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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 13(8): e1006984, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28827800

RESUMO

HSPB7 is a member of the small heat-shock protein (HSPB) family and is expressed in the cardiomyocytes from cardiogenesis onwards. A dramatic increase in HSPB7 is detected in the heart and blood plasma immediately after myocardial infarction. Additionally, several single-nucleotide polymorphisms of HSPB7 have been identified to be associated with heart failure caused by cardiomyopathy in human patients. Although a recent study has shown that HSPB7 is required for maintaining myofiber structure in skeletal muscle, its molecular and physiological functions in the heart remain unclear. In the present study, we generated a cardiac-specific inducible HSPB7 knockout mouse and demonstrated that the loss of HSPB7 in cardiomyocytes results in rapid heart failure and sudden death. The electrocardiogram showed cardiac arrhythmia with abnormal conduction in the HSPB7 mutant mice before death. In HSPB7 CKO cardiomyocytes, no significant defect was detected in the organization of contractile proteins in sarcomeres, but a severe structural disruption was observed in the intercalated discs. The expression of connexin 43, a gap-junction protein located at the intercalated discs, was downregulated in HSPB7 knockout cardiomyocytes. Mislocalization of desmoplakin, and N-cadherin, the intercalated disc proteins, was also observed in the HSPB7 CKO hearts. Furthermore, filamin C, the interaction protein of HSPB7, was upregulated and aggregated in HSPB7 mutant cardiomyocytes. In conclusion, our findings characterize HSPB7 as an intercalated disc protein and suggest it has an essential role in maintaining intercalated disc integrity and conduction function in the adult heart.


Assuntos
Cardiomiopatias/genética , Proteínas de Choque Térmico HSP27/genética , Insuficiência Cardíaca/genética , Miócitos Cardíacos/metabolismo , Animais , Síndrome de Brugada/genética , Síndrome de Brugada/patologia , Caderinas/genética , Doença do Sistema de Condução Cardíaco , Cardiomiopatias/fisiopatologia , Conexina 43/genética , Modelos Animais de Doenças , Eletrocardiografia , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Sarcômeros/metabolismo , Sarcômeros/patologia
2.
J Cell Sci ; 129(8): 1661-70, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929074

RESUMO

HSPB7 belongs to the small heat-shock protein (sHSP) family, and its expression is restricted to cardiac and skeletal muscles from embryonic stages to adulthood. Here, we found that skeletal-muscle-specific ablation of the HspB7 does not affect myogenesis during embryonic stages to postnatal day 1 (P1), but causes subsequent postnatal death owing to a respiration defect, with progressive myopathy phenotypes in the diaphragm. Deficiency of HSPB7 in the diaphragm muscle resulted in muscle fibrosis, sarcomere disarray and sarcolemma integrity loss. We identified dimerized filamin C (FLNC) as an interacting partner of HSPB7. Immunofluorescence studies demonstrated that the aggregation and mislocalization of FLNC occurred in the muscle of HspB7 mutant adult mice. Furthermore, the components of dystrophin glycoprotein complex, γ- and δ-sarcoglycan, but not dystrophin, were abnormally upregulated and mislocalized in HSPB7 mutant muscle. Collectively, our findings suggest that HSPB7 is essential for maintaining muscle integrity, which is achieved through its interaction with FLNC, in order to prevent the occurrence and progression of myopathy.


Assuntos
Diafragma/patologia , Filaminas/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Músculo Esquelético/fisiologia , Doenças Musculares/metabolismo , Animais , Células Cultivadas , Dimerização , Fibrose , Proteínas de Choque Térmico HSP27/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Desenvolvimento Muscular/genética , Doenças Musculares/genética , Ligação Proteica , Transporte Proteico/genética , Respiração/genética , Sarcoglicanas/metabolismo
3.
Cell Rep ; 42(11): 113416, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37967007

RESUMO

Differentiated cardiomyocytes (CMs) must undergo diverse morphological and functional changes during postnatal development. However, the mechanisms underlying initiation and coordination of these changes remain unclear. Here, we delineate an integrated, time-ordered transcriptional network that begins with expression of genes for cell-cell connections and leads to a sequence of structural, cell-cycle, functional, and metabolic transitions in mouse postnatal hearts. Depletion of histone H2B ubiquitin ligase RNF20 disrupts this gene network and impairs CM polarization. Subsequently, assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis confirmed that RNF20 contributes to chromatin accessibility in this context. As such, RNF20 is likely to facilitate binding of transcription factors at the promoters of genes involved in cell-cell connections and actin organization, which are crucial for CM polarization and functional integration. These results suggest that CM polarization is one of the earliest events during postnatal heart development and provide insights into how RNF20 regulates CM polarity and the postnatal gene program.


Assuntos
Miócitos Cardíacos , Ubiquitina-Proteína Ligases , Animais , Camundongos , Miócitos Cardíacos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Histonas/metabolismo , Cromatina , Epigênese Genética , Expressão Gênica
4.
FEBS Lett ; 583(17): 2765-71, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19619540

RESUMO

Ankyrin repeat domain 17 (Ankrd17) encodes an ubiquitously expressed protein with two clusters of ankyrin repeats. We have used gene targeting strategy to ablate the Ankrd17 gene in mouse. The Ankrd17-deficient mice died between embryonic day (E) 10.5 and E11.5 due to cardiovascular defects. Serious hemorrhages were detected and the vascular smooth muscle cells (vSMCs) surrounding the vessels were drastically reduced in the Ankrd17-deficient embryos, suggesting that the vascular maturation was not completed. Interestingly, vSMC differentiation marker genes were up-regulated in the mutant embryos. Our data have demonstrated the indispensability of Ankrd17 functioning for vascular maturation during early development. The Ankrd17-deficient mice also provide a new animal model for the analysis of the regulatory pathways of the differentiation of vSMC precursor cells.


Assuntos
Anquirinas/metabolismo , Vasos Sanguíneos/embriologia , Desenvolvimento Embrionário/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Proteínas/metabolismo , Animais , Anquirinas/genética , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Diferenciação Celular/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Hemorragia , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/citologia , Proteínas/genética , Proteínas de Ligação a RNA
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa