Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Biol Chem ; 287(18): 14325-35, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22399302

RESUMO

Under conditions of reduced mitogen or nutritional substrate levels, the serine/threonine kinase target of rapamycin can augment the nuclear content of distinct transcription factors and promote the induction of stress response genes. In its latent (i.e., unphosphorylated) form, the transcription factor STAT1 regulates a subset of genes involved in immune modulation and apoptosis. Based on previous work indicating a functional relationship between mammalian target of rapamycin (mTOR) and the nuclear content of latent STAT1, we investigated the mechanism by which mTOR controls STAT1 nuclear import. By fluorescence confocal microscopy, inactivation of mTOR with rapamycin promoted the nuclear translocation of unphosphorylated STAT1, but not that of a STAT1 mutant incapable of binding its nuclear import adaptor karyopherin-α1 (KPNA1). By immunoprecipitation, KPNA1 was physically associated with mTOR and STAT1 in a complex that translocated to the nucleus in response to rapamycin. Although mTOR is not a kinase for KPNA1, the mTOR-associated phosphatase protein phosphatase 2A catalytic interacted directly with KPNA1 and regulated nuclear import of the mTOR-KPNA1 complex. KPNA1, or its interaction with STAT1, was required for the nuclear import of latent STAT1, transcriptional induction of the STAT1 gene, and caspase-3 activation under conditions of reduced mTOR activity (i.e. rapamycin, glucose starvation, serum withdrawal). Therefore, at low mitogen or nutrient levels, mTOR and protein phosphatase 2A catalytically control the constitutive nuclear import of latent STAT1 by KPNA1, which are key modulators of STAT1 expression and apoptosis.


Assuntos
Núcleo Celular/metabolismo , Serina-Treonina Quinases TOR/metabolismo , alfa Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Apoptose/fisiologia , Caspase 3/fisiologia , Núcleo Celular/genética , Ativação Enzimática/fisiologia , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Mutação , Fosforilação/fisiologia , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Serina-Treonina Quinases TOR/genética , alfa Carioferinas/genética
2.
Am J Physiol Renal Physiol ; 298(4): F951-61, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20071462

RESUMO

The tyrosine phosphorylation of nephrin is reported to regulate podocyte morphology via the Nck adaptor proteins. The Pak family of kinases are regulators of the actin cytoskeleton and are recruited to the plasma membrane via Nck. Here, we investigated the role of Pak in podocyte morphology. Pak1/2 were expressed in cultured podocytes. In mouse podocytes, Pak2 was predominantly phosphorylated, concentrated at the tips of the cellular processes, and its expression and/or phosphorylation were further increased when differentiated. Overexpression of rat nephrin in podocytes increased Pak1/2 phosphorylation, which was abolished when the Nck binding sites were mutated. Furthermore, dominant-negative Nck constructs blocked the Pak1 phosphorylation induced by antibody-mediated cross linking of nephrin. Transient transfection of constitutively kinase-active Pak1 into differentiated mouse podocytes decreased stress fibers, increased cortical F-actin, and extended the cellular processes, whereas kinase-dead mutant, kinase inhibitory construct, and Pak2 knockdown by shRNA had the opposite effect. In a rat model of puromycin aminonucleoside nephrosis, Pak1/2 phosphorylation was decreased in glomeruli, concomitantly with a decrease of nephrin tyrosine phosphorylation. These results suggest that Pak contributes to remodeling of the actin cytoskeleton in podocytes. Disturbed nephrin-Nck-Pak interaction may contribute to abnormal morphology of podocytes and proteinuria.


Assuntos
Actinas/fisiologia , Glomérulos Renais/citologia , Proteínas de Membrana/metabolismo , Podócitos/citologia , Quinases Ativadas por p21/metabolismo , Animais , Antibióticos Antineoplásicos/efeitos adversos , Linhagem Celular , Regulação da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica , Humanos , Isoenzimas , Glomérulos Renais/metabolismo , Proteínas de Membrana/genética , Camundongos , Nefrose/induzido quimicamente , Nefrose/metabolismo , Fosforilação , Plaquinas/fisiologia , Podócitos/metabolismo , Puromicina Aminonucleosídeo/efeitos adversos , Ratos , Quinases Ativadas por p21/genética
3.
Nephron Exp Nephrol ; 114(3): e93-e106, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19955829

RESUMO

BACKGROUND/AIMS: The function of glomerular podocytes is closely associated with the actin cytoskeleton. In this study, we studied the role of the small Rho-GTPase, Rac1, in actin organization in podocytes. METHODS: Conditionally immortalized mouse podocytes (MP) stably expressing nephrin or control plasmid were used. RESULTS: In MP, Rac1 activity increased significantly at 1 week of differentiation. MP stably expressing nephrin showed Rac1 activity significantly higher and more sustained than vector-expressing control cells. Antibody-mediated cross-linking of nephrin also activated Rac1. Differentiated MP showed more distinct lamellipodia/cellular processes, as compared with undifferentiated cells, which was further augmented by nephrin expression. Transient transfection of constitutively active Rac1 markedly increased the number of lamellipodia/cellular processes in undifferentiated MP, while the Rac1 inhibitor caused actin cytoskeleton derangement in differentiating MP. In the rat model of puromycin aminonucleoside nephrosis, RhoA activity was increased at Day 7 (at the peak of proteinuria), while Rac1 activity increased significantly only at Day 14, when the recovery process had started. CONCLUSION: Rac1 is activated in differentiating MP and nephrin potentiates Rac1 activation. Rac1 likely contributes to lamellipodia formation in differentiating MP and may contribute to process formation in podocytes recovering from injuries.


Assuntos
Actinas/metabolismo , Glomérulos Renais/citologia , Podócitos/metabolismo , Proteínas rac1 de Ligação ao GTP/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Ativação Enzimática , Glomérulos Renais/metabolismo , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/farmacologia , Camundongos , Nefrose/induzido quimicamente , Puromicina Aminonucleosídeo , Ratos , Ratos Sprague-Dawley , Proteínas Ativadoras de ras GTPase/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo
5.
Sci Rep ; 9(1): 17620, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31772273

RESUMO

Inactivation of the protein complex 'mechanistic target of rapamycin complex 1' (mTORC1) can increase the nuclear content of transcriptional regulators of metabolism and apoptosis. Previous studies established that nuclear import of signal transducer and activator of transcription-1 (STAT1) requires the mTORC1-associated adaptor karyopherin-α1 (KPNA1) when mTORC1 activity is reduced. However, the role of other mTORC1-interacting proteins in the complex, including 'protein kinase C delta' (PKCδ), have not been well characterized. In this study, we demonstrate that PKCδ, a STAT1 kinase, contains a functional 'target of rapamycin signaling' (TOS) motif that directs its interaction with mTORC1. Depletion of KPNA1 by RNAi prevented the nuclear import of PKCδ in cells exposed to the mTORC1 inhibitor rapamycin or amino acid restriction. Mutation of the TOS motif in PKCδ led to its loss of regulation by mTORC1 or karyopherin-α1, resulting in increased constitutive nuclear content. In cells expressing wild-type PKCδ, STAT1 activity and apoptosis were increased by rapamycin or interferon-ß. Those expressing the PKCδ TOS mutant exhibited increased STAT1 activity and apoptosis; further enhancement by rapamycin or interferon-ß, however, was lost. Therefore, the TOS motif in PKCδ is a novel structural mechanism by which mTORC1 prevents PKCδ and STAT1 nuclear import, and apoptosis.


Assuntos
Núcleo Celular/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteína Quinase C-delta/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Motivos de Aminoácidos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular , Humanos , Modelos Moleculares , Mutação de Sentido Incorreto , Mutação Puntual , Conformação Proteica , Mapeamento de Interação de Proteínas , Proteína Quinase C-delta/química , Proteína Quinase C-delta/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Recombinantes/metabolismo , Proteína Regulatória Associada a mTOR/metabolismo , Fator de Transcrição STAT1/biossíntese , Alinhamento de Sequência , Sirolimo/farmacologia , alfa Carioferinas/antagonistas & inibidores , alfa Carioferinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA