Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-32322239

RESUMO

The neuromodulator oxytocin, since its first synthesis by du Vigneaud in 1953, has mainly been associated with beneficial physiological effects, as well as positive social and emotional behaviors. This overall positive picture of oxytocin as the "love-, cuddle-, or bonding-hormone" has repeatedly been challenged since then. Oxytocin-induced effects that would be perceived as negative by the individual, such as increased anxiety or potentiation of stress-induced ACTH release, as well as the regulation of negative approach-related emotions, such as envy and schadenfreude (gloating) have been described. The general consent is that oxytocin, instead of acting unidirectional, induces changes in the salience network to shift the emphasis of emotional contexts, and therefore can, e.g., produce both anxiolytic as well as anxiogenic behavioral outcomes. However, the underlying mechanisms leading to alterations in the salience network are still unclear. With the aim to understand the manifold effects of oxytocin on a cellular/molecular level, a set of oxytocin receptor-coupled signaling cascades and downstream effectors regulating transcription and translation has been identified. Those oxytocin-driven effectors, such as MEF2 and CREB, are known modulators of the neuronal and glial cytoarchitecture. We hypothesize that, by determining cellular morphology and connectivity, MEF2 is one of the key factors that might contribute to the diverse behavioral effects of oxytocin.


Assuntos
Ansiolíticos/farmacologia , Ansiedade/induzido quimicamente , Fatores de Transcrição MEF2/fisiologia , Ocitocina/efeitos adversos , Ocitocina/farmacologia , Animais , Comportamento/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Emoções/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
2.
J Neurosci ; 39(40): 7958-7967, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31420455

RESUMO

The transcription factor, myocyte enhancer factor-2 (MEF2), is required for normal circadian behavior in Drosophila; however, its role in the mammalian circadian system has not been established. Of the four mammalian Mef2 genes, Mef2d is highly expressed in the suprachiasmatic nucleus (SCN) of the hypothalamus, a region critical for coordinating peripheral circadian clocks. Using both conventional and brain-specific Mef2d KO (Mef2d-/-) mouse lines, we demonstrate that MEF2D is essential for maintaining the length of the circadian free-running period of locomotor activity and normal sleep patterns in male mice. Crossing Mef2d-/- with Per2::luc reporter mice, we show that these behavioral changes are achieved without altering the endogenous period of the master circadian oscillator in the SCN. Together, our data suggest that alterations in behavior in Mef2d-/- mice may be the result of an effect on SCN output, rather than an effect on timekeeping within the SCN itself. These findings add to the growing body of evidence that MEF2 proteins play important roles in the brain.SIGNIFICANCE STATEMENT These studies are the first to show a role for MEF2 proteins in the brain outside of the hippocampus, and our findings suggest that these proteins may play diverse roles in the CNS. It is important to continue to build on our understanding of the roles of proteins acting in the SCN because SCN dysfunction underlies jet lag in humans and influences the response to shift work schedules, which are now known as risk factors for the development of cancer. Our work on MEF2D could be the basis for opening new lines of research in the development and regulation of circadian rhythms.


Assuntos
Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Sono/genética , Sono/fisiologia , Animais , Comportamento Animal , Proteínas CLOCK/biossíntese , Proteínas CLOCK/genética , Luz , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , RNA/biossíntese , RNA/genética , Transtornos do Sono-Vigília/genética , Transtornos do Sono-Vigília/psicologia , Núcleo Supraquiasmático/fisiologia
3.
BMC Mol Biol ; 20(1): 8, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30885136

RESUMO

BACKGROUND: Myocyte enhancer factor 2A (MEF2A) plays an important role in cell proliferation, differentiation and survival. Functional deletion or mutation in MEF2A predisposes individuals to cardiovascular disease mainly caused by vascular endothelial dysfunction. However, the effect of the inhibition of MEF2A expression on human coronary artery endothelial cells (HCAECs) is unclear. In this study, expression of MEF2A was inhibited by specific small interference RNA (siRNA), and changes in mRNA profiles in response to MEF2A knockdown were analyzed using an Agilent human mRNA array. RESULTS: Silencing of MEF2A in HCAECs accelerated cell senescence and suppressed cell proliferation. Microarray analysis identified 962 differentially expressed genes (DEGs) between the MEF2A knockdown group and the negative control group. Annotation clustering analysis showed that the DEGs were preferentially enriched in gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related to proliferation, development, survival, and inflammation. Furthermore, 61 of the 578 downregulated DEGs have at least one potential MEF2A binding site in the proximal promoter and were mostly enriched in the GO terms "reproduction" and "cardiovascular." The protein-protein interaction network analyzed for the downregulated DEGs and the DEGs in the GO terms "cardiovascular" and "aging" revealed that PIK3CG, IL1B, IL8, and PRKCB were included in hot nodes, and the regulation of the longevity-associated gene PIK3CG by MEF2A has been verified at the protein level, suggesting that PIK3CG might play a key role in MEF2A knockdown induced HCAEC senescence. CONCLUSIONS: MEF2A knockdown accelerates HCAEC senescence, and the underlying molecular mechanism may be involved in down-regulation of the genes related with cell proliferation, development, inflammation and survival, in which PIK3CG may play a key role.


Assuntos
Senescência Celular/genética , Vasos Coronários/citologia , Células Endoteliais , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Células Endoteliais/citologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia
4.
Anat Rec (Hoboken) ; 302(1): 136-145, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30289203

RESUMO

Primary cilia are small organelles projecting from the cell surface of many cell types. They play a crucial role in the regulation of various signaling pathway. In this study, we investigated the importance of cilia for heart development by conditionally deleting intraflagellar transport protein Ift88 using the col3.6-cre mouse. Analysis of col3.6;Ift88 offspring showed a wide spectrum of cardiovascular defects including double outlet right ventricle and atrioventricular septal defects. In addition, we found that in the majority of specimens the pulmonary veins did not properly connect to the developing left atrium. The abnormal connections found resemble those seen in patients with total anomalous pulmonary venous return. Analysis of mutant hearts at early stages of development revealed abnormal development of the dorsal mesocardium, a second heart field-derived structure at the venous pole intrinsically related to the development of the pulmonary veins. Data presented support a crucial role for primary cilia in outflow tract development and atrioventricular septation and their significance for the formation of the second heart field-derived tissues at the venous pole including the dorsal mesocardium. Furthermore, the results of this study indicate that proper formation of the dorsal mesocardium is critically important for the development of the pulmonary veins. Anat Rec, 302:136-145, 2019. © 2018 Wiley Periodicals, Inc.


Assuntos
Cílios/patologia , Modelos Animais de Doenças , Comunicação Interatrial/patologia , Veias Pulmonares/anormalidades , Síndrome de Cimitarra/patologia , Animais , Colágeno Tipo III/fisiologia , Fatores de Transcrição MEF2/fisiologia , Masculino , Camundongos , Camundongos Knockout , Penetrância , Proteínas Supressoras de Tumor/fisiologia
5.
Dev Biol ; 445(2): 170-177, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30521808

RESUMO

MEF2C is a member of the highly conserved MEF2 family of transcription factors and is a key regulator of cardiovascular development. In mice, Mef2c is expressed in the developing heart and vasculature, including the endothelium. Loss of Mef2c function in germline knockout mice leads to early embryonic demise and profound developmental abnormalities in the cardiovascular system. Previous attempts to uncover the cause of embryonic lethality by specifically disrupting Mef2c function in the heart or vasculature failed to recapitulate the global Mef2c knockout phenotype and instead resulted in relatively minor defects that did not compromise viability or result in significant cardiovascular defects. However, previous studies examined the requirement of Mef2c in the myocardial and endothelial lineages using Cre lines that begin to be expressed after the expression of Mef2c has already commenced. Here, we tested the requirement of Mef2c in the myocardial and endothelial lineages using conditional knockout approaches in mice with Cre lines that deleted Mef2c prior to onset of its expression in embryonic development. We found that deletion of Mef2c in the early myocardial lineage using Nkx2-5Cre resulted in cardiac and vascular abnormalities that were indistinguishable from the defects in the global Mef2c knockout. In contrast, early deletion of Mef2c in the vascular endothelium using an Etv2::Cre line active prior to the onset of Mef2c expression resulted in viable offspring that were indistinguishable from wild type controls with no overt defects in vascular development, despite nearly complete early deletion of Mef2c in the vascular endothelium. Thus, these studies support the idea that the requirement of MEF2C for vascular development is secondary to its requirement in the heart and suggest that the observed failure in vascular remodeling in Mef2c knockout mice results from defective heart function.


Assuntos
Sistema Cardiovascular/embriologia , Animais , Fenômenos Fisiológicos Cardiovasculares/genética , Endotélio Vascular/anormalidades , Endotélio Vascular/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Fatores de Transcrição MEF2/deficiência , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Organogênese/genética , Organogênese/fisiologia , Gravidez
6.
BMC Cancer ; 18(1): 1217, 2018 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-30514247

RESUMO

BACKGROUND: Agarose encapsulated murine renal adenocarcinoma cells (RENCA macrobeads) are currently being investigated in clinical trials as a treatment for therapy-resistant metastatic colorectal cancer. We have previously demonstrated the capacity of RENCA macrobeads to produce diffusible substances that markedly inhibit the proliferation of epithelial-derived tumor cells outside the macrobead environment. This study examined the molecular mechanisms underlying the observed inhibition in targeted tumor cells exposed to RENCA macrobeads. METHODS: We evaluated changes in transcription factor responses, participating intracellular signaling pathways and the involvement of specific cellular receptors in targeted tumor cells exposed to RENCA macrobeads. RESULTS: Factors secreted by RENCA macrobeads significantly up-regulated the activity of the MEF2 transcription factor as well as altered the transcription of MEF2b and MEF2d isoforms in targeted tumor cells. Suppression of individual or multiple MEF2 isoforms in target tumor cells markedly reduced the growth inhibitory effects of RENCA macrobeads. Furthermore, these effects were linked to the activation of the EGF receptor as attenuation of EGFR resulted in a substantial reduction of the cancer cell growth-inhibitory effect. CONCLUSIONS: Since interruption of the EGFR signaling cascade did not eliminate RENCA macrobead-induced growth control, our data suggests that RENCA macrobeads exert their full growth inhibitory effects through the simultaneous activation of multiple signaling pathways. In contrast to a precision medicine approach targeting single molecular abnormalities, the RENCA macrobead functions as a biological-systems therapy to re-establish regulation in a highly dysfunctional and dysregulated cancer system.


Assuntos
Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Microesferas , Transdução de Sinais/fisiologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Receptores ErbB/metabolismo , Gefitinibe/farmacologia , Humanos , Fatores de Transcrição MEF2/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos
7.
Gene ; 675: 312-321, 2018 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-29981832

RESUMO

Wnt16 is a major Wnt ligand involved in the regulation of postnatal bone homeostasis. Previous studies have shown that Wnt16 promotes bone formation and inhibits bone resorption, suggesting that this molecule could be targeted for therapeutic interventions to treat bone thinning disorders such as osteoporosis. However, the molecular mechanisms by which Wnt16 regulates bone metabolism is not yet fully understood. To better understand the molecular mechanisms by which Wnt16 promotes bone formation and to identify the target genes regulated by Wnt16 in osteoblasts, we treated calvarial osteoblasts purified from C57Bl/6 mice with recombinant Wnt16 and profiled the gene expression changes by RNA-seq at 24 h post-treatment. We also compared gene expression profiles of Wnt16-treated osteoblasts to canonical Wnt3a- and non-canonical Wnt5a-treated osteoblasts. This study identified 576 genes differentially expressed in Wnt16-treated osteoblasts compared to sham-treated controls; these included several members of Wnt pathway (Wnt2b, Wnt7b, Wnt11, Axin2, Sfrp2, Sfrp4, Fzd5 etc.) and TGF-ß/BMP signaling pathway (Bmp7, Inhba, Inhbb, Tgfb2 etc.). Wnt16 also regulated a large number of genes with known bone phenotypes. We also found that about 37% (215/576) of the Wnt16 targets overlapped with Wnt3a targets and ~15% (86/576) overlapped with Wnt5a targets, suggesting that Wnt16 activates both canonical and non-canonical Wnt signaling targets in osteoblasts. Transcription factor binding motif enrichment analysis in the promoter regions of Wnt16 targets identified noncanonical Wnt/JNK pathway activated transcription factors Fosl2 and Fosl1 as two of the most significantly enriched transcription factors associated with genes activated by Wnt16 while Mef2c was the most significantly enriched transcription factor associated with genes repressed by Wnt16. We also found that a large number of Mef2c targets overlapped with genes down-regulated by Wnt16 and Mef2c itself was transcriptionally repressed by Wnt16 suggesting that Mef2c plays a role in Wnt16-mediated transcriptional regulation.


Assuntos
Redes Reguladoras de Genes , Proteínas Wnt/fisiologia , Animais , Células Cultivadas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Wnt/genética , Via de Sinalização Wnt/genética
8.
Biochim Biophys Acta Rev Cancer ; 1870(2): 261-273, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29879430

RESUMO

While MEF2 transcription factors are well known to cooperate in orchestrating cell fate and adaptive responses during development and adult life, additional studies over the last decade have identified a wide spectrum of genetic alterations of MEF2 in different cancers. The consequences of these alterations, including triggering and maintaining the tumorigenic process, are not entirely clear. A deeper knowledge of the molecular pathways that regulate MEF2 expression and function, as well as the nature and consequences of MEF2 mutations are necessary to fully understand the many roles of MEF2 in malignant cells. This review discusses the current knowledge of MEF2 transcription factors in cancer.


Assuntos
Carcinogênese/metabolismo , Fatores de Transcrição MEF2/fisiologia , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Carcinogênese/patologia , Humanos
9.
PLoS One ; 13(4): e0196255, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29698438

RESUMO

Myocyte enhancer factor 2A (MEF2A) is widely distributed in various tissues or organs and plays crucial roles in multiple biological processes. To examine the potential effects of MEF2A on skeletal muscle myoblast, the functional role of MFE2A in myoblast proliferation and differentiation was investigated. In this study, we found that the mRNA expression level of Mef2a was dramatically increased during the myogenesis of bovine skeletal muscle primary myoblast. Overexpression of MEF2A significantly promoted myoblast proliferation, while knockdown of MEF2A inhibited the proliferation and differentiation of myoblast. RT-PCR and western blot analysis revealed that this positive effect of MEF2A on the proliferation of myoblast was carried out by triggering cell cycle progression by activating CDK2 protein expression. Besides, MEF2A was found to be an important transcription factor that bound to the myozenin 2 (MyoZ2) proximal promoter and performed upstream of MyoZ2 during myoblast differentiation. This study provides the first experimental evidence that MEF2A is a positive regulator in skeletal muscle myoblast proliferation and suggests that MEF2A regulates myoblast differentiation via regulating MyoZ2.


Assuntos
Fatores de Transcrição MEF2/fisiologia , Músculo Esquelético/fisiologia , Mioblastos Esqueléticos/fisiologia , Mioblastos Esqueléticos/ultraestrutura , Adenoviridae/genética , Animais , Bovinos , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Citometria de Fluxo , Regulação da Expressão Gênica , Desenvolvimento Muscular , Proteínas Musculares/fisiologia , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo
10.
Nat Commun ; 7: 12376, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27507714

RESUMO

DNA double strand break (DSB) repair is critical for generation of B-cell receptors, which are pre-requisite for B-cell progenitor survival. However, the transcription factors that promote DSB repair in B cells are not known. Here we show that MEF2C enhances the expression of DNA repair and recombination factors in B-cell progenitors, promoting DSB repair, V(D)J recombination and cell survival. Although Mef2c-deficient mice maintain relatively intact peripheral B-lymphoid cellularity during homeostasis, they exhibit poor B-lymphoid recovery after sub-lethal irradiation and 5-fluorouracil injection. MEF2C binds active regulatory regions with high-chromatin accessibility in DNA repair and V(D)J genes in both mouse B-cell progenitors and human B lymphoblasts. Loss of Mef2c in pre-B cells reduces chromatin accessibility in multiple regulatory regions of the MEF2C-activated genes. MEF2C therefore protects B lymphopoiesis during stress by ensuring proper expression of genes that encode DNA repair and B-cell factors.


Assuntos
Quebras de DNA de Cadeia Dupla , Hematopoese/fisiologia , Células Precursoras de Linfócitos B/fisiologia , Recombinação V(D)J/fisiologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Cromatina/metabolismo , Feminino , Fluoruracila/farmacologia , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Fatores de Transcrição MEF2/fisiologia , Masculino , Camundongos , Células Precursoras de Linfócitos B/efeitos dos fármacos , Células Precursoras de Linfócitos B/efeitos da radiação , Irradiação Corporal Total/efeitos adversos
11.
Oncotarget ; 7(29): 45398-45413, 2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27322685

RESUMO

Recently, we identified deregulated expression of the B-cell specific transcription factor MEF2C in T-cell acute lymphoid leukemia (T-ALL). Here, we performed sequence analysis of a regulatory upstream section of MEF2C in T-ALL cell lines which, however, proved devoid of mutations. Unexpectedly, we found strong conservation between the regulatory upstream region of MEF2C (located at chromosomal band 5q14) and an intergenic stretch at 7q11 located between STAG3L4 and AUTS2, covering nearly 20 kb. While the non-coding gene STAG3L4 was inconspicuously expressed, AUTS2 was aberrantly upregulated in 6% of T-ALL patients (public dataset GSE42038) and in 3/24 T-ALL cell lines, two of which represented very immature differentiation stages. AUTS2 expression was higher in normal B-cells than in T-cells, indicating lineage-specific activity in lymphopoiesis. While excluding chromosomal aberrations, examinations of AUTS2 transcriptional regulation in T-ALL cells revealed activation by IL7-IL7R-STAT5-signalling and MEF2C. AUTS2 protein has been shown to interact with polycomb repressor complex 1 subtype 5 (PRC1.5), transforming this particular complex into an activator. Accordingly, expression profiling and functional analyses demonstrated that AUTS2 activated while PCGF5 repressed transcription of NKL homeobox gene MSX1 in T-ALL cells. Forced expression and pharmacological inhibition of EZH2 in addition to H3K27me3 analysis indicated that PRC2 repressed MSX1 as well. Taken together, we found that AUTS2 and MEF2C, despite lying on different chromosomes, share strikingly similar regulatory upstream regions and aberrant expression in T-ALL subsets. Our data implicate chromatin complexes PRC1/AUTS2 and PRC2 in a gene network in T-ALL regulating early lymphoid differentiation.


Assuntos
Complexo Repressor Polycomb 1/fisiologia , Complexo Repressor Polycomb 2/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas/fisiologia , Linhagem Celular Tumoral , Proteínas do Citoesqueleto , Redes Reguladoras de Genes , Humanos , Fatores de Transcrição MEF2/fisiologia , Fator de Transcrição MSX1/genética , Fator de Transcrição STAT5/fisiologia , Fatores de Transcrição
12.
Development ; 143(5): 774-9, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26811383

RESUMO

Congenital heart defects are the most common birth defects in humans, and those that affect the proper alignment of the outflow tracts and septation of the ventricles are a highly significant cause of morbidity and mortality in infants. A late differentiating population of cardiac progenitors, referred to as the anterior second heart field (AHF), gives rise to the outflow tract and the majority of the right ventricle and provides an embryological context for understanding cardiac outflow tract alignment and membranous ventricular septal defects. However, the transcriptional pathways controlling AHF development and their roles in congenital heart defects remain incompletely elucidated. Here, we inactivated the gene encoding the transcription factor MEF2C in the AHF in mice. Loss of Mef2c function in the AHF results in a spectrum of outflow tract alignment defects ranging from overriding aorta to double-outlet right ventricle and dextro-transposition of the great arteries. We identify Tdgf1, which encodes a Nodal co-receptor (also known as Cripto), as a direct transcriptional target of MEF2C in the outflow tract via an AHF-restricted Tdgf1 enhancer. Importantly, both the MEF2C and TDGF1 genes are associated with congenital heart defects in humans. Thus, these studies establish a direct transcriptional pathway between the core cardiac transcription factor MEF2C and the human congenital heart disease gene TDGF1. Moreover, we found a range of outflow tract alignment defects resulting from a single genetic lesion, supporting the idea that AHF-derived outflow tract alignment defects may constitute an embryological spectrum rather than distinct anomalies.


Assuntos
Fator de Crescimento Epidérmico/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/genética , Feminino , Deleção de Genes , Coração/embriologia , Cardiopatias Congênitas/genética , Comunicação Interventricular/genética , Ventrículos do Coração , Humanos , Hibridização In Situ , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Morfogênese/genética , Proteínas de Neoplasias/genética , Organogênese , Análise de Sequência de RNA , Distribuição Tecidual , Transcrição Gênica , Transposição dos Grandes Vasos/genética
13.
Hepatol Int ; 10(3): 493-500, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26797719

RESUMO

BACKGROUND: Pokemon, a master oncogene crucial for the tumorigenicity and progression of a variety of cancers, has been demonstrated to enhance the proliferation and survival of hepatocellular carcinoma (HCC). However, the contribution of Pokemon to the invasiveness of HCC has not yet been studied. METHODS: In this study, we employed HCC cells to investigate the role of Pokemon in the invasion of HCC with multidisciplinary approaches. RESULTS: Pokemon overexpression was found to be closely associated with invasion and intrahepatic metastasis of HCC in clinical specimens. Suppression of Pokemon attenuated the invasion of HCC cells by in vitro transwell and wound-healing assays. Myocyte enhancer factor 2D (MEF2D), an oncogene that can promote the invasiveness of HCC, was found to be underexpressed during Pokemon silencing in HCC cells. Restoration of MEF2D abolished the effect of Pokemon downregulation on the migration of HCC cells. Further experiments verified that Pokemon binds two putative recognition sites located within the upstream region of the MEF2D promoter and enhances its transcription. The association between Pokemon and MEF2D was further confirmed in HCC specimens. Animal experiments further confirmed that Pokemon downregulation attenuated the metastasis of HCC cells in mice. CONCLUSION: Collectively, Pokemon was found to enhance the migration and invasion of HCC by increasing MEF2D expression. Thus, targeting Pokemon and MEF2D may be an effective strategy to suppress the metastasis of HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Proteínas de Ligação a DNA/fisiologia , Neoplasias Hepáticas/patologia , Fatores de Transcrição/fisiologia , Animais , Western Blotting , Carcinoma Hepatocelular/fisiopatologia , Linhagem Celular Tumoral , Células Hep G2 , Humanos , Neoplasias Hepáticas/fisiopatologia , Fatores de Transcrição MEF2/fisiologia , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica
14.
Tumour Biol ; 37(3): 2891-900, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26409449

RESUMO

Lung carcinoma is a deadly malignant disease with poor prognosis and increasing incidence in recent years. However, the molecular mechanism underlying the initiation and progression of lung cancer is still not completely elucidated. Recently, myocyte enhancer factor 2D (MEF2D) has been reported to promote the growth of liver cancer, but its implication in lung cancer is still unknown. This study is aimed to determine the role of MEF2D in lung carcinoma. Quantitative PCR (qPCR) and immunoblot assays showed that MEF2D was overexpressed in lung cancer tissues and cell lines, compared with the matched normal tissues and cell lines. Small interfering RNA (siRNA) suppression of MEF2D was able to reduce the proliferation, survival, and invasion of lung carcinoma cells. The transfection of MEF2D-expressing constructs into normal lung fibroblast cells promoted their proliferation and motility. The role of MEF2D in the growth of lung cancer was also confirmed in mice. Further study revealed that miR-218, which was underexpressed in lung carcinoma, was predicted to bind the 3'-untranslated region (UTR) of MEF2D mRNA. miR-218 was shown to suppress the activity of luciferase with MEF2D 3'-UTR. The changes in miR-218 levels affected the expression of MEF2D in lung cancer cells and normal fibroblast cells. There is also an inverse association between miR-218 abundance and MEF2D levels in the lung carcinoma specimen. Furthermore, the transfection of a plasmid that expressed MEF2D resistance to miR-218 regulation abolished the inhibitory effect of miR-218 on lung cancer cells. Collectively, MEF2D overexpression participated in the growth of lung cancers and its aberrant expression may result from the reduction of tumor suppressor miR-218.


Assuntos
Neoplasias Pulmonares/patologia , MicroRNAs/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Fatores de Transcrição MEF2/análise , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica
15.
Proc Natl Acad Sci U S A ; 112(51): 15755-60, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26647181

RESUMO

Ketamine produces rapid antidepressant-like effects in animal assays for depression, although the molecular mechanisms underlying these behavioral actions remain incomplete. Here, we demonstrate that ketamine rapidly stimulates histone deacetylase 5 (HDAC5) phosphorylation and nuclear export in rat hippocampal neurons through calcium/calmodulin kinase II- and protein kinase D-dependent pathways. Consequently, ketamine enhanced the transcriptional activity of myocyte enhancer factor 2 (MEF2), which leads to regulation of MEF2 target genes. Transfection of a HDAC5 phosphorylation-defective mutant (Ser259/Ser498 replaced by Ala259/Ala498, HDAC5-S/A), resulted in resistance to ketamine-induced nuclear export, suppression of ketamine-mediated MEF2 transcriptional activity, and decreased expression of MEF2 target genes. Behaviorally, viral-mediated hippocampal knockdown of HDAC5 blocked or occluded the antidepressant effects of ketamine both in unstressed and stressed animals. Taken together, our results reveal a novel role of HDAC5 in the actions of ketamine and suggest that HDAC5 could be a potential mechanism contributing to the therapeutic actions of ketamine.


Assuntos
Antidepressivos/farmacologia , Núcleo Celular/metabolismo , Histona Desacetilases/metabolismo , Ketamina/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Células Cultivadas , Hipocampo/metabolismo , Fatores de Transcrição MEF2/fisiologia , Fosforilação , Ratos , Ratos Sprague-Dawley
17.
Cell Death Dis ; 6: e1944, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26512955

RESUMO

Exposure to metabolic disease during fetal development alters cellular differentiation and perturbs metabolic homeostasis, but the underlying molecular regulators of this phenomenon in muscle cells are not completely understood. To address this, we undertook a computational approach to identify cooperating partners of the myocyte enhancer factor-2 (MEF2) family of transcription factors, known regulators of muscle differentiation and metabolic function. We demonstrate that MEF2 and the serum response factor (SRF) collaboratively regulate the expression of numerous muscle-specific genes, including microRNA-133a (miR-133a). Using tandem mass spectrometry techniques, we identify a conserved phosphorylation motif within the MEF2 and SRF Mcm1 Agamous Deficiens SRF (MADS)-box that regulates miR-133a expression and mitochondrial function in response to a lipotoxic signal. Furthermore, reconstitution of MEF2 function by expression of a neutralizing mutation in this identified phosphorylation motif restores miR-133a expression and mitochondrial membrane potential during lipotoxicity. Mechanistically, we demonstrate that miR-133a regulates mitochondrial function through translational inhibition of a mitophagy and cell death modulating protein, called Nix. Finally, we show that rodents exposed to gestational diabetes during fetal development display muscle diacylglycerol accumulation, concurrent with insulin resistance, reduced miR-133a, and elevated Nix expression, as young adult rats. Given the diverse roles of miR-133a and Nix in regulating mitochondrial function, and proliferation in certain cancers, dysregulation of this genetic pathway may have broad implications involving insulin resistance, cardiovascular disease, and cancer biology.


Assuntos
Diferenciação Celular/genética , Fatores de Transcrição MEF2/química , Mitocôndrias/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Resposta Sérica/química , Motivos de Aminoácidos , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Diabetes Gestacional , Feminino , Regulação da Expressão Gênica , Humanos , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/fisiologia , Potencial da Membrana Mitocondrial/genética , MicroRNAs/metabolismo , Mitocôndrias/genética , Fibras Musculares Esqueléticas/citologia , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/citologia , Miócitos de Músculo Liso/citologia , Fosforilação , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Fator de Resposta Sérica/metabolismo , Fator de Resposta Sérica/fisiologia , Espectrometria de Massas em Tandem
18.
Metabolism ; 64(12): 1682-93, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26455966

RESUMO

OBJECTIVE: Evidence shows that both macrophage migration inhibitory factor (MIF) and GLUT4 glucose transporter are involved in diabetic cardiomyopathy (DCM), but it remains largely unknown whether and how MIF regulates GLUT4 expression in cardiomyocytes. The present study aims to investigate the mechanism underlying the modulation of GLUT4 by MIF in cardiomyocytes. MATERIAL AND METHODS: Activations of AKT and AMPK signaling, and expressions of MIF, GLUT4 and the candidate GLUT4 regulation associated transcription factors in the diabetic mouse myocardium were determined. The screened transcription factors mediating MIF-promoted GLUT4 expression were verified by RNA interference (RNAi) and electrophoretic mobility shift assay (EMSA), respectively. RESULTS: MIF was increased, but GLUT4 was decreased in the diabetic mouse myocardium. MIF could enhance glucose uptake and up-regulate GLUT4 expression in NMVCs. Expressions of transcription factor MEF2A, -2C, -2D and Zac1 were significantly up-regulated in MIF-treated neonatal mouse ventricular cardiomyocytes (NMVCs), and markedly reduced in the diabetic myocardium. Knockdown of MEF2A, -2C, -2D and Zac1 could significantly inhibit glucose uptake and GLUT4 expression in cardiomyocytes. Moreover, EMSA results revealed that transcriptional activities of MEF2 and Zac1 were significantly increased in MIF-treated NMVCs. AMPK signaling was activated in MIF-stimulated NMVCs, and AMPK activator AICAR could enhance MEF2A, -2C, -2D, Zac1 and GLUT4 expression. Additionally, MIF effects were inhibited by an AMPK inhibitor compound C and siRNA targeting MIF receptor CD74, suggesting the involvement of CD74-dependent AMPK activation. CONCLUSIONS: Transcription factor MEF2 and Zac1 mediate MIF-induced GLUT4 expression through CD74-dependent AMPK activation in cardiomyocytes.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Genes Supressores de Tumor/fisiologia , Transportador de Glucose Tipo 4/genética , Oxirredutases Intramoleculares/fisiologia , Fatores de Transcrição MEF2/fisiologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Antígenos de Diferenciação de Linfócitos B/fisiologia , Células Cultivadas , Cardiomiopatias Diabéticas/fisiopatologia , Antígenos de Histocompatibilidade Classe II/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Insulina/fisiologia , Função Ventricular Esquerda
19.
J Biol Chem ; 290(40): 24367-80, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26294766

RESUMO

The cardiomyocyte cell cycle is a poorly understood process. Mammalian cardiomyocytes permanently withdraw from the cell cycle shortly after birth but can re-enter the cell cycle and proliferate when subjected to injury within a brief temporal window in the neonatal period. Thus, investigating the mechanisms of cell cycle regulation in neonatal cardiomyocytes may provide critical insight into the molecular events that prevent adult myocytes from proliferating in response to injury or stress. MEF2D is a key transcriptional mediator of pathological remodeling in the adult heart downstream of various stress-promoting insults. However, the specific gene programs regulated by MEF2D in cardiomyocytes are unknown. By performing genome-wide transcriptome analysis using MEF2D-depleted neonatal cardiomyocytes, we found a significant impairment in the cell cycle, characterized by the up-regulation of numerous positive cell cycle regulators. Expression of Pten, the primary negative regulator of PI3K/Akt, was significantly reduced in MEF2D-deficient cardiomyocytes and found to be a direct target gene of MEF2D. Consistent with these findings mutant cardiomyocytes showed activation of the PI3K/Akt survival pathway. Paradoxically, prolonged deficiency of MEF2D in neonatal cardiomyocytes did not trigger proliferation but instead resulted in programmed cell death, which is likely mediated by the E2F transcription factor. These results demonstrate a critical role for MEF2D in cell cycle regulation of post-mitotic, neonatal cardiomyocytes in vitro.


Assuntos
Miócitos Cardíacos/citologia , Animais , Animais Recém-Nascidos , Apoptose , Caspase 3/metabolismo , Ciclo Celular , Proliferação de Células , Sobrevivência Celular , Fatores de Transcrição E2F/metabolismo , Fibroblastos/metabolismo , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Transcriptoma
20.
FEBS J ; 282(15): 2930-47, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26038288

RESUMO

In Xenopus, animal cap (AC) cells give rise to ectoderm and its derivatives: epidermis and the central nervous system. Ectoderm has long been considered a default pathway of embryonic development, with cells that are not under the influence of vegetal Nodal signaling adopting an ectodermal program of gene expression. In the present study, we describe the involvement of the animally-localized maternal transcription factor myocyte enhancer factor (Mef) 2D in regulating the identity of AC cells. We find that Mef2D is required for the formation of both ectodermal lineages: neural and epidermis. Gain and loss of function experiments indicate that Mef2D regulates early gastrula expression of key ectodermal/epidermal genes in the animal region. Mef2D controls the activity of zygotic bone morphogenetic protein (BMP) signaling known to dictate the epidermal differentiation program. Exogenous expression of Mef2D in vegetal blastomeres was sufficient to induce ectopic expression of ectoderm/epidermal genes in the vegetal half of the embryo, when Nodal signaling was inhibited. Depletion of Mef2D caused a loss of AC cell adhesion that was rescued by the expression of E-cadherin or bone morphogenetic protein 4. In addition, expression of Mef2D in the prospective endoderm caused unusual aggregation of vegetal cells with animal cells in vitro and inappropriate segregation to other germ layers in vivo. Mef2D cooperates with another animally-expressed transcription factor, FoxI1e. Together, they regulate the expression of genes encoding signaling proteins and the transcription factors that control the regional identity of animal cells. Therefore, we describe a new role for the animally-localized Mef2D protein in early ectoderm specification, which is similar to that of the vegetally-localized VegT in endoderm and mesoderm formation.


Assuntos
Adesão Celular/fisiologia , Ectoderma/citologia , Fatores de Transcrição MEF2/fisiologia , Xenopus/embriologia , Animais , Linhagem da Célula , Gástrula/química , Transdução de Sinais , Xenopus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA