Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 71(1): 129-141.e8, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29979962

RESUMO

The enhancer regions of the myogenic master regulator MyoD give rise to at least two enhancer RNAs. Core enhancer eRNA (CEeRNA) regulates transcription of the adjacent MyoD gene, whereas DRReRNA affects expression of Myogenin in trans. We found that DRReRNA is recruited at the Myogenin locus, where it colocalizes with Myogenin nascent transcripts. DRReRNA associates with the cohesin complex, and this association correlates with its transactivating properties. Despite being expressed in undifferentiated cells, cohesin is not loaded on Myogenin until the cells start expressing DRReRNA, which is then required for cohesin chromatin recruitment and maintenance. Functionally, depletion of either cohesin or DRReRNA reduces chromatin accessibility, prevents Myogenin activation, and hinders muscle cell differentiation. Thus, DRReRNA ensures spatially appropriate cohesin loading in trans to regulate gene expression.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Proteínas Cromossômicas não Histona/biossíntese , Elementos Facilitadores Genéticos , Músculo Esquelético/metabolismo , Miogenina/biossíntese , RNA não Traduzido/metabolismo , Transcrição Gênica , Animais , Proteínas de Ciclo Celular/genética , Diferenciação Celular , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Células HEK293 , Humanos , Camundongos , Músculo Esquelético/citologia , Proteína MyoD/biossíntese , Proteína MyoD/genética , Miogenina/genética , RNA não Traduzido/genética , Coesinas
2.
J Anat ; 234(3): 359-367, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30657171

RESUMO

Skeletal muscle has the remarkable capability to regenerate itself following injury. Adult myogenic stem cells (MSCs) are responsible for the repair and regeneration, and their activity is controlled by intrinsic and extrinsic factors. The aim of this study was to examine and compare the expression levels of Pax3, Pax7, MRF and p38 proteins during the course of regeneration and in different areas of the focal freeze-lesion damaged adult rat TA muscle. Using the focal freeze injury model, immunohistochemistry, laser-capture micro-dissection and Western blot analysis were performed. The results show that (1) in the severely damaged area, the focal freeze-lesion injury significantly activated Pax7 and myogenin expression within 7 days and down-regulated Pax3, MyoD and Myf-5 within 1 or 3 days, and (2) the level of the p38 protein was strongly and transiently up-regulated in the whole muscle on day 7 following injury, whereas the level of the pp38 protein was down-regulated within 3 days in the severely damaged and non-damaged areas. These findings indicate that the temporal (e.g. the time course of regeneration) and spatial (e.g. three zones created by the focal freeze-lesion) cues in a regenerating muscle have a significant impact on the activity of the adult MSCs.


Assuntos
Proteínas Musculares/metabolismo , Fatores de Regulação Miogênica/metabolismo , Miogenina/metabolismo , Regeneração/fisiologia , Animais , Diferenciação Celular/fisiologia , Congelamento , Imuno-Histoquímica/métodos , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/patologia , Miogenina/biossíntese , Fator de Transcrição PAX3/metabolismo , Fator de Transcrição PAX7/metabolismo , Ratos , Ferimentos e Lesões , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Dev Biol ; 402(1): 61-71, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25796573

RESUMO

Klhl31 is a member of the Kelch-like family in vertebrates, which are characterized by an amino-terminal broad complex tram-track, bric-a-brac/poxvirus and zinc finger (BTB/POZ) domain, carboxy-terminal Kelch repeats and a central linker region (Back domain). In developing somites Klhl31 is highly expressed in the myotome downstream of myogenic regulators (MRF), and it remains expressed in differentiated skeletal muscle. In vivo gain- and loss-of-function approaches in chick embryos reveal a role of Klhl31 in skeletal myogenesis. Targeted mis-expression of Klhl31 led to a reduced size of dermomyotome and myotome as indicated by detection of relevant myogenic markers, Pax3, Myf5, myogenin and myosin heavy chain (MF20). The knock-down of Klhl31 in developing somites, using antisense morpholinos (MO), led to an expansion of Pax3, Myf5, MyoD and myogenin expression domains and an increase in the number of mitotic cells in the dermomyotome and myotome. The mechanism underlying this phenotype was examined using complementary approaches, which show that Klhl31 interferes with ß-catenin dependent Wnt signaling. Klhl31 reduced the Wnt-mediated activation of a luciferase reporter in cultured cells. Furthermore, Klhl31 attenuated secondary axis formation in Xenopus embryos in response to Wnt1 or ß-catenin. Klhl31 mis-expression in the developing neural tube affected its dorso-ventral patterning and led to reduced dermomyotome and myotome size. Co-transfection of a Wnt3a expression vector with Klhl31 in somites or in the neural tube rescued the phenotype and restored the size of dermomyotome and myotome. Thus, Klhl31 is a novel modulator of canonical Wnt signaling, important for vertebrate myogenesis. We propose that Klhl31 acts in the myotome to support cell cycle withdrawal and differentiation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Proliferação de Células , Embrião de Galinha , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Hibridização In Situ , Mitose , Músculos/embriologia , Miogenina/biossíntese , Tubo Neural/metabolismo , Fenótipo , Transdução de Sinais , Somitos/metabolismo , Xenopus laevis , beta Catenina/genética
4.
Dev Growth Differ ; 58(6): 546-61, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27224259

RESUMO

Muscle regeneration is an important process for skeletal muscle growth and recovery. Repair of muscle damage is exquisitely programmed by cellular mechanisms inherent in myogenic stem cells, also known as muscle satellite cells. We demonstrated previously the involvement of homeobox transcription factors, SIX1, SIX4 and SIX5, in the coordinated proliferation and differentiation of isolated satellite cells in vitro. However, their roles in adult muscle regeneration in vivo remain elusive. To investigate SIX4 and SIX5 functions during muscle regeneration, we introduced knockout alleles of Six4 and Six5 into an animal model of Duchenne Muscular Dystrophy (DMD), mdx (Dmd(mdx) /Y) mice, characterized by frequent degeneration-regeneration cycles in muscles. A lower number of small myofibers, higher number of thick ones and lower serum creatine kinase and lactate dehydrogenase activities were noted in 50-week-old Six4(+/-) 5(+/-) Dmd(mdx) /Y mice than Dmd(mdx) /Y mice, indicating improvement of dystrophic phenotypes of Dmd(mdx) /Y mice. Higher proportions of cells positive for MYOD1 and MYOG (markers of regenerating myonuclei) and SIX1 (a marker of regenerating myoblasts and newly regenerated myofibers) in 12-week-old Six4(+/-) 5(+/-) Dmd(mdx) /Y mice suggested enhanced regeneration, compared with Dmd(mdx) /Y mice. Although grip strength was comparable in Six4(+/-) 5(+/-) Dmd(mdx) /Y and Dmd(mdx) /Y mice, treadmill exercise did not induce muscle weakness in Six4(+/-) 5(+/-) Dmd(mdx) /Y mice, suggesting higher regeneration capacity. In addition, Six4(+/-) 5(+/-) Dmd(mdx) /Y mice showed 33.8% extension of life span. The results indicated that low Six4 and Six5 gene dosage improved dystrophic phenotypes of Dmd(mdx) /Y mice by enhancing muscle regeneration, and suggested that SIX4 and SIX5 are potentially useful de novo targets in therapeutic applications against muscle disorders, including DMD.


Assuntos
Dosagem de Genes/fisiologia , Proteínas de Homeodomínio/metabolismo , Longevidade/fisiologia , Músculo Esquelético/fisiologia , Regeneração/fisiologia , Transativadores/metabolismo , Animais , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos mdx , Camundongos Knockout , Proteína MyoD/biossíntese , Proteína MyoD/genética , Miogenina/biossíntese , Miogenina/genética , Transativadores/genética
5.
Cryobiology ; 73(2): 112-9, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27593478

RESUMO

Foxo4 and MyoG proteins regulate the transcription of numerous genes, including the E3 ubiquitin ligases MAFbx and MuRF1, which are activated in skeletal muscle under atrophy-inducing conditions. In the thirteen-lined ground squirrel, there is little muscle wasting that occurs during hibernation, a process characterized by bouts of torpor and arousal, despite virtual inactivity. Consequently, we were interested in studying the regulatory role of Foxo4 and MyoG on ubiquitin ligases throughout torpor-arousal cycles. Findings indicate that MAFbx and MuRF1 decreased during early torpor (ET) by 42% and 40%, respectively, relative to euthermic control (EC), although MuRF1 expression subsequently increased at late torpor (LT). The expression pattern of MyoG most closely resembled that of MAFbx, with levels decreasing during LT. In addition, the phosphorylation of Foxo4 at Thr-451 showed an initial increase during EN, followed by a decline throughout the remainder of the torpor-arousal cycle, suggesting Foxo4 inhibition. This trend was mirrored by inhibition of the Ras-Ral pathway, as the Ras and Ral proteins were decreased by 77% and 41% respectively, at ET. Foxo4 phosphorylation at S197 was depressed during entrance and torpor, suggesting Foxo4 nuclear localization, and possibly regulating the increase in MuRF1 levels at LT. These findings indicate that signaling pathways involved in regulating muscle atrophy, such as MyoG and Foxo4 through the Ras-Ral pathway, contribute to important muscle-specific changes during hibernation. Therefore, this data provides novel insight into the molecular mechanisms regulating muscle remodeling in a hibernator model.


Assuntos
Atrofia/fisiopatologia , Hibernação/fisiologia , Músculo Esquelético/metabolismo , Miogenina/biossíntese , Sciuridae/fisiologia , Torpor/fisiologia , Fatores de Transcrição/metabolismo , Animais , Regulação para Baixo , Fosforilação , Transdução de Sinais , Transcrição Gênica/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo
6.
J Biol Chem ; 289(7): 3923-35, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24385428

RESUMO

The complex process of skeletal muscle differentiation is organized by the myogenic regulatory factors (MRFs), Myf5, MyoD, Myf6, and myogenin, where myogenin plays a critical role in the regulation of the final stage of muscle differentiation. In an effort to investigate the role microRNAs (miRNAs) play in regulating myogenin, a bioinformatics approach was used and six miRNAs (miR-182, miR-186, miR-135, miR-491, miR-329, and miR-96) were predicted to bind the myogenin 3'-untranslated region (UTR). However, luciferase assays showed only miR-186 inhibited translation and 3'-UTR mutagenesis analysis confirmed this interaction was specific. Interestingly, the expression of miR-186 mirrored that of its host gene, ZRANB2, during development. Functional studies demonstrated that miR-186 overexpression inhibited the differentiation of C2C12 and primary muscle cells. Our findings therefore identify miR-186 as a novel regulator of myogenic differentiation.


Assuntos
Regiões 3' não Traduzidas/fisiologia , Diferenciação Celular/fisiologia , MicroRNAs/metabolismo , Músculo Esquelético/metabolismo , Miogenina/biossíntese , Animais , Linhagem Celular , Camundongos , MicroRNAs/genética , Músculo Esquelético/citologia , Miogenina/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
7.
Mol Cell Biochem ; 409(1-2): 271-82, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26272338

RESUMO

Several hormones and growth factors, including adipokines, play important roles during muscle development and regeneration. CTRP3, a paralog of adiponectin, is a member of the C1q and tumor necrosis factor-related protein (CTRP) superfamily. CTRP3 is a novel adipokine previously reported to reduce glucose output in hepatocytes and lower glucose levels in mice models. In the present study, we provide the first evidence for a physiological role of the CTRP3 in myogenesis using C2C12 myoblasts. CTRP3 was expressed in developing skeletal muscle tissues, and the expression level of CTRP3 was increased during myogenic differentiation of C2C12 cells. Recombinant CTRP3 (rCTRP3) promoted the proliferation of undifferentiated C2C12 myoblasts and this response required activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. In contrary, rCTRP3 inhibited myogenic differentiation and fusion of C2C12 cells by suppressing the expression of myogenic marker genes (myogenin and myosin heavy chain). CTRP3 mRNA expression was increased in C2C12 myoblasts treated with transforming growth factor-ß3 (TGF-ß3), suggesting that TGF-ß3 is one of the extracellular factors regulating CTRP3 expression during myogenesis. These results indicate a novel physiological role for CTRP3 during skeletal myogenesis.


Assuntos
Adipocinas/metabolismo , Diferenciação Celular/fisiologia , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/embriologia , Mioblastos/metabolismo , Adipocinas/genética , Animais , Linhagem Celular , Proliferação de Células/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Miogenina/biossíntese , Cadeias Pesadas de Miosina/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fator de Crescimento Transformador beta3/farmacologia
8.
Histochem Cell Biol ; 141(3): 289-300, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24186058

RESUMO

The embryonic muscles of the axial skeleton and limbs take their origin from the dermomyotomes of the somites. During embryonic myogenesis, muscle precursors delaminate from the dermomyotome giving rise to the hypaxial and epaxial myotome. Mutant studies for myogenic regulatory factors have shown that the development of the hypaxial myotome differs from the formation of the epaxial myotome and that the development of the hypaxial myotome depends on the latter within the trunk region. The transcriptional networks that regulate the transition of proliferative dermomyotomal cells into the predominantly post-mitotic hypaxial myotome, as well as the eventual patterning of the myotome, are not fully understood. Similar transitions occurring during the development of the neural system have been shown to be controlled by the Atonal family of helix-loop-helix transcription factors. Here, we demonstrate that ATOH8, a member of the Atonal family, is expressed in a subset of embryonic muscle cells in the dermomyotome and myotome. Using the RNAi approach, we show that loss of ATOH8 in the lateral somites at the trunk level results in a blockage of differentiation and thus causes cells to be maintained in a predetermined state. Furthermore, we show that ATOH8 is also expressed in cultured C2C12 mouse myoblasts and becomes dramatically downregulated during their differentiation. We propose that ATOH8 plays a role during the transition of myoblasts from the proliferative phase to the differentiation phase and in the regulation of myogenesis in the hypaxial myotome of the trunk.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Padronização Corporal/genética , Desenvolvimento Muscular/genética , Músculo Esquelético/embriologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula , Embrião de Galinha , Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Mioblastos/citologia , Fator Regulador Miogênico 5/biossíntese , Miogenina/biossíntese , Fator de Transcrição PAX7/biossíntese , Interferência de RNA , RNA Interferente Pequeno , Somitos/fisiologia
9.
Mol Biol Rep ; 41(2): 1003-13, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24385300

RESUMO

This experiment was conducted to explore the biological functions of myogenin (MyoG) gene. MyoG gene was cloned from genome of Hu sheep by overlap extension PCR. Then, pEGFP-C1-MyoG and pcDNA3.0-MyoG fusion expression vectors was constructed and pEGFP-C1-MyoG vector had been transfected into NIH-3T3 cells by liposomes-mediated method, and MyoG was detected in vitro by RT-PCR,western blotting and its subcellular localization by EGFP marker. pcDNA3.0-MyoG was transfected into goat embryonic fibroblasts (GEF) cells in order to detect the myogenic function of MyoG in vitro. Then pEGFP-C1-MyoG plasmid was injected into the testes of sheep and goat, respectively, to produce the transgenic generation. The results showed that the length of MyoG coding region of Hu sheep was 675 bp, encoding 224 amino acids. Compared with goat, cattle, pig and rat, the sequence homology of sheep MyoG cDNA was 99.26, 97.04, 92.00, and 87.70 %, respectively. The bioinformatics prediction showed that MyoG protein contained a typical bHLH structure, but without a short signal peptide, revealing that MyoG protein might be a non-secretory protein. The result of RT-PCR and western blotting demonstrated that MyoG could be expressed successfully in the transfected cells in vitro and the MyoG protein was located in nucleus. The positive transfected GEF cells with pcDNA3.0-MyoG were found to express desmin protein. The positive rates of transgenic sheep and transgenic goat were 7.1 and 7.4 % in F1 generation, respectively. Conclusively, MyoG cDNA from Hu sheep had been cloned successfully. The subcellular localization and myogenic activity of MyoG were exactly detected on the basis of multiple biological analyses, which expanded our understanding of the biological function of MyoG.


Assuntos
Animais Geneticamente Modificados/genética , Clonagem Molecular , Miogenina/genética , Carneiro Doméstico/genética , Animais , DNA Complementar/genética , Vetores Genéticos , Cabras/genética , Camundongos , Miogenina/biossíntese , Células NIH 3T3 , Ratos
10.
J Musculoskelet Neuronal Interact ; 14(3): 325-33, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25198228

RESUMO

Heat stress could promote skeletal muscle regeneration. But, in the regeneration process, effects of heat stress on myogenic cells and the regulating factors is unknown. Therefore, Influences of heat stress soon after injury on distribution of the myogenic cells and chronological changes in expression of MyoD and myogenin were examined. The first peak of MyoD expression was temporally correlated with the time when proliferating satellite cells began to appear, and the rapid decline of the MyoD expression from the first peak, with the appearance time of myoblasts, respectively in both the non-Heat and Heat groups. The first peak of myogenin expression was temporally correlated with the time when multinuclear cells began to form in the both groups. Due to the heat stress, proliferation and differentiation of myogenic cells and chronological changes in these factors were accelerated one day earlier than in the non-Heat group. As MyoD and myogenin are regulating factor of proliferation and differentiation, heat stress soon after the muscle injury could accelerate the proliferation and differentiation of myogenic cells and the expression of their regulating factors MyoD and myogenin.


Assuntos
Transtornos de Estresse por Calor/metabolismo , Músculo Esquelético/lesões , Proteína MyoD/biossíntese , Miogenina/biossíntese , Regeneração , Animais , Diferenciação Celular , Transtornos de Estresse por Calor/complicações , Transtornos de Estresse por Calor/patologia , Masculino , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/patologia , Ratos , Células Satélites de Músculo Esquelético/metabolismo
11.
Genet Mol Res ; 13(2): 2772-83, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24782091

RESUMO

In the present study, real time-polymerase chain reaction was applied to analyze the expression of IGF-I and MyoG genes in Hu sheep longissimus dorsi at different growth stages and their association with meat traits. Expression of the IGF-I gene in Hu sheep differed significantly between males and females at the two day-old (0.01 < P < 0.05), one-month old (0.01 < P < 0.05), and three month-old (P < 0.01) stages. IGF-I gene expression in male longissimus muscles was higher than that of females at all growth stages, except for the three month-old stage. There was no significant difference (P > 0.05) between males and females at any growth stage in expression of the MyoG gene. MyoG gene expression in male longissimus muscles tended to be higher than that of females at all growth stages, except for the six month-old stage. IGF-I gene expression was significantly and positively correlated with live weight (P < 0.01) and carcass weight (0.01< P < 0.05), and was non-significantly positively correlated with net meat weight (P > 0.05). In contrast, MyoG gene expression was non-significantly and positively correlated with live weight, carcass, and net meat weight (P > 0.05). Carcass traits showed highly significant positive correlations (P < 0.01). Furthermore, expressions of IGF-I and MyoG genes showed highly significant positive correlations (P < 0.01). We conclude that the expressions of IGF-I and MyoG genes are significantly and positively correlated with early muscle traits of Hu sheep.


Assuntos
Fator de Crescimento Insulin-Like I/biossíntese , Carne , Miogenina/biossíntese , Carneiro Doméstico/genética , Animais , Peso Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento , Fator de Crescimento Insulin-Like I/genética , Músculo Esquelético/crescimento & desenvolvimento , Miogenina/genética , Fenótipo
12.
Am J Physiol Cell Physiol ; 304(2): C128-36, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23114963

RESUMO

Myogenic differentiation of skeletal muscle cells is characterized by a sequence of events that include activation of signal transducer and activator of transcription 3 (STAT3) and enhanced expression of its target gene Socs3. Autocrine effects of IL-6 may contribute to the activation of the STAT3-Socs3 cascade and thus to myogenic differentiation. The importance of IL-6 and STAT3 for the differentiation process was studied in C2C12 cells and in primary mouse wild-type and IL-6(-/-) skeletal muscle cells. In differentiating C2C12 myoblasts, the upregulation of IL-6 mRNA expression and protein secretion started after increased phosphorylation of STAT3 on tyrosine 705 and increased mRNA expression of Socs3 was observed. Knockdown of STAT3 and IL-6 mRNA in differentiating C2C12 myoblasts impaired the expression of the myogenic markers myogenin and MyHC IIb and subsequently myotube fusion. However, the knockdown of IL-6 did not prevent the induction of STAT3 tyrosine phosphorylation. The IL-6-independent activation of STAT3 was verified in differentiating primary IL-6(-/-) myoblasts. The phosphorylation of STAT3 and the expression levels of STAT3, Socs3, and myogenin during differentiation were comparable in the primary myoblasts independent of the genotype. However, IL-6(-/-) cells failed to induce MyHC IIb expression to the same level as in wild-type cells and showed reduced myotube formation. Supplementation of IL-6 could partially restore the fusion of IL-6(-/-) cells. These data demonstrate that IL-6 depletion during myogenic differentiation does not reduce the activation of the STAT3-Socs3 cascade, while IL-6 and STAT3 are both necessary to promote myotube fusion.


Assuntos
Diferenciação Celular , Interleucina-6/fisiologia , Desenvolvimento Muscular , Mioblastos Esqueléticos/citologia , Fator de Transcrição STAT3/metabolismo , Animais , Células Cultivadas , Técnicas de Silenciamento de Genes , Interleucina-6/genética , Camundongos , Camundongos Mutantes , Fibras Musculares Esqueléticas/metabolismo , Miogenina/biossíntese , Cadeias Pesadas de Miosina/biossíntese , Fosforilação , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Tirosina/metabolismo
13.
Biochim Biophys Acta ; 1819(11-12): 1208-16, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23092676

RESUMO

In the nucleus of differentiated osteoblasts, the DNA-binding αNAC protein acts as a transcriptional coactivator of the Osteocalcin gene. Chromatin immunoprecipitation-microarray assays (ChIP-chip) showed that αNAC binds the Osteocalcin promoter but also identified the Myogenin promoter as an αNAC target. Here, we confirm these array data using quantitative ChIP and further detected that αNAC binds to these promoters in myoblasts. Since these genes are differentially regulated during osteoblastogenesis or myogenesis, these results suggest cell- and promoter-context specific functions for αNAC. We hypothesized that αNAC dynamically recruits corepressors to inhibit Myogenin expression in cells committing to the osteoblastic lineage or to inhibit Osteocalcin transcription in differentiating myoblasts. Using co-immunoprecipitation assays, we detected complexes between αNAC and the corepressors HDAC1 and HDAC3, in myoblasts and osteoblasts. Sequential ChIP confirmed HDAC1 recruitment by αNAC at the Osteocalcin and Myogenin promoters. Interaction with the corepressors was detectable in pre-osteoblasts and in myoblasts but disappeared as the cells differentiate. Treatment with an HDAC inhibitor caused de-repression of Osteocalcin expression in myoblasts. Overexpression of αNAC in myoblasts inhibits expression of Myogenin and differentiation. However, overexpression of an N-terminus truncated αNAC mutant allowed myoblasts to express Myogenin and differentiate, and this mutant did not interact with HDAC1 or HDAC3. This study identified an additional DNA-binding target and novel protein-protein interactions for αNAC. We propose that αNAC plays a role in regulating gene transcription during mesenchymal cell differentiation by differentially recruiting corepressors at target promoters.


Assuntos
Regulação da Expressão Gênica/fisiologia , Histona Desacetilase 1/metabolismo , Histona Desacetilases/metabolismo , Chaperonas Moleculares/metabolismo , Mioblastos/metabolismo , Miogenina/biossíntese , Osteoblastos/metabolismo , Osteocalcina/biossíntese , Regiões Promotoras Genéticas/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Histona Desacetilase 1/genética , Histona Desacetilases/genética , Camundongos , Chaperonas Moleculares/genética , Mioblastos/citologia , Miogenina/genética , Osteoblastos/citologia , Osteocalcina/genética , Transcrição Gênica/fisiologia
14.
Histopathology ; 63(4): 545-50, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23944986

RESUMO

AIMS: Myogenin (myf4) is a nuclear transcription factor that is considered to be a sensitive and highly specific marker for skeletal muscle differentiation. Following the identification of focal strong nuclear staining with myogenin in two fibroepithelial polyps of the lower female genital tract (the index cases), we stained a series of vulvovaginal spindle cell lesions with this marker in order to investigate how widespread myogenin staining is in these lesions. METHODS AND RESULTS: Fibroepithelial polyps (n = 13), other vulvovaginal mesenchymal lesions (n = 21) and vulval or vaginal spindle cell squamous carcinomas (n = 4) were stained for myogenin. Apart from the index cases, all of the other cases were negative, except for one vaginal spindle cell squamous carcinoma, which showed focal weak nuclear immunoreactivity. Ten of 12 embryonal rhabdomyosarcomas of the lower female genital tract were myogenin-positive, as was a single vaginal rhabdomyoma. CONCLUSIONS: Our study illustrates that focal myogenin immunoreactivity occurs uncommonly in fibroepithelial polyps of the lower female genital tract. This may result in diagnostic confusion and misdiagnosis as a skeletal muscle neoplasm, especially the sarcoma botryoides variant of embryonal rhabdomyosarcoma.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células Escamosas/diagnóstico , Diagnóstico Diferencial , Miogenina/biossíntese , Pólipos/diagnóstico , Neoplasias Vaginais/diagnóstico , Adulto , Carcinoma de Células Escamosas/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Miogenina/análise , Pólipos/metabolismo , Rabdomiossarcoma/diagnóstico , Neoplasias Vaginais/metabolismo
15.
Artigo em Inglês | MEDLINE | ID: mdl-23319163

RESUMO

The hypothesis of this study was that 17ß-estradiol (estradiol) stimulates turkey skeletal muscle growth by influencing myogenic satellite cell proliferation, differentiation, and the gene expression of selected proteins important in regulating growth and development. Increasing levels of estradiol were administered in basal medium containing additional nutrients. Female-derived pectoralis major (PM) satellite cell proliferation was stimulated by estradiol at a level of 10(-9)M following 4days of treatment. Male PM and biceps femoris (BF) satellite cell proliferation was increased at 10(-12)M estradiol. Turkey embryonic myoblast proliferation, however, decreased with 10(-9)M and 10(-5)M estradiol following 3days under these conditions. Estradiol had no effect on the differentiation of any of the 4 groups of cells. Likewise, glypican-1 expression was unaffected by estradiol treatment. MyoD expression decreased in male PM but not BF cells. MyoD expression in female PM cells and embryonic myoblasts were also unaffected by estradiol administration. Estradiol decreased myogenin expression in male satellite cells, but had no effect on female cells. There was a slight decrease in myogenin expression in embryonic myoblasts. The results demonstrate a direct effect of estradiol on avian satellite cell proliferation independent of glypican-1, and decreased expression of MyoD and myogenin in some myogenic cells, coinciding with increased cellular proliferation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Estradiol/farmacologia , Glipicanas/biossíntese , Proteína MyoD/biossíntese , Miogenina/biossíntese , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Glipicanas/genética , Glipicanas/metabolismo , Masculino , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/genética , Proteína MyoD/genética , Proteína MyoD/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Músculos Peitorais/efeitos dos fármacos , Músculos Peitorais/crescimento & desenvolvimento , Músculos Peitorais/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Perus/genética , Perus/crescimento & desenvolvimento , Perus/metabolismo
16.
Genet Mol Res ; 12(4): 6733-42, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24391014

RESUMO

To confirm the entire developmental process and transition point of embryonic Pekin duck pectoral muscle, and to investigate the association between pectoral muscle development and their regulating genes, anatomical and morphological analyses of embryonic Pekin duck skeletal muscles were performed, and the expression patterns of its regulating genes were investigated. The anatomical analysis revealed that body weight increased with age, while increases in pectoral muscle weight nearly ceased after the embryo was 20 days of hatching (E20). The developmental morphological characteristics of Pekin duck pectoral muscle at the embryonic stage showed that E20 was the transition point (from proliferation to fusion) of Pekin duck pectoral muscle. The expression patterns of MRF4, MyoG, and MSTN indicated that E19 or E20 was the fastest point of pectoral muscle development and the crucial transition for Pekin duck pectoral muscle development during the embryonic stage. Together, these findings imply that E20 is the crucial transition point (from proliferation to fusion) of Pekin duck pectoral muscle and that there is no muscle fiber hypertrophy after E20. Results of this study provide further understanding of the developmental process and transition point of Pekin duck pectoral muscle during the embryo stage.


Assuntos
Patos/embriologia , Perfilação da Expressão Gênica/veterinária , Músculos Peitorais/embriologia , Animais , Peso Corporal , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Fatores de Regulação Miogênica/biossíntese , Fatores de Regulação Miogênica/genética , Miogenina/biossíntese , Miogenina/genética , Miostatina/biossíntese , Miostatina/genética , Músculos Peitorais/anatomia & histologia , Músculos Peitorais/crescimento & desenvolvimento , RNA Mensageiro/biossíntese
17.
J Biol Chem ; 286(26): 23498-510, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21543328

RESUMO

In adult muscles and under normal physiological conditions, satellite cells are found in a quiescent state but can be induced to enter the cell cycle by signals resulting from exercise, injury-induced muscle regeneration, or specific disease states. Once activated, satellite cells proliferate, self-renew, and differentiate to form myofibers. In the present study, we found that the zinc finger-containing factor Teashirt-3 (TSHZ3) was expressed in quiescent satellite cells of adult mouse skeletal muscles. We showed that following treatment with cardiotoxin TSHZ3 was strongly expressed in satellite cells of regenerating muscles. Moreover, immunohistochemical analysis indicated that TSHZ3 was expressed in both quiescent and activated satellite cells on intact myofibers in culture. TSHZ3 expression was maintained in myoblasts but disappeared with myotube formation. In C2C12 myoblasts, we showed that overexpression of Tshz3 impaired myogenic differentiation and promoted the down-regulation of myogenin (Myog) and up-regulation of paired-box factor 7 (Pax7). Moreover, knockdown experiments revealed a selective effect of Tshz3 on Myog regulation, and transcriptional reporter experiments indicated that TSHZ3 repressed Myog promoter. We identified the BRG1-associated factor 57 (BAF57), a subunit of the SWI/SNF complex, as a partner of TSHZ3. We showed that TSHZ3 cooperated with BAF57 to repress MYOD-dependent Myog expression. These results suggest a novel mechanism for transcriptional repression by TSHZ3 in which TSHZ3 and BAF57 cooperate to modulate MyoD activity on the Myog promoter to regulate skeletal muscle differentiation.


Assuntos
Diferenciação Celular/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Regulação da Expressão Gênica/fisiologia , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/metabolismo , Miogenina/biossíntese , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Cardiotoxinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Desenvolvimento Muscular/efeitos dos fármacos , Músculo Esquelético/citologia , Miogenina/genética , Fator de Transcrição PAX7/genética , Fator de Transcrição PAX7/metabolismo , Regiões Promotoras Genéticas/fisiologia , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Proteínas Repressoras/genética , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Fatores de Transcrição/genética
18.
J Cell Biochem ; 113(1): 209-19, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21898542

RESUMO

Differentiation of skeletal muscle myoblasts involves activation of muscle-specific markers such as MyoD, Myf5, MRF4, and myogenin, followed by exit from the cell cycle, expression of structural proteins, and fusion into multinucleated myotubes. Cyclin D3 is upregulated during muscle differentiation, and expression of cyclin D3 in proliferating myoblasts causes early activation of myogenesis. In this study, we have identified the genes activated by cyclin D3 expression in C2C12 myoblasts and differentiated cells by real-time PCR analysis. Cyclin D3 expression induced faster differentiation kinetics and increase in levels of myogenic genes such as MyoD, Myf5, and myogenin at an early stage during the differentiation process, although long-term myogenic differentiation was not affected. Transcript levels of the transcription factor Pax7 that is expressed in muscle progenitors were enhanced by cyclin D3 expression in myoblasts. Components of a histone methyltransferase complex recruited by Pax7 to myogenic gene promoters were also regulated by cyclin D3. Further, the Pax7 promoter was upregulated in myoblasts expressing cyclin D3. Myoblasts that expressed cyclin D3 showed moderately higher levels of the cyclin-dependent kinase inhibitor p21 and were stalled in G2/M phase of the cell cycle. Our findings suggest that cyclin D3 primes myoblasts for differentiation by enhancing muscle specific gene expression and cell cycle exit.


Assuntos
Pontos de Checagem do Ciclo Celular , Diferenciação Celular/genética , Ciclina D3/metabolismo , Fator de Transcrição PAX7/metabolismo , Animais , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/metabolismo , Camundongos , Células Musculares/fisiologia , Desenvolvimento Muscular/genética , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/metabolismo , Miogenina/biossíntese , Fator de Transcrição PAX7/genética , Regiões Promotoras Genéticas
19.
Pharmacol Res ; 65(2): 221-30, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22178654

RESUMO

Anabolic/androgenic steroids (AAS) are drugs that enhance muscle mass, and are often illegally utilized in athletes to improve their performances. Recent data suggest that the increased risk for amyotrophic lateral sclerosis (ALS) in male soccer and football players could be linked to AAS abuse. ALS is a motor neuron disease mainly occurring in sporadic (sALS) forms, but some familial forms (fALS) exist and have been linked to mutations in different genes. Some of these, in their wild type (wt) form, have been proposed as risk factors for sALS, i.e. superoxide dismutase 1 (SOD1) gene, whose mutations are causative of about 20% of fALS. Notably, SOD1 toxicity might occur both in motor neurons and in muscle cells. Using gastrocnemius muscles of mice overexpressing human mutant SOD1 (mutSOD1) at different disease stages, we found that the expression of a selected set of genes associated to muscle atrophy, MyoD, myogenin, atrogin-1, and transforming growth factor (TGF)ß1, is up-regulated already at the presymptomatic stage. Atrogin-1 gene expression was increased also in mice overexpressing human wtSOD1. Similar alterations were found in axotomized mouse muscles and in cultured ALS myoblast models. In these ALS models, we then evaluated the pharmacological effects of the synthetic AAS nandrolone on the expression of the genes modified in ALS muscle. Nandrolone administration had no effects on MyoD, myogenin, and atrogin-1 expression, but it significantly increased TGFß1 expression at disease onset. Altogether, these data suggest that, in fALS, muscle gene expression is altered at early stages, and AAS may exacerbate some of the alterations induced by SOD1 possibly acting as a contributing factor also in sALS.


Assuntos
Esclerose Lateral Amiotrófica/genética , Expressão Gênica/efeitos dos fármacos , Mutação , Nandrolona/farmacologia , Superóxido Dismutase/genética , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/metabolismo , Anabolizantes/farmacologia , Androgênios/farmacologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Proteína MyoD/biossíntese , Proteína MyoD/genética , Proteína MyoD/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Miogenina/biossíntese , Miogenina/genética , Miogenina/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Superóxido Dismutase/biossíntese , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
Exp Cell Res ; 316(17): 2932-44, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20696153

RESUMO

Muscle satellite cells are essential for muscle growth and regeneration and their morphology, behavior and gene expression have been extensively studied. However, the mechanisms involved in their proliferation and differentiation remain elusive. Six1 and Six4 proteins were expressed in the nuclei of myofibers of adult mice and the numbers of myoblasts positive for Six1 and Six4 increased during regeneration of skeletal muscles. Six1 and Six4 were expressed in quiescent, activated and differentiated muscle satellite cells isolated from adult skeletal muscle. Overexpression of Six4 and Six5 repressed the proliferation and differentiation of satellite cells. Conversely, knockdown of Six5 resulted in augmented proliferation, and that of Six4 inhibited differentiation. Muscle satellite cells isolated from Six4(+/-)Six5(-/-) mice proliferated to higher cell density though their differentiation was not altered. Meanwhile, overproduction of Six1 repressed proliferation and promoted differentiation of satellite cells. In addition, Six4 and Six5 repressed, while Six1 activated myogenin expression, suggesting that the differential regulation of myogenin expression is responsible for the differential effects of Six genes. The results indicated the involvement of Six genes in the behavior of satellite cells and identified Six genes as potential target for manipulation of proliferation and differentiation of muscle satellite cells for therapeutic applications.


Assuntos
Diferenciação Celular/genética , Proliferação de Células , Proteínas de Homeodomínio/genética , Células Satélites de Músculo Esquelético/citologia , Transativadores/genética , Adulto , Animais , Regulação da Expressão Gênica/fisiologia , Proteínas de Homeodomínio/fisiologia , Humanos , Camundongos , Camundongos Knockout , Células Musculares/citologia , Miogenina/biossíntese , Transativadores/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA