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













Base de datos
Intervalo de año de publicación
1.
Kidney Int ; 105(4): 844-864, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38154558

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are the predominant cause for chronic kidney disease below age 30 years. Many monogenic forms have been discovered due to comprehensive genetic testing like exome sequencing. However, disease-causing variants in known disease-associated genes only explain a proportion of cases. Here, we aim to unravel underlying molecular mechanisms of syndromic CAKUT in three unrelated multiplex families with presumed autosomal recessive inheritance. Exome sequencing in the index individuals revealed three different rare homozygous variants in FOXD2, encoding a transcription factor not previously implicated in CAKUT in humans: a frameshift in the Arabic and a missense variant each in the Turkish and the Israeli family with segregation patterns consistent with autosomal recessive inheritance. CRISPR/Cas9-derived Foxd2 knockout mice presented with a bilateral dilated kidney pelvis accompanied by atrophy of the kidney papilla and mandibular, ophthalmologic, and behavioral anomalies, recapitulating the human phenotype. In a complementary approach to study pathomechanisms of FOXD2-dysfunction-mediated developmental kidney defects, we generated CRISPR/Cas9-mediated knockout of Foxd2 in ureteric bud-induced mouse metanephric mesenchyme cells. Transcriptomic analyses revealed enrichment of numerous differentially expressed genes important for kidney/urogenital development, including Pax2 and Wnt4 as well as gene expression changes indicating a shift toward a stromal cell identity. Histology of Foxd2 knockout mouse kidneys confirmed increased fibrosis. Further, genome-wide association studies suggest that FOXD2 could play a role for maintenance of podocyte integrity during adulthood. Thus, our studies help in genetic diagnostics of monogenic CAKUT and in understanding of monogenic and multifactorial kidney diseases.


Asunto(s)
Estructuras Embrionarias , Factores de Transcripción Forkhead , Enfermedades Renales , Riñón , Nefronas , Sistema Urinario , Anomalías Urogenitales , Reflujo Vesicoureteral , Adulto , Animales , Humanos , Ratones , Estudio de Asociación del Genoma Completo , Riñón/anomalías , Riñón/embriología , Enfermedades Renales/genética , Ratones Noqueados , Nefronas/embriología , Factores de Transcripción/genética , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/genética , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/metabolismo
2.
Kidney Int ; 104(2): 378-387, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37230223

RESUMEN

Nephronophthisis (NPH) is an autosomal-recessive ciliopathy representing one of the most frequent causes of kidney failure in childhood characterized by a broad clinical and genetic heterogeneity. Applied to one of the worldwide largest cohorts of patients with NPH, genetic analysis encompassing targeted and whole exome sequencing identified disease-causing variants in 600 patients from 496 families with a detection rate of 71%. Of 788 pathogenic variants, 40 known ciliopathy genes were identified. However, the majority of patients (53%) bore biallelic pathogenic variants in NPHP1. NPH-causing gene alterations affected all ciliary modules defined by structural and/or functional subdomains. Seventy six percent of these patients had progressed to kidney failure, of which 18% had an infantile form (under five years) and harbored variants affecting the Inversin compartment or intraflagellar transport complex A. Forty eight percent of patients showed a juvenile (5-15 years) and 34% a late-onset disease (over 15 years), the latter mostly carrying variants belonging to the Transition Zone module. Furthermore, while more than 85% of patients with an infantile form presented with extra-kidney manifestations, it only concerned half of juvenile and late onset cases. Eye involvement represented a predominant feature, followed by cerebellar hypoplasia and other brain abnormalities, liver and skeletal defects. The phenotypic variability was in a large part associated with mutation types, genes and corresponding ciliary modules with hypomorphic variants in ciliary genes playing a role in early steps of ciliogenesis associated with juvenile-to-late onset NPH forms. Thus, our data confirm a considerable proportion of late-onset NPH suggesting an underdiagnosis in adult chronic kidney disease.


Asunto(s)
Ciliopatías , Enfermedades Renales Quísticas , Fallo Renal Crónico , Enfermedades Renales Poliquísticas , Adulto , Humanos , Fallo Renal Crónico/diagnóstico , Enfermedades Renales Poliquísticas/complicaciones , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Mutación , Ciliopatías/genética
3.
medRxiv ; 2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36993625

RESUMEN

Background: Congenital anomalies of the kidney and urinary tract (CAKUT) are the predominant cause for chronic kidney disease below 30 years of age. Many monogenic forms have been discovered mainly due to comprehensive genetic testing like exome sequencing (ES). However, disease-causing variants in known disease-associated genes still only explain a proportion of cases. Aim of this study was to unravel the underlying molecular mechanism of syndromic CAKUT in two multiplex families with presumed autosomal recessive inheritance. Methods and Results: ES in the index individuals revealed two different rare homozygous variants in FOXD2, a transcription factor not previously implicated in CAKUT in humans: a frameshift in family 1 and a missense variant in family 2 with family segregation patterns consistent with autosomal-recessive inheritance. CRISPR/Cas9-derived Foxd2 knock-out (KO) mice presented with bilateral dilated renal pelvis accompanied by renal papilla atrophy while extrarenal features included mandibular, ophthalmologic, and behavioral anomalies, recapitulating the phenotype of humans with FOXD2 dysfunction. To study the pathomechanism of FOXD2-dysfunction-mediated developmental renal defects, in a complementary approach, we generated CRISPR/Cas9-mediated KO of Foxd2 in ureteric-bud-induced mouse metanephric mesenchyme cells. Transcriptomic analyses revealed enrichment of numerous differentially expressed genes important in renal/urogenital development, including Pax2 and Wnt4 as well as gene expression changes indicating a cell identity shift towards a stromal cell identity. Histology of Foxd2 KO mouse kidneys confirmed increased fibrosis. Further, GWAS data (genome-wide association studies) suggests that FOXD2 could play a role for maintenance of podocyte integrity during adulthood. Conclusions: In summary, our data implicate that FOXD2 dysfunction is a very rare cause of autosomal recessive syndromic CAKUT and suggest disturbances of the PAX2-WNT4 cell signaling axis contribute to this phenotype.

4.
Hum Mutat ; 43(3): 347-361, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35005812

RESUMEN

We report the screening of a large panel of genes in a series of 100 fetuses (98 families) affected with severe renal defects. Causative variants were identified in 22% of cases, greatly improving genetic counseling. The percentage of variants explaining the phenotype was different according to the type of phenotype. The highest diagnostic yield was found in cases affected with the ciliopathy-like phenotype (11/15 families and, in addition, a single heterozygous or a homozygous Class 3 variant in PKHD1 in three unrelated cases with autosomal recessive polycystic kidney disease). The lowest diagnostic yield was observed in cases with congenital anomalies of the kidney and urinary tract (9/78 families and, in addition, Class 3 variants in GREB1L in three unrelated cases with bilateral renal agenesis). Inheritance was autosomal recessive in nine genes (PKHD1, NPHP3, CEP290, TMEM67, DNAJB11, FRAS1, ACE, AGT, and AGTR1), and autosomal dominant in six genes (PKD1, PKD2, PAX2, EYA1, BICC1, and MYOCD). Finally, we developed an original approach of next-generation sequencing targeted RNA sequencing using the custom capture panel used for the sequencing of DNA, to validate one MYOCD heterozygous splicing variant identified in two male siblings with megabladder and inherited from their healthy mother.


Asunto(s)
Enfermedades Renales , Riñón Poliquístico Autosómico Dominante , Antígenos de Neoplasias , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Femenino , Feto/anomalías , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Riñón/anomalías , Enfermedades Renales/congénito , Enfermedades Renales/diagnóstico , Enfermedades Renales/genética , Masculino , Mutación , Riñón Poliquístico Autosómico Dominante/genética
5.
Pediatr Nephrol ; 36(8): 2361-2369, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33580824

RESUMEN

BACKGROUND: Co-occurrence of polycystic kidney disease and hyperinsulinemic hypoglycemia has been reported in children in a few families associated with a variant in the promotor of the PMM2 gene, at position -167 upstream of the coding sequence. PMM2 encodes phosphomannomutase 2, a key enzyme in N-glycosylation. While biallelic coding PMM2 mutations are involved in congenital disorder of glycosylation CDG1A, that particular variant in the promoter of the gene, either in the homozygous state or associated with a mutation in the coding exons of the gene, is thought to restrict the N-glycosylation defect to the kidney and the pancreas. METHODS: Targeted exome sequencing of a panel of genes involved in monogenic kidney diseases. RESULTS: We identified a PMM2 variant at position -167 associated with a pathogenic PMM2 variant in the coding exons in 3 families, comprising 6 cases affected with a cystic kidney disease. The spectrum of phenotypes was very broad, from extremely enlarged fetal cystic kidneys in the context of a COACH-like syndrome, to isolated cystic kidney disease with small kidneys, slowly progressing toward kidney failure in adulthood. Hypoglycemia was reported only in one case. CONCLUSION: These data show that the PMM2 promotor variation, in trans of a PMM2 coding mutation, is associated with a wide spectrum of kidney phenotypes, and is not always associated with extra-renal symptoms. When present, extra-renal defects may include COACH-like syndrome. These data prompt screening of PMM2 in unresolved cases of fetal hyperechogenic/cystic kidneys as well as in cystic kidney disease in children and adults. Graphical Abstract.


Asunto(s)
Enfermedades Renales Poliquísticas , Hiperinsulinismo Congénito , Humanos , Mutación , Fenotipo , Fosfotransferasas (Fosfomutasas) , Regiones Promotoras Genéticas , Síndrome
6.
Hum Mol Genet ; 28(16): 2720-2737, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31042281

RESUMEN

Mutations in genes encoding components of the intraflagellar transport (IFT) complexes have previously been associated with a spectrum of diseases collectively termed ciliopathies. Ciliopathies relate to defects in the formation or function of the cilium, a sensory or motile organelle present on the surface of most cell types. IFT52 is a key component of the IFT-B complex and ensures the interaction of the two subcomplexes, IFT-B1 and IFT-B2. Here, we report novel IFT52 biallelic mutations in cases with a short-rib thoracic dysplasia (SRTD) or a congenital anomaly of kidney and urinary tract (CAKUT). Combining in vitro and in vivo studies in zebrafish, we showed that SRTD-associated missense mutation impairs IFT-B complex assembly and IFT-B2 ciliary localization, resulting in decreased cilia length. In comparison, CAKUT-associated missense mutation has a mild pathogenicity, thus explaining the lack of skeletal defects in CAKUT case. In parallel, we demonstrated that the previously reported homozygous nonsense IFT52 mutation associated with Sensenbrenner syndrome [Girisha et al. (2016) A homozygous nonsense variant in IFT52 is associated with a human skeletal ciliopathy. Clin. Genet., 90, 536-539] leads to exon skipping and results in a partially functional protein. Finally, our work uncovered a novel role for IFT52 in microtubule network regulation. We showed that IFT52 interacts and partially co-localized with centrin at the distal end of centrioles where it is involved in its recruitment and/or maintenance. Alteration of this function likely contributes to centriole splitting observed in Ift52-/- cells. Altogether, our findings allow a better comprehensive genotype-phenotype correlation among IFT52-related cases and revealed a novel, extra-ciliary role for IFT52, i.e. disruption may contribute to pathophysiological mechanisms.


Asunto(s)
Proteínas Portadoras/genética , Centrosoma/metabolismo , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Microtúbulos/metabolismo , Mutación , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Niño , Preescolar , Cilios/metabolismo , Consanguinidad , Análisis Mutacional de ADN , Femenino , Genotipo , Homocigoto , Humanos , Lactante , Péptidos y Proteínas de Señalización Intracelular , Masculino , Linaje , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Combinación Trimetoprim y Sulfametoxazol/metabolismo , Secuenciación del Exoma , Pez Cebra
7.
Hum Mol Genet ; 28(5): 778-795, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388224

RESUMEN

Mutations in KIF14 have previously been associated with either severe, isolated or syndromic microcephaly with renal hypodysplasia (RHD). Syndromic microcephaly-RHD was strongly reminiscent of clinical ciliopathies, relating to defects of the primary cilium, a signalling organelle present on the surface of many quiescent cells. KIF14 encodes a mitotic kinesin, which plays a key role at the midbody during cytokinesis and has not previously been shown to be involved in cilia-related functions. Here, we analysed four families with fetuses presenting with the syndromic form and harbouring biallelic variants in KIF14. Our functional analyses showed that the identified variants severely impact the activity of KIF14 and likely correspond to loss-of-function mutations. Analysis in human fetal tissues further revealed the accumulation of KIF14-positive midbody remnants in the lumen of ureteric bud tips indicating a shared function of KIF14 during brain and kidney development. Subsequently, analysis of a kif14 mutant zebrafish line showed a conserved role for this mitotic kinesin. Interestingly, ciliopathy-associated phenotypes were also present in mutant embryos, supporting a potential direct or indirect role for KIF14 at cilia. However, our in vitro and in vivo analyses did not provide evidence of a direct role for KIF14 in ciliogenesis and suggested that loss of kif14 causes ciliopathy-like phenotypes through an accumulation of mitotic cells in ciliated tissues. Altogether, our results demonstrate that KIF14 mutations result in a severe syndrome associating microcephaly and RHD through its conserved function in cytokinesis during kidney and brain development.


Asunto(s)
Anomalías Congénitas/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Enfermedades Renales/congénito , Riñón/anomalías , Cinesinas/genética , Mutación con Pérdida de Función , Microcefalia/genética , Proteínas Oncogénicas/genética , Animales , Anomalías Congénitas/metabolismo , Citocinesis/genética , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Genes Letales , Estudios de Asociación Genética/métodos , Sitios Genéticos , Humanos , Riñón/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Masculino , Microcefalia/metabolismo , Microcefalia/patología , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Linaje , Fenotipo , Relación Estructura-Actividad , Pez Cebra
8.
Am J Hum Genet ; 101(5): 803-814, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100091

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) constitute a major cause of chronic kidney disease in children and 20% of prenatally detected anomalies. CAKUT encompass a spectrum of developmental kidney defects, including renal agenesis, hypoplasia, and cystic and non-cystic dysplasia. More than 50 genes have been reported as mutated in CAKUT-affected case subjects. However, the pathophysiological mechanisms leading to bilateral kidney agenesis (BKA) remain largely elusive. Whole-exome or targeted exome sequencing of 183 unrelated familial and/or severe CAKUT-affected case subjects, including 54 fetuses with BKA, led to the identification of 16 heterozygous variants in GREB1L (growth regulation by estrogen in breast cancer 1-like), a gene reported as a target of retinoic acid signaling. Four loss-of-function and 12 damaging missense variants, 14 being absent from GnomAD, were identified. Twelve of them were present in familial or simplex BKA-affected case subjects. Female BKA-affected fetuses also displayed uterus agenesis. We demonstrated a significant association between GREB1L variants and BKA. By in situ hybridization, we showed expression of Greb1l in the nephrogenic zone in developing mouse kidney. We generated a Greb1l knock-out mouse model by CRISPR-Cas9. Analysis at E13.5 revealed lack of kidneys and genital tract anomalies in male and female Greb1l-/- embryos and a slight decrease in ureteric bud branching in Greb1l+/- embryos. We showed that Greb1l invalidation in mIMCD3 cells affected tubulomorphogenesis in 3D-collagen culture, a phenotype rescued by expression of the wild-type human protein. This demonstrates that GREB1L plays a major role in early metanephros and genital development in mice and humans.


Asunto(s)
Anomalías Congénitas/genética , Enfermedades Renales/congénito , Riñón/anomalías , Mutación/genética , Proteínas de Neoplasias/genética , Proteínas/genética , Animales , Niño , Exoma/genética , Femenino , Feto/anomalías , Heterocigoto , Humanos , Enfermedades Renales/genética , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Sistema Urinario/anomalías , Anomalías Urogenitales/genética
9.
J Am Soc Nephrol ; 28(10): 2901-2914, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28566479

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) occur in three to six of 1000 live births, represent about 20% of the prenatally detected anomalies, and constitute the main cause of CKD in children. These disorders are phenotypically and genetically heterogeneous. Monogenic causes of CAKUT in humans and mice have been identified. However, despite high-throughput sequencing studies, the cause of the disease remains unknown in most patients, and several studies support more complex inheritance and the role of environmental factors and/or epigenetics in the pathophysiology of CAKUT. Here, we report the targeted exome sequencing of 330 genes, including genes known to be involved in CAKUT and candidate genes, in a cohort of 204 unrelated patients with CAKUT; 45% of the patients were severe fetal cases. We identified pathogenic mutations in 36 of 204 (17.6%) patients. These mutations included five de novo heterozygous loss of function mutations/deletions in the PBX homeobox 1 gene (PBX1), a gene known to have a crucial role in kidney development. In contrast, the frequency of SOX17 and DSTYK variants recently reported as pathogenic in CAKUT did not indicate causality. These findings suggest that PBX1 is involved in monogenic CAKUT in humans and call into question the role of some gene variants recently reported as pathogenic in CAKUT. Targeted exome sequencing also proved to be an efficient and cost-effective strategy to identify pathogenic mutations and deletions in known CAKUT genes.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas Proto-Oncogénicas/genética , Anomalías Urogenitales/genética , Estudios de Cohortes , Análisis Mutacional de ADN , Exoma , Femenino , Humanos , Masculino , Factor de Transcripción 1 de la Leucemia de Células Pre-B
10.
Hum Mutat ; 37(10): 1025-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27319779

RESUMEN

Neonatal sclerosing cholangitis (NSC) is a rare biliary disease leading to liver transplantation in childhood. Patients with NSC and ichtyosis have already been identified with a CLDN1 mutation, encoding a tight-junction protein. However, for the majority of patients, the molecular basis of NSC remains unknown. We identified biallelic missense mutations or in-frame deletion in DCDC2 in four affected children. Mutations involve highly conserved amino acids in the doublecortin domains of the protein. In cholangiocytes, DCDC2 protein is normally located in the cytoplasm and cilia, whereas in patients the mutated protein is accumulated in the cytoplasm, absent from cilia, and associated with ciliogenesis defect. This is the first report of DCDC2 mutations in NSC. This data expands the molecular spectrum of NSC, that can be considered as a ciliopathy and also expands the clinical spectrum of the DCDC2 mutations, previously reported in dyslexia, deafness, and nephronophtisis.


Asunto(s)
Colangitis Esclerosante/genética , Cilios/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Colangitis Esclerosante/metabolismo , Citoplasma/metabolismo , Femenino , Humanos , Masculino , Proteínas Asociadas a Microtúbulos/química , Mutación Missense , Linaje , Dominios Proteicos , Eliminación de Secuencia
11.
PLoS Genet ; 12(3): e1005894, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26967905

RESUMEN

Ciliopathies are a group of genetic multi-systemic disorders related to dysfunction of the primary cilium, a sensory organelle present at the cell surface that regulates key signaling pathways during development and tissue homeostasis. In order to identify novel genes whose mutations would cause severe developmental ciliopathies, >500 patients/fetuses were analyzed by a targeted high throughput sequencing approach allowing exome sequencing of >1200 ciliary genes. NEK8/NPHP9 mutations were identified in five cases with severe overlapping phenotypes including renal cystic dysplasia/hypodysplasia, situs inversus, cardiopathy with hypertrophic septum and bile duct paucity. These cases highlight a genotype-phenotype correlation, with missense and nonsense mutations associated with hypodysplasia and enlarged cystic organs, respectively. Functional analyses of NEK8 mutations in patient fibroblasts and mIMCD3 cells showed that these mutations differentially affect ciliogenesis, proliferation/apoptosis/DNA damage response, as well as epithelial morphogenesis. Notably, missense mutations exacerbated some of the defects due to NEK8 loss of function, highlighting their likely gain-of-function effect. We also showed that NEK8 missense and loss-of-function mutations differentially affect the regulation of the main Hippo signaling effector, YAP, as well as the expression of its target genes in patient fibroblasts and renal cells. YAP imbalance was also observed in enlarged spheroids of Nek8-invalidated renal epithelial cells grown in 3D culture, as well as in cystic kidneys of Jck mice. Moreover, co-injection of nek8 MO with WT or mutated NEK8-GFP RNA in zebrafish embryos led to shortened dorsally curved body axis, similar to embryos injected with human YAP RNA. Finally, treatment with Verteporfin, an inhibitor of YAP transcriptional activity, partially rescued the 3D spheroid defects of Nek8-invalidated cells and the abnormalities of NEK8-overexpressing zebrafish embryos. Altogether, our study demonstrates that NEK8 human mutations cause major organ developmental defects due to altered ciliogenesis and cell differentiation/proliferation through deregulation of the Hippo pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Cilios/genética , Fosfoproteínas/genética , Enfermedades Renales Poliquísticas/genética , Proteínas Quinasas/genética , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Diferenciación Celular/genética , Cilios/patología , Femenino , Estudios de Asociación Genética , Humanos , Riñón/metabolismo , Riñón/patología , Ratones , Morfogénesis/genética , Mutación , Quinasas Relacionadas con NIMA , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/biosíntesis , Enfermedades Renales Poliquísticas/patología , Porfirinas/administración & dosificación , Transducción de Señal , Factores de Transcripción , Verteporfina , Proteínas Señalizadoras YAP , Pez Cebra
12.
Hum Genet ; 135(1): 69-87, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26572137

RESUMEN

Congenital anomalies of the kidneys and urinary tract (CAKUT) are genetically highly heterogeneous leaving most cases unclear after mutational analysis of the around 30 causative genes known so far. Assuming that phenotypes frequently showing dominant inheritance, such as CAKUT, can be caused by de novo mutations, de novo analysis of whole-exome sequencing data was done on two patient-parent-trios to identify novel CAKUT genes. In one case, we detected a heterozygous de novo frameshift variant in TBC1D1 encoding a Rab-GTPase-activating protein regulating glucose transporter GLUT4 translocation. Sequence analysis of 100 further CAKUT cases yielded three novel or rare inherited heterozygous TBC1D1 missense variants predicted to be pathogenic. TBC1D1 mutations affected Ser237-phosphorylation or protein stability and thereby act as hypomorphs. Tbc1d1 showed widespread expression in the developing murine urogenital system. A mild CAKUT spectrum phenotype, including anomalies observed in patients carrying TBC1D1 mutations, was found in kidneys of some Tbc1d1 (-/-) mice. Significantly reduced Glut4 levels were detected in kidneys of Tbc1d1 (-/-) mice and the dysplastic kidney of a TBC1D1 mutation carrier versus controls. TBC1D1 and SLC2A4 encoding GLUT4 were highly expressed in human fetal kidney. The patient with the truncating TBC1D1 mutation showed evidence for insulin resistance. These data demonstrate heterozygous deactivating TBC1D1 mutations in CAKUT patients with a similar renal and ureteral phenotype, and provide evidence that TBC1D1 mutations may contribute to CAKUT pathogenesis, possibly via a role in glucose homeostasis.


Asunto(s)
Exoma , Proteínas Activadoras de GTPasa/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mutación , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Niño , Preescolar , Femenino , Proteínas Activadoras de GTPasa/química , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Linaje , Homología de Secuencia de Aminoácido , Adulto Joven
13.
Dis Model Mech ; 8(8): 903-17, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26035382

RESUMEN

Wilms' tumours, paediatric kidney cancers, are the archetypal example of tumours caused through the disruption of normal development. The genetically best-defined subgroup of Wilms' tumours is the group caused by biallelic loss of the WT1 tumour suppressor gene. Here, we describe a developmental series of mouse models with conditional loss of Wt1 in different stages of nephron development before and after the mesenchymal-to-epithelial transition (MET). We demonstrate that Wt1 is essential for normal development at all kidney developmental stages under study. Comparison of genome-wide expression data from the mutant mouse models with human tumour material of mutant or wild-type WT1 datasets identified the stage of origin of human WT1-mutant tumours, and emphasizes fundamental differences between the two human tumour groups due to different developmental stages of origin.


Asunto(s)
Nefronas/crecimiento & desarrollo , Nefronas/metabolismo , Proteínas WT1/metabolismo , Tumor de Wilms/patología , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Regulación Neoplásica de la Expresión Génica , Genoma , Integrasas/metabolismo , Ratones Endogámicos C57BL , Ratones Mutantes , Estadificación de Neoplasias , Nefronas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Imagen de Lapso de Tiempo , Proteínas WT1/genética , Tumor de Wilms/genética
14.
Am J Hum Genet ; 94(2): 288-94, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24439109

RESUMEN

Renal hypodysplasia (RHD) is a heterogeneous condition encompassing a spectrum of kidney development defects including renal agenesis, hypoplasia, and (cystic) dysplasia. Heterozygous mutations of several genes have been identified as genetic causes of RHD with various severity. However, these genes and mutations are not associated with bilateral renal agenesis, except for RET mutations, which could be involved in a few cases. The pathophysiological mechanisms leading to total absence of kidney development thus remain largely elusive. By using a whole-exome sequencing approach in families with several fetuses with bilateral renal agenesis, we identified recessive mutations in the integrin α8-encoding gene ITGA8 in two families. Itga8 homozygous knockout in mice is known to result in absence of kidney development. We provide evidence of a damaging effect of the human ITGA8 mutations. These results demonstrate that mutations of ITGA8 are a genetic cause of bilateral renal agenesis and that, at least in some cases, bilateral renal agenesis is an autosomal-recessive disease.


Asunto(s)
Anomalías Congénitas/genética , Genes Recesivos , Cadenas alfa de Integrinas/genética , Enfermedades Renales/congénito , Riñón/anomalías , Anomalías Urogenitales/genética , Anomalías Congénitas/patología , Femenino , Feto/anomalías , Homocigoto , Humanos , Cadenas alfa de Integrinas/metabolismo , Riñón/patología , Enfermedades Renales/genética , Enfermedades Renales/patología , Masculino , Mutación , Linaje , Anomalías Urogenitales/patología
16.
Clin J Am Soc Nephrol ; 8(7): 1179-87, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23539225

RESUMEN

BACKGROUND AND OBJECTIVES: Congenital anomalies of the kidney and urinary tract (CAKUT) are a frequent cause of renal failure in children, and their detection in utero is now common with fetal screening ultrasonography. The clinical course of CAKUT detected before birth is very heterogeneous and depends on the level of nephron reduction. The most severe forms cause life-threatening renal failure, leading to perinatal death or the need for very early renal replacement therapy. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: This study reports the screening of two genes (HNF1B and PAX2) involved in monogenic syndromic CAKUT in a cohort of 103 fetuses from 91 families with very severe CAKUT that appeared isolated by fetal ultrasound examination and led to termination of pregnancy. RESULTS: This study identified a disease-causing mutation in HNF1B in 12 cases from 11 families and a mutation in PAX2 in 4 unrelated cases. Various renal phenotypes were observed, but no case of bilateral agenesis was associated with HNF1B or PAX2 mutations. Autopsy identified extrarenal abnormalities not detected by ultrasonography in eight cases but confirmed the absence of extrarenal defects in eight other cases. A positive family history of renal disease was not significantly more frequent in cases with an identified mutation. Moreover, in cases with an inherited mutation, there was a great phenotypic variability regarding the severity of the renal disease within a single family. CONCLUSIONS: Our results suggest that mutations in genes involved in syndromic CAKUT with Mendelian inheritance are not rare in fetal cases with severe CAKUT appearing isolated at prenatal ultrasound, a finding of clinical importance because of genetic counseling.


Asunto(s)
Factor Nuclear 1-beta del Hepatocito/genética , Mutación , Factor de Transcripción PAX2/genética , Diagnóstico Prenatal , Reflujo Vesicoureteral/diagnóstico , Reflujo Vesicoureteral/genética , Aborto Terapéutico , Autopsia , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Herencia , Humanos , Masculino , Linaje , Fenotipo , Valor Predictivo de las Pruebas , Embarazo , Diagnóstico Prenatal/métodos , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Ultrasonografía Prenatal , Anomalías Urogenitales
17.
Eur J Hum Genet ; 21(7): 784-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23169491

RESUMEN

Nephroblastoma (Wilms' tumor; WT) is the most common renal tumor of childhood. To date, several genetic abnormalities predisposing to WT have been identified in rare overgrowth syndromes. Among them, abnormal methylation of the 11p15 region, GPC3 and DIS3L2 mutations, which are responsible for Beckwith-Wiedemann, Simpson-Golabi-Behmel and Perlman syndromes, respectively. However, the underlying cause of WT remains unknown in the majority of cases. We report three unrelated patients who presented with WT in addition to a constitutional 9q22.3 microdeletion and dysmorphic/overgrowth syndrome. The size of the deletions was variable (ie, from 1.7 to 8.9 Mb) but invariably encompassed the PTCH1 gene. Subsequently, we identified a somatic PTCH1 nonsense mutation in the renal tumor of one patient. In addition, by array comparative genomic hybridization method, we analyzed the DNA extracted from the blood samples of nine patients with overgrowth syndrome and WT, but did not identify any deleterious chromosomal imbalances in these patients. These findings strongly suggest that patients with constitutional 9q22.3 microdeletion have an increased risk of WT, and that PTCH1 have a role in the pathogenesis of nephroblastomas.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 9/genética , Macrosomía Fetal/genética , Neoplasias Renales/genética , Receptores de Superficie Celular/genética , Tumor de Wilms/genética , Adolescente , Adulto , Niño , Hibridación Genómica Comparativa , Femenino , Macrosomía Fetal/patología , Humanos , Neoplasias Renales/patología , Mutación , Receptores Patched , Receptor Patched-1 , Embarazo , Tumor de Wilms/patología
18.
Dev Cell ; 22(6): 1191-207, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22698282

RESUMEN

The identity of niche signals necessary to maintain embryonic nephron progenitors is unclear. Here we provide evidence that Fgf20 and Fgf9, expressed in the niche, and Fgf9, secreted from the adjacent ureteric bud, are necessary and sufficient to maintain progenitor stemness. Reduction in the level of these redundant ligands in the mouse led to premature progenitor differentiation within the niche. Loss of FGF20 in humans, or of both ligands in mice, resulted in kidney agenesis. Sufficiency was shown in vitro where Fgf20 or Fgf9 (alone or together with Bmp7) maintained isolated metanephric mesenchyme or sorted nephron progenitors that remained competent to differentiate in response to Wnt signals after 5 or 2 days in culture, respectively. These findings identify a long-sought-after critical component of the nephron stem cell niche and hold promise for long-term culture and utilization of these progenitors in vitro.


Asunto(s)
Diferenciación Celular , Factor 9 de Crecimiento de Fibroblastos/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Animales , Proteína Morfogenética Ósea 7/fisiología , Anomalías Congénitas/genética , Femenino , Factores de Crecimiento de Fibroblastos/genética , Humanos , Riñón/anomalías , Riñón/crecimiento & desarrollo , Enfermedades Renales/congénito , Enfermedades Renales/genética , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Mutación , Nefronas/crecimiento & desarrollo , Técnicas de Cultivo de Órganos , Nicho de Células Madre/fisiología , Vía de Señalización Wnt
19.
Nephrol Dial Transplant ; 26(12): 3843-51, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22121240

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are the commonest cause of chronic kidney disease in children. Structural anomalies within the CAKUT spectrum include renal agenesis, kidney hypo-/dysplasia, multicystic kidney dysplasia, duplex collecting system, posterior urethral valves and ureter abnormalities. While most CAKUT cases are sporadic, familial clustering of CAKUT is common, emphasizing a strong genetic contribution to CAKUT origin. Animal experiments demonstrate that alterations in genes crucial for kidney development can cause experimental CAKUT, while expression studies implicate mislocalization and/or aberrant levels of the encoded proteins in human CAKUT. Further insight into the pathogenesis of CAKUT will improve strategies for early diagnosis, follow-up and treatment. Here, we outline a collaborative approach to identify and characterize novel factors underlying human CAKUT. This European consortium will share the largest collection of CAKUT patients available worldwide and undertake multidisciplinary research into molecular and genetic pathogenesis, with extension into translational studies to improve long-term patient outcomes.


Asunto(s)
Sistema Urinario/anomalías , Animales , Investigación Biomédica/tendencias , Anomalías Congénitas/diagnóstico , Anomalías Congénitas/etiología , Humanos , Riñón/anomalías , Riñón/crecimiento & desarrollo , Sistema Urinario/crecimiento & desarrollo
20.
Am J Hum Genet ; 89(5): 634-43, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22019273

RESUMEN

A subset of ciliopathies, including Sensenbrenner, Jeune, and short-rib polydactyly syndromes are characterized by skeletal anomalies accompanied by multiorgan defects such as chronic renal failure and retinitis pigmentosa. Through exome sequencing we identified compound heterozygous mutations in WDR19 in a Norwegian family with Sensenbrenner syndrome. In a Dutch family with the clinically overlapping Jeune syndrome, a homozygous missense mutation in the same gene was found. Both families displayed a nephronophthisis-like nephropathy. Independently, we also identified compound heterozygous WDR19 mutations by exome sequencing in a Moroccan family with isolated nephronophthisis. WDR19 encodes IFT144, a member of the intraflagellar transport (IFT) complex A that drives retrograde ciliary transport. We show that IFT144 is absent from the cilia of fibroblasts from one of the Sensenbrenner patients and that ciliary abundance and morphology is perturbed, demonstrating the ciliary pathogenesis. Our results suggest that isolated nephronophthisis, Jeune, and Sensenbrenner syndromes are clinically overlapping disorders that can result from a similar molecular cause.


Asunto(s)
Cilios , Displasia Ectodérmica/genética , Mutación Missense , Enfermedades Renales Poliquísticas/genética , Proteínas/genética , Síndrome de Costilla Pequeña y Polidactilia/genética , Enfermedades Torácicas/genética , Adolescente , Adulto , Niño , Cilios/genética , Cilios/patología , Anomalías Craneofaciales/genética , Proteínas del Citoesqueleto , Exoma/genética , Femenino , Fibroblastos/metabolismo , Flagelos/genética , Flagelos/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Datos de Secuencia Molecular , Marruecos , Países Bajos , Noruega , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Enfermedades Renales Poliquísticas/congénito , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA