Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Exp Cell Res ; 420(2): 113356, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36122768

RESUMO

Actin has important functions in both cytoplasm and nucleus of the cell, with active nuclear transport mechanisms maintaining the cellular actin balance. Nuclear actin levels are subject to regulation during many cellular processes from cell differentiation to cancer. Here we show that nuclear actin levels increase upon differentiation of PC6.3 cells towards neuron-like cells. Photobleaching experiments demonstrate that this increase is due to decreased nuclear export of actin during cell differentiation. Increased nuclear actin levels lead to decreased nuclear localization of MRTF-A, a well-established transcription cofactor of SRF. In line with MRTF-A localization, transcriptomics analysis reveals that MRTF/SRF target gene expression is first transiently activated, but then substantially downregulated during PC6.3 cell differentiation. This study therefore describes a novel cellular context, where regulation of nuclear actin is utilized to tune MRTF/SRF target gene expression during cell differentiation.


Assuntos
Actinas , Transativadores , Actinas/genética , Actinas/metabolismo , Diferenciação Celular/genética , Expressão Gênica , Regulação da Expressão Gênica , Extratos Vegetais , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transativadores/genética , Transativadores/metabolismo
2.
Vascul Pharmacol ; 146: 107091, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35896140

RESUMO

Vascular smooth muscle cells (SMC) possess a unique cytoplasticity, regulated by transcriptional, translational and phenotypic transformation in response to a diverse range of extrinsic and intrinsic pathogenic factors. The mature, differentiated SMC phenotype is physiologically typified transcriptionally by expression of genes encoding "contractile" proteins, such as SMα-actin (ACTA2), SM-MHC (myosin-11) and SM22α (transgelin). When exposed to various pathological conditions (e.g., pro-atherogenic risk factors, hypertension), SMC undergo phenotypic modulation, a bioprocess enabling SMC to de-differentiate in immature stages or trans-differentiate into other cell phenotypes. As recent studies suggest, the process of SMC phenotypic transformation involves five distinct states characterized by different patterns of cell growth, differentiation, migration, matrix protein expression and declined contractility. These changes are mediated via the action of several transcriptional regulators, including myocardin and serum response factor. Conversely, other factors, including Kruppel-like factor 4 and nuclear factor-κB, can inhibit SMC differentiation and growth arrest, while factors such as yin yang-1, can promote SMC differentiation whilst inhibiting proliferation. This article reviews recent advances in our understanding of regulatory mechanisms governing SMC phenotypic modulation. We propose the concept that transcription factors mediating this switching are important biomarkers and potential pharmacological targets for therapeutic intervention in cardiovascular disease.


Assuntos
Músculo Liso Vascular , Fator de Resposta Sérica , Actinas/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Fenótipo , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo
3.
Circulation ; 137(21): 2256-2273, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29217642

RESUMO

BACKGROUND: Myocardial metabolic impairment is a major feature in chronic heart failure. As the major coenzyme in fuel oxidation and oxidative phosphorylation and a substrate for enzymes signaling energy stress and oxidative stress response, nicotinamide adenine dinucleotide (NAD+) is emerging as a metabolic target in a number of diseases including heart failure. Little is known on the mechanisms regulating homeostasis of NAD+ in the failing heart. METHODS: To explore possible alterations of NAD+ homeostasis in the failing heart, we quantified the expression of NAD+ biosynthetic enzymes in the human failing heart and in the heart of a mouse model of dilated cardiomyopathy (DCM) triggered by Serum Response Factor transcription factor depletion in the heart (SRFHKO) or of cardiac hypertrophy triggered by transverse aorta constriction. We studied the impact of NAD+ precursor supplementation on cardiac function in both mouse models. RESULTS: We observed a 30% loss in levels of NAD+ in the murine failing heart of both DCM and transverse aorta constriction mice that was accompanied by a decrease in expression of the nicotinamide phosphoribosyltransferase enzyme that recycles the nicotinamide precursor, whereas the nicotinamide riboside kinase 2 (NMRK2) that phosphorylates the nicotinamide riboside precursor is increased, to a higher level in the DCM (40-fold) than in transverse aorta constriction (4-fold). This shift was also observed in human failing heart biopsies in comparison with nonfailing controls. We show that the Nmrk2 gene is an AMP-activated protein kinase and peroxisome proliferator-activated receptor α responsive gene that is activated by energy stress and NAD+ depletion in isolated rat cardiomyocytes. Nicotinamide riboside efficiently rescues NAD+ synthesis in response to FK866-mediated inhibition of nicotinamide phosphoribosyltransferase and stimulates glycolysis in cardiomyocytes. Accordingly, we show that nicotinamide riboside supplementation in food attenuates the development of heart failure in mice, more robustly in DCM, and partially after transverse aorta constriction, by stabilizing myocardial NAD+ levels in the failing heart. Nicotinamide riboside treatment also robustly increases the myocardial levels of 3 metabolites, nicotinic acid adenine dinucleotide, methylnicotinamide, and N1-methyl-4-pyridone-5-carboxamide, that can be used as validation biomarkers for the treatment. CONCLUSIONS: The data show that nicotinamide riboside, the most energy-efficient among NAD precursors, could be useful for treatment of heart failure, notably in the context of DCM, a disease with few therapeutic options.


Assuntos
Cardiomiopatia Dilatada/tratamento farmacológico , Niacinamida/análogos & derivados , Proteínas Quinases Ativadas por AMP/metabolismo , Acrilamidas/uso terapêutico , Animais , Ácido Cítrico/metabolismo , Citocinas/genética , Citocinas/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Insuficiência Cardíaca/prevenção & controle , Metaboloma/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , NAD/metabolismo , Niacinamida/uso terapêutico , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , PPAR alfa/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Piperidinas/uso terapêutico , Compostos de Piridínio , Ratos , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética
4.
PLoS One ; 9(11): e112109, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25427253

RESUMO

We have generated a novel monoclonal antibody targeting human FGFR1c (R1c mAb) that caused profound body weight and body fat loss in diet-induced obese mice due to decreased food intake (with energy expenditure unaltered), in turn improving glucose control. R1c mAb also caused weight loss in leptin-deficient ob/ob mice, leptin receptor-mutant db/db mice, and in mice lacking either the melanocortin 4 receptor or the melanin-concentrating hormone receptor 1. In addition, R1c mAb did not change hypothalamic mRNA expression levels of Agrp, Cart, Pomc, Npy, Crh, Mch, or Orexin, suggesting that R1c mAb could cause food intake inhibition and body weight loss via other mechanisms in the brain. Interestingly, peripherally administered R1c mAb accumulated in the median eminence, adjacent arcuate nucleus and in the circumventricular organs where it activated the early response gene c-Fos. As a plausible mechanism and coinciding with the initiation of food intake suppression, R1c mAb induced hypothalamic expression levels of the cytokines Monocyte chemoattractant protein 1 and 3 and ERK1/2 and p70 S6 kinase 1 activation.


Assuntos
Anticorpos Monoclonais/farmacologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Órgãos Circunventriculares/efeitos dos fármacos , Intolerância à Glucose/tratamento farmacológico , Hipotálamo/efeitos dos fármacos , Obesidade/tratamento farmacológico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiopatologia , Quimiocina CCL2/agonistas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL7/agonistas , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Órgãos Circunventriculares/metabolismo , Órgãos Circunventriculares/fisiopatologia , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético , Feminino , Regulação da Expressão Gênica , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/fisiopatologia , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Leptina/deficiência , Leptina/genética , Camundongos , Camundongos Knockout , Camundongos Obesos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Obesidade/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 4 de Melanocortina/deficiência , Receptor Tipo 4 de Melanocortina/genética , Receptores de Somatostatina/deficiência , Receptores de Somatostatina/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fator de Resposta Sérica/agonistas , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transdução de Sinais
5.
J Physiol ; 591(15): 3749-63, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23753523

RESUMO

The striated muscle activator of Rho signalling (STARS) pathway is suggested to provide a link between external stress responses and transcriptional regulation in muscle. However, the sensitivity of STARS signalling to different mechanical stresses has not been investigated. In a comparative study, we examined the regulation of the STARS signalling pathway in response to unilateral resistance exercise performed as either eccentric (ECC) or concentric (CONC) contractions as well as prolonged training; with and without whey protein supplementation. Skeletal muscle STARS, myocardian-related transcription factor-A (MRTF-A) and serum response factor (SRF) mRNA and protein, as well as muscle cross-sectional area and maximal voluntary contraction, were measured. A single-bout of exercise produced increases in STARS and SRF mRNA and decreases in MRTF-A mRNA with both ECC and CONC exercise, but with an enhanced response occurring following ECC exercise. A 31% increase in STARS protein was observed exclusively after CONC exercise (P < 0.001), while pSRF protein levels increased similarly by 48% with both CONC and ECC exercise (P < 0.001). Prolonged ECC and CONC training equally stimulated muscle hypertrophy and produced increases in MRTF-A protein of 125% and 99%, respectively (P < 0.001). No changes occurred for total SRF protein. There was no effect of whey protein supplementation. These results show that resistance exercise provides an acute stimulation of the STARS pathway that is contraction mode dependent. The responses to acute exercise were more pronounced than responses to accumulated training, suggesting that STARS signalling is primarily involved in the initial phase of exercise-induced muscle adaptations.


Assuntos
Suplementos Nutricionais , Proteínas dos Microfilamentos/metabolismo , Proteínas do Leite/farmacologia , Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Treinamento Resistido , Fatores de Transcrição/metabolismo , Adulto , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Masculino , Proteínas dos Microfilamentos/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transdução de Sinais , Transativadores , Fatores de Transcrição/genética , Proteínas do Soro do Leite , Adulto Jovem
6.
J Appl Physiol (1985) ; 107(4): 1204-12, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628726

RESUMO

Skeletal muscles, especially weight-bearing muscles, are very sensitive to changes in loading state. The aim of this paper was to characterize the dynamic changes in the unloaded soleus muscle in vivo following a short bout of hindlimb suspension (HS), testing the hypothesis that transcriptional events respond early to the atrophic stimulus. In fact, we observed that after only 1 day of HS, primary transcript levels of skeletal alpha-actin and type I myosin heavy chain (MHC) genes were significantly reduced by more than 50% compared with ground control levels. The degree of the decline for the mRNA expression of actin and type I MHC lagged behind that of the pre-mRNA levels after 1 day of HS, but by 2 and 7 days of HS, large decreases were observed. Although the faster MHC isoforms, IIx and IIb, began to be expressed in soleus after 1 day of HS, a relatively significant shift in mRNA expression from the slow MHC isoform type I toward these fast MHC isoforms did not emerge until 7 days of HS. One day of HS was sufficient to show significant decreases in mRNA levels of putative signaling factors serum response factor (SRF), suppressor of cytokine signaling-3 (SOCS3), and striated muscle activator of Rho signaling (STARS), although transcription factors yin-yang-1 (YY1) and transcriptional enhancing factor-1 (TEF-1) were not significantly affected by HS. The protein levels of actin and type I MHC were significantly decreased after 2 days of HS, and SRF protein was significantly decreased after 7 days HS. Our results show that after only 1 day of unloading, pre-mRNA and mRNA expression of muscle proteins and muscle-specific signaling factors are significantly reduced, suggesting that the downregulation of the synthesis side of the protein balance equation that occurs in atrophying muscle is initiated rapidly.


Assuntos
Actinas/genética , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Cadeias Pesadas de Miosina/genética , Transcrição Gênica , Actinas/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Elevação dos Membros Posteriores , Músculo Esquelético/patologia , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Miofibrilas/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Tamanho do Órgão , Isoformas de Proteínas , Precursores de RNA/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Resposta Sérica/genética , Transdução de Sinais/genética , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Fatores de Tempo , Fator de Transcrição YY1/genética
7.
Genes Dev ; 18(21): 2627-38, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15520282

RESUMO

The Ezh2 protein endows the Polycomb PRC2 and PRC3 complexes with histone lysine methyltransferase (HKMT) activity that is associated with transcriptional repression. We report that Ezh2 expression was developmentally regulated in the myotome compartment of mouse somites and that its down-regulation coincided with activation of muscle gene expression and differentiation of satellite-cell-derived myoblasts. Increased Ezh2 expression inhibited muscle differentiation, and this property was conferred by its SET domain, required for the HKMT activity. In undifferentiated myoblasts, endogenous Ezh2 was associated with the transcriptional regulator YY1. Both Ezh2 and YY1 were detected, with the deacetylase HDAC1, at genomic regions of silent muscle-specific genes. Their presence correlated with methylation of K27 of histone H3. YY1 was required for Ezh2 binding because RNA interference of YY1 abrogated chromatin recruitment of Ezh2 and prevented H3-K27 methylation. Upon gene activation, Ezh2, HDAC1, and YY1 dissociated from muscle loci, H3-K27 became hypomethylated and MyoD and SRF were recruited to the chromatin. These findings suggest the existence of a two-step activation mechanism whereby removal of H3-K27 methylation, conferred by an active Ezh2-containing protein complex, followed by recruitment of positive transcriptional regulators at discrete genomic loci are required to promote muscle gene expression and cell differentiation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Proteínas/metabolismo , Animais , Diferenciação Celular/fisiologia , Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste , Fatores de Ligação de DNA Eritroide Específicos , Extremidades/embriologia , Histona Desacetilase 1 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histona-Lisina N-Metiltransferase , Lisina/metabolismo , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos , Proteína MyoD/genética , Proteína MyoD/metabolismo , Complexo Repressor Polycomb 2 , Estrutura Terciária de Proteína , Proteínas/genética , RNA Interferente Pequeno , Sequências Reguladoras de Ácido Nucleico , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Fator de Transcrição YY1
8.
Gastroenterology ; 126(7): 1809-18, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15188176

RESUMO

BACKGROUND & AIMS: Serum response factor (SRF) regulates transcription of immediate early genes and muscle genes. In this study, we examined the role of SRF in gastric ulcer healing and the mechanisms involved. METHODS: Gastric ulcers were induced in rats by serosal application of acetic acid. Gastric specimens were obtained sequentially after ulcer induction for analyses of SRF messenger RNA (mRNA), protein expression, and for immunohistochemistry. We examined the role of SRF in ulcer healing by local injection of an SRF expression plasmid into ulcers (gene therapy). To elucidate the cellular mechanisms of the action of SRF, we examined the effect of SRF overexpression on actin dynamics, cell migration, and proliferation in rat gastric epithelial cell (RGM1) and smooth muscle cell (A7R5). To determine the clinical relevance, we examined SRF expression in human gastric ulcer specimens. RESULTS: Gastric ulceration activated SRF expression in epithelial cells lining regenerating glands and in myofibroblasts and smooth muscle cells of granulation tissue. SRF up-regulation in human gastric ulcers was similar to that found in rat gastric ulcers. Gene therapy with SRF significantly accelerated experimental gastric ulcer healing and promoted re-epithelialization and muscle restoration. Overexpression of SRF in RGM1 and A7R5 cells accelerates migration and proliferation of these cells by promoting actin polymerization and activation of immediately early genes. CONCLUSIONS: Activation of SRF is an important component of ulcer healing. SRF promotes migration and proliferation of gastric epithelial and smooth muscle cells, which are essential for re-epithelialization and restoration of muscular structures.


Assuntos
Células Epiteliais/fisiologia , Miócitos de Músculo Liso/fisiologia , Fator de Resposta Sérica/genética , Úlcera Gástrica/fisiopatologia , Cicatrização/fisiologia , Animais , Divisão Celular , Movimento Celular , DNA Complementar/farmacologia , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Células Epiteliais/citologia , Expressão Gênica , Terapia Genética , Proteínas Imediatamente Precoces/genética , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Miócitos de Músculo Liso/citologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Fator de Resposta Sérica/metabolismo , Úlcera Gástrica/patologia , Úlcera Gástrica/terapia , Fibras de Estresse/metabolismo , Fatores de Transcrição/genética
9.
Biochem Biophys Res Commun ; 306(1): 32-8, 2003 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12788062

RESUMO

Serum stimulation leads to activation of the serum response factor (SRF)-mediated transcription of immediate-early genes such as c-fos via various signal transduction pathways. We have previously reported that promyelocytic leukemia protein (PML) is involved in the transcriptional regulation by SRF. PML is one of the well-known substrates for modification by small ubiquitin-related modifier-1 (SUMO-1) and several SUMO-1-modified proteins associate with PML. Here, we report that SRF is modified by SUMO-1 chiefly at lysine(147) within the DNA-binding domain. Substitution of this target lysine for alanine did not affect the translocation of SRF to PML-nuclear bodies. The SRF mutant augmented the transcriptional activity under Rho A-stimulated condition but not under serum-starved condition, suggesting that activated SRF is suppressed by its sumoylation. These data support the transcriptional role of SUMO-1 conjugating system in cellular serum response.


Assuntos
Proteínas Nucleares , Proteína SUMO-1/metabolismo , Fator de Resposta Sérica/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , DNA Complementar/genética , Células HeLa , Humanos , Técnicas In Vitro , Lisina/química , Proteínas de Neoplasias/metabolismo , Proteína da Leucemia Promielocítica , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína SUMO-1/genética , Fator de Resposta Sérica/química , Fator de Resposta Sérica/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA