Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Genet ; 12(2): e1005785, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26859289

RESUMO

Rho family GTPases act as molecular switches regulating actin cytoskeleton dynamics. Attenuation of their signaling capacity is provided by GTPase-activating proteins (GAPs), including p190A, that promote the intrinsic GTPase activity of Rho proteins. In the current study we have performed a small-scale ENU mutagenesis screen and identified a novel loss of function allele of the p190A gene Arhgap35, which introduces a Leu1396 to Gln substitution in the GAP domain. This results in decreased GAP activity for the prototypical Rho-family members, RhoA and Rac1, likely due to disrupted ordering of the Rho binding surface. Consequently, Arhgap35-deficient animals exhibit hypoplastic and glomerulocystic kidneys. Investigation into the cystic phenotype shows that p190A is required for appropriate primary cilium formation in renal nephrons. P190A specifically localizes to the base of the cilia to permit axoneme elongation, which requires a functional GAP domain. Pharmacological manipulations further reveal that inhibition of either Rho kinase (ROCK) or F-actin polymerization is able to rescue the ciliogenesis defects observed upon loss of p190A activity. We propose a model in which p190A acts as a modulator of Rho GTPases in a localized area around the cilia to permit the dynamic actin rearrangement required for cilia elongation. Together, our results establish an unexpected link between Rho GTPase regulation, ciliogenesis and glomerulocystic kidney disease.


Assuntos
Cílios/metabolismo , Proteínas Ativadoras de GTPase/genética , Doenças Renais Císticas/genética , Glomérulos Renais/patologia , Organogênese , Mutação Puntual/genética , Proteínas Repressoras/genética , Actinas/metabolismo , Alelos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Citoesqueleto/metabolismo , Embrião de Mamíferos/citologia , Etilnitrosoureia , Feminino , Fibroblastos/metabolismo , Proteínas Ativadoras de GTPase/química , Proteínas Ativadoras de GTPase/metabolismo , Doenças Renais Císticas/patologia , Glomérulos Renais/metabolismo , Túbulos Renais/anormalidades , Túbulos Renais/patologia , Masculino , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Defeitos do Tubo Neural/patologia , Fenótipo , Estrutura Terciária de Proteína , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Reprodutibilidade dos Testes
2.
Development ; 142(15): 2696-703, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26243870

RESUMO

Despite the high occurrence of congenital abnormalities of the lower urinary tract in humans, the molecular, cellular and morphological aspects of their development are still poorly understood. Here, we use a conditional knockout approach to inactivate within the nephric duct (ND) lineage the two effectors of the Hippo pathway, Yap and Taz. Deletion of Yap leads to hydronephrotic kidneys with blind-ending megaureters at birth. In Yap mutants, the ND successfully migrates towards, and contacts, the cloaca. However, close analysis reveals that the tip of the Yap(-/-) ND forms an aberrant connection with the cloaca and does not properly insert into the cloaca, leading to later detachment of the ND from the cloaca. Taz deletion from the ND does not cause any defect, but analysis of Yap(-/-);Taz(-/-) NDs indicates that both genes play partially redundant roles in ureterovesical junction formation. Aspects of the Yap(-/-) phenotype resemble hypersensitivity to RET signaling, including excess budding of the ND, increased phospho-ERK and increased expression of Crlf1, Sprouty1, Etv4 and Etv5. Importantly, the Yap(ND) (-/-) ND phenotype can be largely rescued by reducing Ret gene dosage. Taken together, these results suggest that disrupting Yap/Taz activities enhances Ret pathway activity and contributes to pathogenesis of lower urinary tract defects in human infants.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Morfogênese/fisiologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Transdução de Sinais/fisiologia , Sistema Urinário/embriologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Bromodesoxiuridina , Proteínas de Ciclo Celular , Imunofluorescência , Galactosídeos , Técnicas de Inativação de Genes , Humanos , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Indóis , Camundongos , Fosfoproteínas/genética , Transativadores , Proteínas de Sinalização YAP
3.
Dev Biol ; 382(2): 555-66, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23920117

RESUMO

Translating the developmental program encoded in the genome into cellular and morphogenetic functions requires the deployment of elaborate gene regulatory networks (GRNs). GRNs are especially crucial at the onset of organ development where a few regulatory signals establish the different programs required for tissue organization. In the renal system primordium (the pro/mesonephros), important regulators have been identified but their hierarchical and regulatory organization is still elusive. Here, we have performed a detailed analysis of the GRN underlying mouse pro/mesonephros development. We find that a core regulatory subcircuit composed of Pax2/8, Gata3 and Lim1 turns on a deeper layer of transcriptional regulators while activating effector genes responsible for cell signaling and tissue organization. Among the genes directly affected by the core components are the key developmental molecules Nephronectin (Npnt) and Plac8. Hence, the pro/mesonephros GRN links together several essential genes regulating tissue morphogenesis. This renal GRN sheds new light on the disease group Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) in that gene mutations are expected to generate different phenotypic outcomes as a consequence of regulatory network deficiencies rather than threshold effects from single genes.


Assuntos
Fator de Transcrição GATA3/genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Proteínas com Homeodomínio LIM/genética , Mesonefro/embriologia , Fator de Transcrição PAX2/genética , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição/genética , Animais , Linhagem Celular , Rim/anormalidades , Mesonefro/metabolismo , Camundongos , Morfogênese/genética , Fator de Transcrição PAX8
4.
PLoS Genet ; 4(12): e1000316, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19112489

RESUMO

Metanephric kidney induction critically depends on mesenchymal-epithelial interactions in the caudal region of the nephric (or Wolffian) duct. Central to this process, GDNF secreted from the metanephric mesenchyme induces ureter budding by activating the Ret receptor expressed in the nephric duct epithelium. A failure to regulate this pathway is believed to be responsible for a large proportion of the developmental anomalies affecting the urogenital system. Here, we show that the nephric duct-specific inactivation of the transcription factor gene Gata3 leads to massive ectopic ureter budding. This results in a spectrum of urogenital malformations including kidney adysplasia, duplex systems, and hydroureter, as well as vas deferens hyperplasia and uterine agenesis. The variability of developmental defects is reminiscent of the congenital anomalies of the kidney and urinary tract (CAKUT) observed in human. We show that Gata3 inactivation causes premature nephric duct cell differentiation and loss of Ret receptor gene expression. These changes ultimately affect nephric duct epithelium homeostasis, leading to ectopic budding of interspersed cells still expressing the Ret receptor. Importantly, the formation of these ectopic buds requires both GDNF/Ret and Fgf signaling activities. We further identify Gata3 as a central mediator of beta-catenin function in the nephric duct and demonstrate that the beta-catenin/Gata3 pathway prevents premature cell differentiation independently of its role in regulating Ret expression. Together, these results establish a genetic cascade in which Gata3 acts downstream of beta-catenin, but upstream of Ret, to prevent ectopic ureter budding and premature cell differentiation in the nephric duct.


Assuntos
Fator de Transcrição GATA3/metabolismo , Rim/embriologia , Transdução de Sinais , Ductos Mesonéfricos/anormalidades , beta Catenina/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Fator de Transcrição GATA3/genética , Humanos , Rim/anormalidades , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ureter/anormalidades , Ureter/citologia , Ureter/metabolismo , Ductos Mesonéfricos/embriologia , Ductos Mesonéfricos/crescimento & desenvolvimento , Ductos Mesonéfricos/metabolismo , beta Catenina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...