative frequency of inherited AS, aHUS and NS in a typical pediatric cohort with proteinuria and hematuria. Important therapeutic and preventative measures may result from mutational analysis in individuals with proteinuria and hematuria.


Asunto(s)
Secuenciación del Exoma/métodos , Marcadores Genéticos , Mutación , Nefritis/diagnóstico , Nefritis/genética , Nefrosis/diagnóstico , Nefrosis/genética , Adolescente , Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/genética , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Diagnóstico Diferencial , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Nefritis Hereditaria/diagnóstico , Nefritis Hereditaria/genética , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/genética , Pronóstico
14.
J Am Soc Nephrol ; 29(1): 36-50, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29079659

RESUMEN

Congenital anomalies of the kidneys and urinary tract (CAKUT) comprise a large spectrum of congenital malformations ranging from severe manifestations, such as renal agenesis, to potentially milder conditions, such as vesicoureteral reflux. CAKUT causes approximately 40% of ESRD that manifests within the first three decades of life. Several lines of evidence indicate that CAKUT is often caused by recessive or dominant mutations in single (monogenic) genes. To date, approximately 40 monogenic genes are known to cause CAKUT if mutated, explaining 5%-20% of patients. However, hundreds of different monogenic CAKUT genes probably exist. The discovery of novel CAKUT-causing genes remains challenging because of this pronounced heterogeneity, variable expressivity, and incomplete penetrance. We here give an overview of known genetic causes for human CAKUT and shed light on distinct renal morphogenetic pathways that were identified as relevant for CAKUT in mice and humans.


Asunto(s)
Anomalías Congénitas/genética , Matriz Extracelular/genética , Morfogénesis/genética , Transducción de Señal/genética , Sistema Urinario/anomalías , Sistema Urinario/embriología , Animales , Matriz Extracelular/metabolismo , Humanos
15.
J Am Soc Nephrol ; 29(8): 2123-2138, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29959197

RESUMEN

BACKGROUND: Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of CKD. The discovery of monogenic causes of SRNS has revealed specific pathogenetic pathways, but these monogenic causes do not explain all cases of SRNS. METHODS: To identify novel monogenic causes of SRNS, we screened 665 patients by whole-exome sequencing. We then evaluated the in vitro functional significance of two genes and the mutations therein that we discovered through this sequencing and conducted complementary studies in podocyte-like Drosophila nephrocytes. RESULTS: We identified conserved, homozygous missense mutations of GAPVD1 in two families with early-onset NS and a homozygous missense mutation of ANKFY1 in two siblings with SRNS. GAPVD1 and ANKFY1 interact with the endosomal regulator RAB5. Coimmunoprecipitation assays indicated interaction between GAPVD1 and ANKFY1 proteins, which also colocalized when expressed in HEK293T cells. Silencing either protein diminished the podocyte migration rate. Compared with wild-type GAPVD1 and ANKFY1, the mutated proteins produced upon ectopic expression of GAPVD1 or ANKFY1 bearing the patient-derived mutations exhibited altered binding affinity for active RAB5 and reduced ability to rescue the knockout-induced defect in podocyte migration. Coimmunoprecipitation assays further demonstrated a physical interaction between nephrin and GAPVD1, and immunofluorescence revealed partial colocalization of these proteins in rat glomeruli. The patient-derived GAPVD1 mutations reduced nephrin-GAPVD1 binding affinity. In Drosophila, silencing Gapvd1 impaired endocytosis and caused mistrafficking of the nephrin ortholog. CONCLUSIONS: Mutations in GAPVD1 and probably in ANKFY1 are novel monogenic causes of NS. The discovery of these genes implicates RAB5 regulation in the pathogenesis of human NS.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Síndrome Nefrótico/genética , Podocitos/metabolismo , Proteínas de Unión al GTP rab5/genética , Animales , Movimiento Celular/genética , Células Cultivadas , Estudios de Cohortes , Progresión de la Enfermedad , Drosophila melanogaster , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Tamizaje Masivo/métodos , Mutación Missense , Síndrome Nefrótico/patología , Linaje , Proteínas de Unión a Fosfato , Podocitos/patología , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Secuenciación del Exoma
16.
J Am Soc Nephrol ; 29(9): 2348-2361, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30143558

RESUMEN

BACKGROUND: Congenital anomalies of the kidney and urinary tract (CAKUT) are the most prevalent cause of kidney disease in the first three decades of life. Previous gene panel studies showed monogenic causation in up to 12% of patients with CAKUT. METHODS: We applied whole-exome sequencing to analyze the genotypes of individuals from 232 families with CAKUT, evaluating for mutations in single genes known to cause human CAKUT and genes known to cause CAKUT in mice. In consanguineous or multiplex families, we additionally performed a search for novel monogenic causes of CAKUT. RESULTS: In 29 families (13%), we detected a causative mutation in a known gene for isolated or syndromic CAKUT that sufficiently explained the patient's CAKUT phenotype. In three families (1%), we detected a mutation in a gene reported to cause a phenocopy of CAKUT. In 15 of 155 families with isolated CAKUT, we detected deleterious mutations in syndromic CAKUT genes. Our additional search for novel monogenic causes of CAKUT in consanguineous and multiplex families revealed a potential single, novel monogenic CAKUT gene in 19 of 232 families (8%). CONCLUSIONS: We identified monogenic mutations in a known human CAKUT gene or CAKUT phenocopy gene as the cause of disease in 14% of the CAKUT families in this study. Whole-exome sequencing provides an etiologic diagnosis in a high fraction of patients with CAKUT and will provide a new basis for the mechanistic understanding of CAKUT.


Asunto(s)
Secuenciación del Exoma/métodos , Predisposición Genética a la Enfermedad/epidemiología , Linaje , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/genética , Animales , Humanos , Incidencia , Riñón/anomalías , Ratones , Fenotipo , Pronóstico , Medición de Riesgo , Sensibilidad y Especificidad , Distribución por Sexo , Sistema Urinario/anomalías , Anomalías Urogenitales/epidemiología , Reflujo Vesicoureteral/epidemiología
17.
J Am Soc Nephrol ; 28(1): 69-75, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27151922

RESUMEN

Congenital anomalies of the kidneys and urinary tract (CAKUT) are the leading cause of CKD in children, featuring a broad variety of malformations. A monogenic cause can be detected in around 12% of patients. However, the morphologic clinical phenotype of CAKUT frequently does not indicate specific genes to be examined. To determine the likelihood of detecting causative recessive mutations by whole-exome sequencing (WES), we analyzed individuals with CAKUT from 33 different consanguineous families. Using homozygosity mapping and WES, we identified the causative mutations in nine of the 33 families studied (27%). We detected recessive mutations in nine known disease-causing genes: ZBTB24, WFS1, HPSE2, ATRX, ASPH, AGXT, AQP2, CTNS, and PKHD1 Notably, when mutated, these genes cause multiorgan syndromes that may include CAKUT as a feature (syndromic CAKUT) or cause renal diseases that may manifest as phenocopies of CAKUT. None of the above monogenic disease-causing genes were suspected on clinical grounds before this study. Follow-up clinical characterization of those patients allowed us to revise and detect relevant new clinical features in a more appropriate pathogenetic context. Thus, applying WES to the diagnostic approach in CAKUT provides opportunities for an accurate and early etiology-based diagnosis and improved clinical management.


Asunto(s)
Exoma/genética , Mutación , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/genética , Humanos , Fenotipo , Síndrome
18.
J Am Soc Nephrol ; 28(8): 2364-2376, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28381549

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of CKD in the first three decades of life. However, for most patients with CAKUT, the causative mutation remains unknown. We identified a kindred with an autosomal dominant form of CAKUT. By whole-exome sequencing, we identified a heterozygous truncating mutation (c.279delG, p.Trp93fs*) of the nuclear receptor interacting protein 1 gene (NRIP1) in all seven affected members. NRIP1 encodes a nuclear receptor transcriptional cofactor that directly interacts with the retinoic acid receptors (RARs) to modulate retinoic acid transcriptional activity. Unlike wild-type NRIP1, the altered NRIP1 protein did not translocate to the nucleus, did not interact with RARα, and failed to inhibit retinoic acid-dependent transcriptional activity upon expression in HEK293 cells. Notably, we also showed that treatment with retinoic acid enhanced NRIP1 binding to RARα RNA in situ hybridization confirmed Nrip1 expression in the developing urogenital system of the mouse. In explant cultures of embryonic kidney rudiments, retinoic acid stimulated Nrip1 expression, whereas a pan-RAR antagonist strongly reduced it. Furthermore, mice heterozygous for a null allele of Nrip1 showed a CAKUT-spectrum phenotype. Finally, expression and knockdown experiments in Xenopus laevis confirmed an evolutionarily conserved role for NRIP1 in renal development. These data indicate that dominant NRIP1 mutations can cause CAKUT by interference with retinoic acid transcriptional signaling, shedding light on the well documented association between abnormal vitamin A levels and renal malformations in humans, and suggest a possible gene-environment pathomechanism in this disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mutación , Proteínas Nucleares/genética , Transducción de Señal/genética , Tretinoina/fisiología , Sistema Urinario/anomalías , Animales , Ratones , Proteína de Interacción con Receptores Nucleares 1
19.
Pediatr Nephrol ; 32(12): 2273-2282, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28779239

RESUMEN

BACKGROUND: Rhabdomyolysis is a clinical emergency that may cause acute kidney injury (AKI). It can be acquired or due to monogenic mutations. Around 60 different rare monogenic forms of rhabdomyolysis have been reported to date. In the clinical setting, identifying the underlying molecular diagnosis is challenging due to nonspecific presentation, the high number of causative genes, and current lack of data on the prevalence of monogenic forms. METHODS: We employed whole exome sequencing (WES) to reveal the percentage of rhabdomyolysis cases explained by single-gene (monogenic) mutations in one of 58 candidate genes. We investigated a cohort of 21 unrelated families with rhabdomyolysis, in whom no underlying etiology had been previously established. RESULTS: Using WES, we identified causative mutations in candidate genes in nine of the 21 families (43%). We detected disease-causing mutations in eight of 58 candidate genes, grouped into the following categories: (1) disorders of fatty acid metabolism (CPT2), (2) disorders of glycogen metabolism (PFKM and PGAM2), (3) disorders of abnormal skeletal muscle relaxation and contraction (CACNA1S, MYH3, RYR1 and SCN4A), and (4) disorders of purine metabolism (AHCY). CONCLUSIONS: Our findings demonstrate a very high detection rate for monogenic etiologies using WES and reveal broad genetic heterogeneity for rhabdomyolysis. These results highlight the importance of molecular genetic diagnostics for establishing an etiologic diagnosis. Because these patients are at risk for recurrent episodes of rhabdomyolysis and subsequent risk for AKI, WES allows adequate prophylaxis and treatment for these patients and their family members and enables a personalized medicine approach.


Asunto(s)
Secuenciación del Exoma/métodos , Rabdomiólisis/genética , Adolescente , Adulto , Árabes/genética , Niño , Exoma , Predisposición Genética a la Enfermedad , Humanos , Judíos/genética , Mutación , Rabdomiólisis/etnología
20.
Nephrol Dial Transplant ; 31(8): 1280-3, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26908769

RESUMEN

BACKGROUND: Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of chronic kidney diseases in children and young adults, accounting for ∼50% of cases. These anomalies represent maldevelopment of the genitourinary system and can be genetically explained in only 10-16% of cases by mutations or by copy number variations in protein coding sequences. Knock-out mouse models, lacking components of the microRNA (miRNA) processing machinery (i.e. Dicer, Drosha, Dgcr8), exhibit kidney malformations resembling human CAKUT. METHODS: Given the Dicer-null mouse phenotype, which implicates a central role for miRNAs gene regulation during kidney development, we hypothesized that miRNAs expressed during kidney development may cause CAKUT in humans if mutated. To evaluate this possibility we carried out Next-Generation sequencing of 96 stem-loop regions of 73 renal developmental miRNA genes in 1248 individuals with non-syndromic CAKUT from 980 families. RESULTS: We sequenced 96 stem-loop regions encoded by 73 miRNA genes that are expressed during kidney development in humans, mice and rats. Overall, we identified in 31/1213 individuals from 26 families with 17 different single nucleotide variants. Two variants did not segregate with the disease and hence were not causative. Thirteen variants were likely benign variants because they occurred in control populations and/or they affected nucleotides of weak evolutionary conservation. Two out of 1213 unrelated individuals had potentially pathogenic variants with unknown biologic relevance affecting miRNAs MIR19B1 and MIR99A. CONCLUSIONS: Our results indicate that mutations affecting mature microRNAs in individuals with CAKUT are rare and thus most likely not a common cause of CAKUT in humans.


Asunto(s)
Riñón/anomalías , MicroARNs/genética , Mutación , Sistema Urinario/anomalías , Anomalías Urogenitales/genética , Adolescente , Animales , Niño , Variaciones en el Número de Copia de ADN , Humanos , Ratones , Ratones Noqueados , Fenotipo , Ratas , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA