Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Brain Res ; 1741: 146878, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32407713

RESUMO

Accumulating evidence suggests that myocyte enhancer factor 2D (MEF2D) is a pro-survival factor for neurons. However, whether MEF2D is involved in protecting neurons from cerebral ischemia/reperfusion injury remains unknown. The current study was designed to investigate the exact role and mechanism of MEF2D in regulating oxygen-glucose deprivation/re-oxygenation (OGD/R)-induced neuronal injury, an in vitro model used to study cerebral ischemia/reperfusion injury. MEF2D expression was significantly induced in neurons in response to OGD/R injury. Functional analysis demonstrated that MEF2D upregulation significantly rescued the decreased viability of OGD/R-injured neurons and suppressed OGD/R-induced apoptosis and reactive oxygen species (ROS) production. By contrast, MEF2D knockdown increased the sensitivity of neurons to OGD/R-induced injury. Moreover, MEF2D overexpression increased the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and enhanced the activation of Nrf2 antioxidant signaling. However, Nrf2 knockdown partially blocked the MEF2D-mediated neuroprotective effect in OGD/R-exposed neurons. Overall, these results reveal that MEF2D overexpression attenuates OGD/R-induced injury by enhancing Nrf2-mediated antioxidant signaling. These findings suggest that MEF2D may serve as a neuroprotective target with a potential application for treatment of cerebral ischemia/reperfusion injury.


Assuntos
Hipóxia Celular/fisiologia , Glucose/deficiência , Fator 2 Relacionado a NF-E2/biossíntese , Neurônios/metabolismo , Regulação para Cima/fisiologia , Animais , Linhagem Celular , Fatores de Transcrição MEF2/biossíntese , Camundongos
3.
Circ Res ; 126(1): 6-24, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31730408

RESUMO

RATIONALE: Genome editing by CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 is evolving rapidly. Recently, second-generation CRISPR/Cas9 activation systems based on nuclease inactive dead (d)Cas9 fused to transcriptional transactivation domains were developed for directing specific guide (g)RNAs to regulatory regions of any gene of interest, to enhance transcription. The application of dCas9 to activate cardiomyocyte transcription in targeted genomic loci in vivo has not been demonstrated so far. OBJECTIVE: We aimed to develop a mouse model for cardiomyocyte-specific, CRISPR-mediated transcriptional modulation, and to demonstrate its versatility by targeting Mef2d and Klf15 loci (2 well-characterized genes implicated in cardiac hypertrophy and homeostasis) for enhanced transcription. METHODS AND RESULTS: A mouse model expressing dCas9 with the VPR transcriptional transactivation domains under the control of the Myh (myosin heavy chain) 6 promoter was generated. These mice innocuously expressed dCas9 exclusively in cardiomyocytes. For initial proof-of-concept, we selected Mef2d, which when overexpressed, led to hypertrophy and heart failure, and Klf15, which is lowly expressed in the neonatal heart. The most effective gRNAs were first identified in fibroblast (C3H/10T1/2) and myoblast (C2C12) cell lines. Using an improved triple gRNA expression system (TRISPR [triple gRNA expression construct]), up to 3 different gRNAs were transduced simultaneously to identify optimal conditions for transcriptional activation. For in vivo delivery of the validated gRNA combinations, we employed systemic administration via adeno-associated virus serotype 9. On gRNA delivery targeting Mef2d expression, we recapitulated the anticipated cardiac hypertrophy phenotype. Using gRNA targeting Klf15, we could enhance its transcription significantly, although Klf15 is physiologically silenced at that time point. We further confirmed specific and robust dCas9VPR on-target effects. CONCLUSIONS: The developed mouse model permits enhancement of gene expression by using endogenous regulatory genomic elements. Proof-of-concept in 2 independent genomic loci suggests versatile applications in controlling transcription in cardiomyocytes of the postnatal heart.


Assuntos
Sistemas CRISPR-Cas , Regulação da Expressão Gênica , Miocárdio/metabolismo , Ativação Transcricional , Animais , Linhagem Celular , Dependovirus/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica/genética , Genes Sintéticos , Vetores Genéticos/genética , Coração/crescimento & desenvolvimento , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/genética , Regiões Promotoras Genéticas , Domínios Proteicos , RNA Polimerase III/genética , RNA Guia de Cinetoplastídeos/genética
4.
PLoS Genet ; 13(4): e1006752, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28419090

RESUMO

The contribution of MEF2 TFs to the tumorigenic process is still mysterious. Here we clarify that MEF2 can support both pro-oncogenic or tumor suppressive activities depending on the interaction with co-activators or co-repressors partners. Through these interactions MEF2 supervise histone modifications associated with gene activation/repression, such as H3K4 methylation and H3K27 acetylation. Critical switches for the generation of a MEF2 repressive environment are class IIa HDACs. In leiomyosarcomas (LMS), this two-faced trait of MEF2 is relevant for tumor aggressiveness. Class IIa HDACs are overexpressed in 22% of LMS, where high levels of MEF2, HDAC4 and HDAC9 inversely correlate with overall survival. The knock out of HDAC9 suppresses the transformed phenotype of LMS cells, by restoring the transcriptional proficiency of some MEF2-target loci. HDAC9 coordinates also the demethylation of H3K4me3 at the promoters of MEF2-target genes. Moreover, we show that class IIa HDACs do not bind all the regulative elements bound by MEF2. Hence, in a cell MEF2-target genes actively transcribed and strongly repressed can coexist. However, these repressed MEF2-targets are poised in terms of chromatin signature. Overall our results candidate class IIa HDACs and HDAC9 in particular, as druggable targets for a therapeutic intervention in LMS.


Assuntos
Histona Desacetilases/biossíntese , Leiomiossarcoma/genética , Proteínas Repressoras/biossíntese , Ativação Transcricional/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Metilação de DNA/genética , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/genética , Humanos , Leiomiossarcoma/patologia , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Proteínas Repressoras/genética
5.
PLoS One ; 11(12): e0166791, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27907007

RESUMO

BACKGROUND: Left atrial enlargement in mitral regurgitation (MR) predicts a poor prognosis. The regulatory mechanisms of atrial myocyte hypertrophy of MR patients remain unknown. METHODS AND RESULTS: This study comprised 14 patients with MR, 7 patients with aortic valve disease (AVD), and 6 purchased samples from normal subjects (NC). We used microarrays, enrichment analysis and quantitative RT-PCR to study the gene expression profiles in the left atria. Microarray results showed that 112 genes were differentially up-regulated and 132 genes were differentially down-regulated in the left atria between MR patients and NC. Enrichment analysis of differentially expressed genes demonstrated that "NFAT in cardiac hypertrophy" pathway was not only one of the significant associated canonical pathways, but also the only one predicted with a non-zero score of 1.34 (i.e. activated) through Ingenuity Pathway Analysis molecule activity predictor. Ingenuity Pathway Analysis Global Molecular Network analysis exhibited that the highest score network also showed high association with cardiac related pathways and functions. Therefore, 5 NFAT associated genes (PPP3R1, PPP3CB, CAMK1, MEF2C, PLCE1) were studies for validation. The mRNA expressions of PPP3CB and MEF2C were significantly up-regulated, and CAMK1 and PPP3R1 were significantly down-regulated in MR patients compared to NC. Moreover, MR patients had significantly increased mRNA levels of PPP3CB, MEF2C and PLCE1 compared to AVD patients. The atrial myocyte size of MR patients significantly exceeded that of the AVD patients and NC. CONCLUSIONS: Differentially expressed genes in the "NFAT in cardiac hypertrophy" pathway may play a critical role in the atrial myocyte hypertrophy of MR patients.


Assuntos
Calcineurina/biossíntese , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/biossíntese , Cardiomegalia/genética , Cardiopatias Congênitas/genética , Doenças das Valvas Cardíacas/genética , Fosfoinositídeo Fosfolipase C/biossíntese , Idoso , Valva Aórtica/fisiopatologia , Doença da Válvula Aórtica Bicúspide , Calcineurina/genética , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/genética , Cardiomegalia/fisiopatologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Cardiopatias Congênitas/fisiopatologia , Doenças das Valvas Cardíacas/fisiopatologia , Humanos , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Insuficiência da Valva Mitral/genética , Insuficiência da Valva Mitral/fisiopatologia , Miócitos Cardíacos/patologia , Fosfoinositídeo Fosfolipase C/genética , RNA Mensageiro/biossíntese
6.
Oncotarget ; 7(21): 31053-66, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27105518

RESUMO

Cardiac fibrosis is an important pathological process of diabetic cardiomyopathy, the underlying mechanism remains elusive. This study sought to identify whether inhibition of Myocyte enhancer factor 2A (MEF2A) alleviates cardiac fibrosis by partially regulating Endothelial-to-mesenchymal transition (EndMT). We induced type 1 diabetes mellitus using the toxin streptozotocin (STZ) in mice and injected with lentivirus-mediated short-hairpin RNA (shRNA) in myocardium to inhibit MEF2A expression. Protein expression, histological and functional parameters were examined twenty-one weeks post-STZ injection. We found that Diabetes mellitus increased cardiac MEF2A expression, aggravated cardiac dysfunction and myocardial fibrosis through the accumulation of fibroblasts via EndMT. All of these features were abolished by MEF2A inhibition. MEF2A gene silencing by shRNA in cultured human umbilical vein endothelial cells (HUVECs) ameliorated high glucose-induced phenotypic transition and acquisition of mesenchymal markers through interaction with p38MAPK and Smad2. We conclude that inhibition of endothelial cell-derived MEF2A might be beneficial in the prevention of diabetes mellitus-induced cardiac fibrosis by partially inhibiting EndMT through interaction with p38MAPK and Smad2.


Assuntos
Diabetes Mellitus Experimental/terapia , Fibrose/terapia , Fatores de Transcrição MEF2/antagonistas & inibidores , Miocárdio/patologia , Animais , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Transição Epitelial-Mesenquimal , Fibrose/genética , Fibrose/metabolismo , Fibrose/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Transdução de Sinais , Transfecção
7.
Tumour Biol ; 36(12): 9885-93, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26164003

RESUMO

Hepatocellular carcinoma (HCC) is one of the most deadly human malignancy, and frequent invasion and metastasis is closely associated with its poor prognosis. However, the molecular mechanism underlying HCC invasion is still not completely elucidated. Pokemon is a well-established oncogene for HCC growth, but its contribution to HCC invasion has not been studied yet. In this paper, Pokemon was found to be overexpressed in MHCC-97H HCC cell line, which possesses higher invasiveness. Downregulation of Pokemon abolished the invasion of MHCC-97H HCC cell lines. Pokemon overexpression was able to enhance the invasion of MHCC-97L cells with lower invasiveness. MEF2D, an oncogene promoting the invasion of HCC cells, was further detected to be upregulated and downregulated when Pokemon was overexpressed and silenced, respectively. Online database analysis indicated that one Pokemon recognition site was located within the promoter of MEF2D. Chromatin co-precipitation, luciferase, and qPCR assays all proved that Pokemon can promote the expression of MEF2D in HCC cells. Restoration of MEF2D expression can prevent the impaired invasion of HCC cells with Pokemon silencing, while suppression of MEF2D abolished the effect of Pokemon overexpression on HCC invasion. More interestingly, MEF2D was also found to increase the transcription of Pokemon by binding myocyte enhancer factor 2 (MEF2) sites within its promoter region, implying an auto-regulatory circuit consisting of these two oncogenes that can promote HCC invasion. Our findings can contribute to the understanding of molecular mechanism underlying HCC invasion, and provided evidence that targeting this molecular loop may be a promising strategy for anti-invasion therapy.


Assuntos
Carcinoma Hepatocelular/genética , Proteínas de Ligação a DNA/biossíntese , Neoplasias Hepáticas/genética , Fatores de Transcrição/biossíntese , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Invasividade Neoplásica/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional
8.
Virchows Arch ; 467(3): 345-55, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26089142

RESUMO

Myocyte enhancer binding factor 2 B (MEF2B) is a member of the evolutionary conserved transcription family MEF2. MEF2B has been shown to directly control biological activity of the B cell lymphoma 6 (BCL6) gene in germinal center (GC) B cells. To validate MEF2B as an immunohistochemical marker, we studied a large consecutive series of hyperplastic lymphoid tissues (n = 38) and malignant lymphoproliferative conditions (n = 471), including all major categories of B and T cell neoplasms. In hyperplastic lymphoid tissues, MEF2B staining revealed intense and crisp nuclear expression confined to GC B cells. Unlike BCL6, MEF2B was not detected in follicular T cells. In addition, weak nuclear staining of plasma cells was noted. MEF2B staining labeled neoplastic cells of follicular lymphoma both in common and variant cases as well as in bone marrow biopsies with high sensitivity, while it was almost consistently negative in marginal zone lymphoma. Consistent MEF2B expression was found in Burkitt lymphoma and nodular lymphocyte predominant Hodgkin lymphoma as well as in the large majority of cases of mantle cell lymphoma and diffuse large cell B cell lymphoma. MEF2B protein expression showed a statistically significant association with that of BCL6 in cases of diffuse large B cell lymphoma, not otherwise specified. We conclude that MEF2B is a valuable marker of normal GC B cells, potentially useful in differential diagnosis of small B cell lymphomas.


Assuntos
Biomarcadores Tumorais/análise , Tecido Linfoide/metabolismo , Linfoma de Células B/diagnóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Imuno-Histoquímica , Linfoma de Células B/metabolismo , Fatores de Transcrição MEF2/análise , Fatores de Transcrição MEF2/biossíntese , Masculino , Pessoa de Meia-Idade , Análise Serial de Tecidos
9.
Exp Cell Res ; 336(2): 287-97, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26101156

RESUMO

Single-nucleotide polymorphisms associated with type 2 diabetes (T2D) have been identified in Jazf1, which is also involved in the oncogenesis of endometrial stromal tumors. To understand how Jazf1 variants confer a risk of tumorigenesis and T2D, we explored the functional roles of JAZF1 and searched for JAZF1 target genes in myogenic C2C12 cells. Consistent with an increase of Jazf1 transcripts during myoblast proliferation and their decrease during myogenic differentiation in regenerating skeletal muscle, JAZF1 overexpression promoted cell proliferation, whereas it retarded myogenic differentiation. Examination of myogenic genes revealed that JAZF1 overexpression transcriptionally repressed MEF2C and MRF4 and their downstream genes. AMP deaminase1 (AMPD1) was identified as a candidate for JAZF1 target by gene array analysis. However, promoter assays of Ampd1 demonstrated that mutation of the putative binding site for the TR4/JAZF1 complex did not alleviate the repressive effects of JAZF1 on promoter activity. Instead, JAZF1-mediated repression of Ampd1 occurred through the MEF2-binding site and E-box within the Ampd1 proximal regulatory elements. Consistently, MEF2C and MRF4 expression enhanced Ampd1 promoter activity. AMPD1 overexpression and JAZF1 downregulation impaired AMPK phosphorylation, while JAZF1 overexpression also reduced it. Collectively, these results suggest that aberrant JAZF1 expression contributes to the oncogenesis and T2D pathogenesis.


Assuntos
AMP Desaminase/genética , Proteínas de Transporte/genética , Transformação Celular Neoplásica/genética , Diabetes Mellitus Tipo 2/genética , Desenvolvimento Muscular/genética , Proteínas Nucleares/genética , AMP Desaminase/biossíntese , Animais , Sítios de Ligação/genética , Proteínas de Transporte/biossíntese , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/genética , Proteínas Correpressoras , Proteínas de Ligação a DNA , Diabetes Mellitus Tipo 2/patologia , Regulação da Expressão Gênica/genética , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Camundongos , Fibras Musculares Esqueléticas/citologia , Fatores de Regulação Miogênica/biossíntese , Fatores de Regulação Miogênica/genética , Proteínas Nucleares/biossíntese , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno , Transcrição Gênica/genética
10.
Cancer Res ; 75(11): 2387-99, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25858143

RESUMO

Hematopoietic stem cells (HSC) promptly adapt hematopoiesis to stress conditions, such as infection and cancer, replenishing bone marrow-derived circulating populations, while preserving the stem cell reservoir. SOCS2, a feedback inhibitor of JAK-STAT pathways, is expressed in most primitive HSC and is upregulated in response to STAT5-inducing cytokines. We demonstrate that Socs2 deficiency unleashes HSC proliferation in vitro, sustaining STAT5 phosphorylation in response to IL3, thrombopoietin, and GM-CSF. In vivo, SOCS2 deficiency leads to unrestricted myelopoietic response to 5-fluorouracil (5-FU) and, in turn, induces exhaustion of long-term HSC function along serial bone marrow transplantations. The emerging role of SOCS2 in HSC under stress conditions prompted the investigation of malignant hematopoiesis. High levels of SOCS2 characterize unfavorable subsets of acute myeloid and lymphoblastic leukemias, such as those with MLL and BCR/ABL abnormalities, and correlate with the enrichment of genes belonging to hematopoietic and leukemic stemness signatures. In this setting, SOCS2 and its correlated genes are part of regulatory networks fronted by IKZF1/Ikaros and MEF2C, two transcriptional regulators involved in normal and leukemic hematopoiesis that have never been linked to SOCS2. Accordingly, a comparison of murine wt and Socs2(-/-) HSC gene expression in response to 5-FU revealed a significant overlap with the molecular programs that correlate with SOCS2 expression in leukemias, particularly with the oncogenic pathways and with the IKZF1/Ikaros and MEF2C-predicted targets. Lentiviral gene transduction of murine hematopoietic precursors with Mef2c, but not with Ikzf1, induces Socs2 upregulation, unveiling a direct control exerted by Mef2c over Socs2 expression.


Assuntos
Células-Tronco Hematopoéticas/patologia , Leucemia/genética , Células-Tronco Neoplásicas/patologia , Proteínas Supressoras da Sinalização de Citocina/genética , Animais , Medula Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fluoruracila/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia/tratamento farmacológico , Leucemia/patologia , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/biossíntese , Células-Tronco Neoplásicas/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/biossíntese
11.
Mol Cell Biochem ; 404(1-2): 241-50, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25763715

RESUMO

An elevated level of homocysteine called hyperhomocysteinemia (HHcy) is associated with pathological cardiac remodeling. Hydrogen sulfide (H2S) acts as a cardioprotective gas; however, the mechanism by which H2S mitigates homocysteine-mediated pathological remodeling in cardiomyocytes is unclear. We hypothesized that H2S ameliorates HHcy-mediated hypertrophy by inducing cardioprotective miR-133a in cardiomyocytes. To test the hypothesis, HL1 cardiomyocytes were treated with (1) plain medium (control, CT), (2) 100 µM of homocysteine (Hcy), (3) Hcy with 30 µM of H2S (Hcy + H2S), and (4) H2S for 24 h. The levels of hypertrophy markers: c-fos, atrial natriuretic peptide (ANP), and beta-myosin heavy chain (ß-MHC), miR-133a, and its transcriptional inducer myosin enhancer factor-2C (MEF2C) were determined by Western blotting, RT-qPCR, and immunofluorescence. The activity of MEF2C was assessed by co-immunoprecipitation of MEF2C with histone deacetylase-1(HDAC1). Our results show that H2S ameliorates homocysteine-mediated up-regulation of c-fos, ANP, and ß-MHC, and down-regulation of MEF2C and miR-133a. HHcy induces the binding of MEF2C with HDAC1, whereas H2S releases MEF2C from MEF2C-HDAC1 complex causing activation of MEF2C. These findings elicit that HHcy induces cardiac hypertrophy by promoting MEF2C-HDAC1 complex formation that inactivates MEF2C causing suppression of anti-hypertrophy miR-133a in cardiomyocytes. H2S mitigates hypertrophy by inducing miR-133a through activation of MEF2C in HHcy cardiomyocytes. To our knowledge, this is a novel mechanism of H2S-mediated activation of MEF2C and induction of miR-133a and inhibition of hypertrophy in HHcy cardiomyocytes.


Assuntos
Sulfeto de Hidrogênio/administração & dosagem , Hiper-Homocisteinemia/genética , MicroRNAs/biossíntese , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Homocisteína/administração & dosagem , Humanos , Hiper-Homocisteinemia/patologia , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Camundongos , MicroRNAs/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Remodelação Ventricular/genética
14.
Mol Cell Biochem ; 400(1-2): 1-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25472877

RESUMO

Oleanolic acid (OA) is a natural compound from plants with anti-tumor activities. However, the mechanism of the inhibitory effect of OA on cell cycle progression has not been completely explored. We employed several lung carcinoma cell lines to investigate the cell cycle-related molecular pathway affected by OA. The data revealed that OA suppressed the proliferation of lung cancer cells in both dose- and time-dependent manners, along with an increase in miR-122 abundance. The suppression of miR-122 abolished the effect of OA on lung cancer cells. CCNG1 and MEF2D, two putative miR-122 targets, were found to be downregulated by OA treatment. Restoring their expression counteracted the effect of OA on lung carcinoma cells. OA was further shown to induce the expression of miR-122-regulating transcriptional factors in lung cancer cells. Collectively, OA induced cell cycle arrest in lung cancer cells through miR-122/Cyclin G1/MEF2D pathway. This finding may contribute to the understanding of the molecular mechanism of OA's anti-tumor activity.


Assuntos
Ciclina G1/biossíntese , Neoplasias Pulmonares/tratamento farmacológico , MicroRNAs/biossíntese , Ácido Oleanólico/administração & dosagem , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina G1/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , MicroRNAs/genética
15.
Circ Res ; 116(2): 237-44, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25416133

RESUMO

RATIONALE: Generation of induced cardiac myocytes (iCMs) directly from fibroblasts offers great opportunities for cardiac disease modeling and cardiac regeneration. A major challenge of iCM generation is the low conversion rate of fibroblasts to fully reprogrammed iCMs, which could in part be attributed to unbalanced expression of reprogramming factors Gata4 (G), Mef2c (M), and Tbx5 (T) using the current gene delivery approach. OBJECTIVE: We aimed to establish a system to express distinct ratios of G, M, T proteins in fibroblasts and determine the effect of G, M, T stoichiometry on iCM reprogramming. METHODS AND RESULTS: We took advantage of the inherent feature of the polycistronic system and generated all possible combinations of G, M, T with identical 2A sequences in a single transgene. We demonstrated that each splicing order of G, M, T gave rise to distinct G, M, T protein expression levels. Combinations that resulted in higher protein level of Mef2c with lower levels of Gata4 and Tbx5 significantly enhanced reprogramming efficiency compared with separate G, M, T transduction. Importantly, after further optimization, the MGT vector resulted in more than 10-fold increase in the number of mature beating iCM loci. Molecular characterization revealed that more optimal G, M, T stoichiometry correlated with higher expression of mature cardiac myocyte markers. CONCLUSIONS: Our results demonstrate that stoichiometry of G, M, T protein expression influences the efficiency and quality of iCM reprogramming. The established optimal G, M, T expression condition will provide a valuable platform for future iCM studies.


Assuntos
Reprogramação Celular/fisiologia , Fator de Transcrição GATA4/biossíntese , Miócitos Cardíacos/fisiologia , Proteínas com Domínio T/biossíntese , Animais , Células Cultivadas , Fator de Transcrição GATA4/genética , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Camundongos , Camundongos Transgênicos , Proteínas com Domínio T/genética
16.
Circ Res ; 116(1): 56-69, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25287062

RESUMO

RATIONALE: Right ventricular (RV) failure is a major cause of morbidity and mortality in pulmonary hypertension, but its mechanism remains unknown. Myocyte enhancer factor 2 (Mef2) has been implicated in RV development, regulating metabolic, contractile, and angiogenic genes. Moreover, Mef2 regulates microRNAs that have emerged as important determinants of cardiac development and disease, but for which the role in RV is still unclear. OBJECTIVE: We hypothesized a critical role of a Mef2-microRNAs axis in RV failure. METHODS AND RESULTS: In a rat pulmonary hypertension model (monocrotaline), we studied RV free wall tissues from rats with normal, compensated, and decompensated RV hypertrophy, carefully defined based on clinically relevant parameters, including RV systolic and end-diastolic pressures, cardiac output, RV size, and morbidity. Mef2c expression was sharply increased in compensating phase of RVH tissues but was lost in decompensation phase of RVH. An unbiased screening of microRNAs in our model resulted to a short microRNA signature of decompensated RV failure, which included the myocardium-specific miR-208, which was progressively downregulated as RV failure progressed, in contrast to what is described in left ventricular failure. With mechanistic in vitro experiments using neonatal and adult RV cardiomyocytes, we showed that miR-208 inhibition, as well as tumor necrosis factor-α, activates the complex mediator of transcription 13/nuclear receptor corepressor 1 axis, which in turn promotes Mef2 inhibition, closing a self-limiting feedback loop, driving the transition from compensating phase of RVH toward decompensation phase of RVH. In our model, serum tumor necrosis factor-α levels progressively increased with time while serum miR-208 levels decreased, mirroring its levels in RV myocardium. CONCLUSIONS: We describe an RV-specific mechanism for heart failure, which could potentially lead to new biomarkers and therapeutic targets.


Assuntos
Insuficiência Cardíaca/metabolismo , Hipertensão Pulmonar/metabolismo , Fatores de Transcrição MEF2/biossíntese , MicroRNAs/biossíntese , Função Ventricular Direita/fisiologia , Animais , Células Cultivadas , Insuficiência Cardíaca/patologia , Hipertensão Pulmonar/patologia , Masculino , Miócitos Cardíacos/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
17.
J Biol Chem ; 290(2): 1256-68, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25416778

RESUMO

Skeletal muscle differentiation requires precisely coordinated transcriptional regulation of diverse gene programs that ultimately give rise to the specialized properties of this cell type. In Drosophila, this process is controlled, in part, by MEF2, the sole member of an evolutionarily conserved transcription factor family. By contrast, vertebrate MEF2 is encoded by four distinct genes, Mef2a, -b, -c, and -d, making it far more challenging to link this transcription factor to the regulation of specific muscle gene programs. Here, we have taken the first step in molecularly dissecting vertebrate MEF2 transcriptional function in skeletal muscle differentiation by depleting individual MEF2 proteins in myoblasts. Whereas MEF2A is absolutely required for proper myoblast differentiation, MEF2B, -C, and -D were found to be dispensable for this process. Furthermore, despite the extensive redundancy, we show that mammalian MEF2 proteins regulate a significant subset of nonoverlapping gene programs. These results suggest that individual MEF2 family members are able to recognize specific targets among the entire cohort of MEF2-regulated genes in the muscle genome. These findings provide opportunities to modulate the activity of MEF2 isoforms and their respective gene programs in skeletal muscle homeostasis and disease.


Assuntos
Diferenciação Celular/genética , Evolução Molecular , Fatores de Transcrição MEF2/biossíntese , Músculo Esquelético/crescimento & desenvolvimento , Isoformas de Proteínas/biossíntese , Animais , Células COS , Chlorocebus aethiops , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição MEF2/antagonistas & inibidores , Fatores de Transcrição MEF2/genética , Mamíferos/genética , Mamíferos/crescimento & desenvolvimento , Camundongos , Desenvolvimento Muscular/genética , Mioblastos/citologia , Mioblastos/metabolismo , Isoformas de Proteínas/genética
18.
Biomed Res Int ; 2015: 748470, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26788505

RESUMO

Our previous study reported that muscle cell enhancement factor 2C (MEF2C) was fully activated after inhibition of the phosphorylation activity of integrin-linked kinase (ILK) in the skeletal muscle cells of goats. It enhanced the binding of promoter or enhancer of transcription factor related to proliferation of muscle cells and then regulated the expression of these genes. In the present investigation, we explored whether ILK activation depended on PI3K to regulate the phosphorylation and transcriptional activity of MEF2C during C2C12 cell proliferation. We inhibited PI3K activity in C2C12 with LY294002 and then found that ILK phosphorylation levels and MEF2C phosphorylation were decreased and that MCK mRNA expression was suppressed significantly. After inhibiting ILK phosphorylation activity with Cpd22 and ILK-shRNA, we found MEF2C phosphorylation activity and MCK mRNA expression were increased extremely significantly. In the presence of Cpd22, PI3K activity inhibition increased MEF2C phosphorylation and MCK mRNA expression indistinctively. We conclude that ILK negatively and independently of PI3K regulated MEF2C phosphorylation activity and MCK mRNA expression in C2C12 cells. The results provide new ideas for the study of classical signaling pathway of PI3K-ILK-related proteins and transcription factors.


Assuntos
Creatina Quinase Forma MM/biossíntese , Regulação da Expressão Gênica/genética , Fatores de Transcrição MEF2/genética , Proteínas Serina-Treonina Quinases/biossíntese , Animais , Linhagem Celular , Proliferação de Células/genética , Creatina Quinase Forma MM/genética , Fatores de Transcrição MEF2/biossíntese , Camundongos , Células Musculares/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/biossíntese , Transdução de Sinais , Ativação Transcricional/genética
19.
J Bone Miner Res ; 30(6): 1044-52, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25529628

RESUMO

Growth plate chondrocytes go through multiple differentiation steps and eventually become hypertrophic chondrocytes. The parathyroid hormone (PTH)-related peptide (PTHrP) signaling pathway plays a central role in regulation of hypertrophic differentiation, at least in part, through enhancing activity of histone deacetylase 4 (HDAC4), a negative regulator of MEF2 transcription factors that drive hypertrophy. We have previously shown that loss of the chondrocyte-specific microRNA (miRNA), miR-140, alters chondrocyte differentiation including mild acceleration of hypertrophic differentiation. Here, we provide evidence that miR-140 interacts with the PTHrP-HDAC4 pathway to control chondrocyte differentiation. Heterozygosity of PTHrP or HDAC4 substantially impaired animal growth in miR-140 deficiency, whereas these mutations had no effect in the presence of miR-140. miR-140-deficient chondrocytes showed increased MEF2C expression with normal levels of total and phosphorylated HDAC4, indicating that the miR-140 pathway merges with the PTHrP-HDAC4 pathway at the level of MEF2C. miR-140 negatively regulated p38 mitogen-activated protein kinase (MAPK) signaling, and inhibition of p38 MAPK signaling reduced MEF2C expression. These results demonstrate that miR-140 ensures the robustness of the PTHrP/HDAC4 regulatory system by suppressing MEF2C-inducing stimuli. © 2014 American Society for Bone and Mineral Research © 2015 American Society for Bone and Mineral Research.


Assuntos
Condrócitos/metabolismo , Histona Desacetilases/metabolismo , MicroRNAs/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Transdução de Sinais , Animais , Condrócitos/patologia , Histona Desacetilases/genética , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/patologia , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética , Proteína Relacionada ao Hormônio Paratireóideo/genética
20.
Mol Biol Cell ; 25(23): 3765-78, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25208565

RESUMO

Recent work has shown that Staufen1 plays key roles in skeletal muscle, yet little is known about its pattern of expression during embryonic and postnatal development. Here we first show that Staufen1 levels are abundant in mouse embryonic muscles and that its expression decreases thereafter, reaching low levels in mature muscles. A similar pattern of expression is seen as cultured myoblasts differentiate into myotubes. Muscle degeneration/regeneration experiments revealed that Staufen1 increases after cardiotoxin injection before returning to the low levels seen in mature muscles. We next prevented the decrease in Staufen1 during differentiation by generating stable C2C12 muscle cell lines overexpressing Staufen1. Cells overexpressing Staufen1 differentiated poorly, as evidenced by reductions in the differentiation and fusion indices and decreases in MyoD, myogenin, MEF2A, and MEF2C, independently of Staufen-mediated mRNA decay. However, levels of c-myc, a factor known to inhibit differentiation, were increased in C2C12 cells overexpressing Staufen1 through enhanced translation. By contrast, the knockdown of Staufen1 decreased c-myc levels in myoblasts. Collectively our results show that Staufen1 is highly expressed during early stages of differentiation/development and that it can impair differentiation by regulating c-myc, thereby highlighting the multifunctional role of Staufen1 in skeletal muscle cells.


Assuntos
Diferenciação Celular/genética , Desenvolvimento Muscular , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas de Ligação a RNA/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição MEF2/biossíntese , Camundongos , Músculos/metabolismo , Proteína MyoD/biossíntese , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas de Ligação a RNA/biossíntese , Regeneração/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA