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










Base de dados
Intervalo de ano de publicação
1.
Cardiovasc Res ; 106(3): 398-407, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25852081

RESUMO

AIMS: Increased myocardial wall strain triggers the cardiac hypertrophic response by increasing cardiomyocyte size, reprogramming gene expression, and enhancing contractile protein synthesis. The LIM protein, migfilin, is a cytoskeleton-associated protein that was found to translocate in vitro into the nucleus in a Ca(2+)-dependent manner, where it co-activates the pivotal cardiac transcription factor Csx/Nkx2.5. However, the in vivo role of migfilin in cardiac function and stress response is unclear. METHODS AND RESULTS: To define the role of migfilin in cardiac hypertrophy, we induced hypertension by transverse aortic constriction (TAC) and compared cardiac morphology and function of migfilin knockout (KO) with wild-type (WT) hearts. Heart size and myocardial contractility were comparable in untreated migfilin KO and WT hearts, but migfilin-null hearts presented a reduced extent of hypertrophic remodelling in response to chronic hypertensile stress. Migfilin KO mice maintained their cardiac function for a longer time period compared with WT mice, which presented extensive fibrosis and death due to heart failure. Migfilin translocated into the nucleus of TAC-treated cardiomyocytes, and migfilin KO hearts showed reduced Akt activation during the early response to pressure overload. CONCLUSIONS: Our findings indicate an important role of migfilin in the regulation of cardiac hypertrophy upon experimental TAC.


Assuntos
Moléculas de Adesão Celular/metabolismo , Hipertensão/complicações , Hipertrofia Ventricular Esquerda/metabolismo , Miócitos Cardíacos/metabolismo , Remodelação Ventricular , Transporte Ativo do Núcleo Celular , Animais , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Modelos Animais de Doenças , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrose , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Hipertensão/fisiopatologia , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fatores de Tempo
2.
Cell Cycle ; 13(3): 462-70, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24280829

RESUMO

The spatiotemporal manipulations of gene expression by the Cre recombinase (Cre) of bacteriophage P1 has become an essential asset to understanding mammalian genetics. Accumulating evidence suggests that Cre activity can, in addition to excising targeted loxP sites, induce cytotoxic effects, including abnormal cell cycle progression, genomic instability, and apoptosis, which can accelerate cancer progression. It is speculated that these defects are caused by Cre-induced DNA damage at off-target sites. Here we report the formation of tetraploid keratinocytes in the epidermis of keratin 5 and/or keratin 14 promoter-driven Cre (KRT5- and KRT14-Cre) expressing mouse skin. Biochemical analyses and flow cytometry demonstrated that Cre expression also induces DNA damage, genomic instability, and tetraploidy in HCT116 cells, and live-cell imaging revealed an extension of the G 2 cell cycle phase followed by defective or skipping of mitosis as cause for the tetraploidy. Since tetraploidy eventually leads to aneuploidy, a hallmark of cancer, our findings highlight the importance of distinguishing non-specific cytopathic effects from specific Cre/loxP-driven genetic manipulations when using Cre-mediated gene deletions.


Assuntos
Dano ao DNA , Integrases/metabolismo , Tetraploidia , Proteínas Virais/metabolismo , Animais , Bacteriófago P1/enzimologia , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Citocinese , Células Epidérmicas , Pontos de Checagem da Fase G2 do Ciclo Celular , Instabilidade Genômica , Células HCT116 , Humanos , Integrases/genética , Queratina-14/genética , Queratina-5/genética , Queratinócitos/citologia , Camundongos Transgênicos , Mitose , Regiões Promotoras Genéticas , Proteínas Virais/genética
3.
J Biol Chem ; 288(26): 18863-71, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23658024

RESUMO

Integrin-linked kinase (ILK) localizes to focal adhesions (FAs) where it regulates cell spreading, migration, and growth factor receptor signaling. Previous reports showed that overexpressed ILK in which Val(386) and Thr(387) were substituted with glycine residues (ILK-VT/GG) could neither interact with paxillin nor localize to FA in cells expressing endogenous wild-type ILK, implying that paxillin binding to ILK is required for its localization to FAs. Here, we show that introducing this mutation into the germ line of mice (ILK-VT/GG) caused vasculogenesis defects, resulting in a general developmental delay and death at around embryonic day 12.5. Fibroblasts isolated from ILK-VT/GG mice contained mutant ILK in FAs, showed normal adhesion to and spreading on extracellular matrix substrates but displayed impaired migration. Biochemical analysis revealed that VT/GG substitutions decreased ILK protein stability leading to decreased ILK levels and reduced binding to paxillin and α-parvin. Because paxillin depletion did not affect ILK localization to FAs, the embryonic lethality and the in vitro migration defects are likely due to the reduced levels of ILK-VT/GG and diminished binding to parvins.


Assuntos
Embrião de Mamíferos/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Mutação , Paxilina/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Motivos de Aminoácidos , Animais , Sítios de Ligação , Adesão Celular , Movimento Celular , Citometria de Fluxo , Adesões Focais/metabolismo , Genes Letais , Camundongos , Proteínas dos Microfilamentos/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Fatores de Tempo
4.
J Cell Sci ; 125(Pt 23): 5690-701, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22992463

RESUMO

Efficient wound repair is essential for the maintenance of the integrity of the skin. The repair process is controlled by a variety of growth factors and cytokines, and their abnormal expression or activity can cause healing disorders. Here, we show that wound repair is severely delayed in mice lacking fibroblast growth factor receptors (FGFR) 1 and 2 in keratinocytes. As the underlying mechanism, we identified impaired wound contraction and a delay in re-epithelialization that resulted from impaired keratinocyte migration at the wound edge. Scratch wounding and transwell assays demonstrated that FGFR1/2-deficient keratinocytes had a reduced migration velocity and impaired directional persistence owing to inefficient formation and turnover of focal adhesions. Underlying this defect, we identified a significant reduction in the expression of major focal adhesion components in the absence of FGFR signaling, resulting in a general migratory deficiency. These results identify FGFs as key regulators of keratinocyte migration in wounded skin.


Assuntos
Queratinócitos/citologia , Queratinócitos/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Pele/metabolismo , Pele/patologia , Cicatrização/fisiologia , Animais , Western Blotting , Adesão Celular/genética , Adesão Celular/fisiologia , Movimento Celular/genética , Movimento Celular/fisiologia , Células Cultivadas , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cicatrização/genética
5.
J Cell Sci ; 124(Pt 3): 414-21, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21224394

RESUMO

Migfilin is a LIM-domain-containing protein of the zyxin family of adaptor proteins and is found at cell-matrix and cell-cell adhesion sites and in the nucleus. In vitro studies have suggested that migfilin promotes ß1 integrin activity, regulates cell spreading and migration and induces cardiomyocyte differentiation. To test directly the function of migfilin in vivo, we generated a migfilin-null mouse strain. Here, we report that loss of migfilin expression permits normal development and normal postnatal aging. Fibroblasts and keratinocytes from migfilin-null mice display normal spreading and adhesion, and normal integrin expression and activation. The migration velocity and directionality of migfilin-null embryonic fibroblasts were normal, whereas the velocity of migfilin-null keratinocytes in wound scratch assays was slightly but significantly reduced. Our findings indicate that the roles of migfilin are functionally redundant during mouse development and tissue homeostasis.


Assuntos
Moléculas de Adesão Celular/genética , Adesão Celular/genética , Proteínas do Citoesqueleto/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Integrina beta1/metabolismo , Animais , Moléculas de Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proteínas do Citoesqueleto/fisiologia , Matriz Extracelular/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Proteínas com Domínio LIM , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...