Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
FASEB J ; 33(8): 9638-9655, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31145867

RESUMO

Here, we performed whole-genome bisulfite sequencing of longissimus dorsi muscle from Landrace and Wuzhishan (WZS) miniature pigs during 18, 21, and 28 d postcoitum. It was uncovered that in regulatory regions only around transcription start sites (TSSs), gene expression and methylation showed negative correlation, whereas in gene bodies, positive correlation occurred. Furthermore, earlier myogenic gene demethylation around TSSs and earlier hypermethylation of myogenic genes in gene bodies were considered to trigger their earlier expression in miniature pigs. Furthermore, by analyzing the methylation pattern of the myogenic differentiation 1(MyoD) promoter and distal enhancer, we found that earlier demethylation of the MyoD distal enhancer in WZSs contributes to its earlier expression. Moreover, DNA demethylase Tet1 was found to be involved in the demethylation of the myogenin promoter and promoted immortalized mouse myoblast cell line (C2C12) and porcine embryonic myogenic cell differentiation. This study reveals that earlier demethylation of myogenic genes contributes to precocious terminal differentiation of myoblasts in miniature pigs.-Zhang, X., Nie, Y., Cai, S., Ding, S., Fu, B., Wei, H., Chen, L., Liu, X., Liu, M., Yuan, R., Qiu, B., He, Z., Cong, P., Chen, Y., Mo, D. Earlier demethylation of myogenic genes contributes to embryonic precocious terminal differentiation of myoblasts in miniature pigs.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Muscular/fisiologia , Mioblastos/citologia , Mioblastos/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Biologia Computacional , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Desmetilação , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Desenvolvimento Muscular/genética , Proteína MyoD/genética , Proteína MyoD/metabolismo , Regiões Promotoras Genéticas/genética , Suínos , Porco Miniatura
2.
BMC Genet ; 20(1): 72, 2019 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-31477002

RESUMO

BACKGROUND: Myogenic Differentiation 1 (MyoD) is a crucial master switch in regulating muscle-specific gene transcription. Forced expression of myoD is equipped to induce several cell lineages into myoblast, which then differentiate and fuse into myotube. Pig is one of the most significant livestock supplying meat, and has been classified into lean, fat and miniature pig breeds. However, the mechanisms underlying muscle mass variation among different pig breeds have remained unclear. Considering the important effect of MyoD on muscle development, it remains to be investigated whether the difference in muscle mass is caused by its single nucleotide polymorphisms (SNPs) which are the major differences among pig breeds at DNA level. RESULTS: In this study, we identified the locations of porcine myoD regulatory regions including proximal regulatory region (PRR), distal regulatory region (DRR), and core enhancer (CE) region. There are 8 SNPs in the regulatory regions and 6 SNPs in gene body region, which were identified from lean, fat and miniature pig populations. However, these SNPs have no effects on its temporal expression and transcriptional activity which might lead to the distinction in postnatal muscle mass. In addition, overexpression of myoD clones across from amphibious to mammals including xenopus tropicalis, chicken, mouse and pig whose gene identities vary from 68 to 84%, could promote myogenesis in NIH3T3 fibroblasts cells. CONCLUSIONS: These results proved that myoD nucleotide variations from different pig populations have no effect on muscle mass, suggesting that the function of myoD is highly conserved not only among different pig breeds, but also across different species. Thus, it would be futile to discover SNPs affecting muscle mass in pig populations with normal muscle development.


Assuntos
Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Proteína MyoD/genética , Polimorfismo de Nucleotídeo Único , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Mutação , Sequências Reguladoras de Ácido Nucleico , Suínos
3.
J Cell Sci ; 129(22): 4305-4316, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27672022

RESUMO

Although the mechanism underlying modulation of transcription factors in myogenesis has been well elucidated, the function of the transcription cofactors involved in this process remains poorly understood. Here, we identified HMGB2 as an essential nuclear transcriptional co-regulator in myogenesis. HMGB2 was highly expressed in undifferentiated myoblasts and regenerating muscle. Knockdown of HMGB2 inhibited myoblast proliferation and stimulated its differentiation. HMGB2 depletion downregulated Myf5 and cyclin A2 at the protein but not mRNA level. In contrast, overexpression of HMGB2 promoted Myf5 and cyclin A2 protein upregulation. Furthermore, we found that the RNA-binding protein IGF2BP2 is a downstream target of HMGB2, as previously shown for HMGA2. IGF2BP2 binds to mRNAs of Myf5 or cyclin A2, resulting in translation enhancement or mRNA stabilization, respectively. Notably, overexpression of IGF2BP2 could partially rescue protein levels of Myf5 and cyclin A2, in response to HMGB2 decrease. Moreover, depletion of HMGB2 in vivo severely attenuated muscle repair; this was due to a decrease in satellite cells. Taken together, these results highlight the previously undiscovered and crucial role of the HMGB2-IGF2BP2 axis in myogenesis and muscle regeneration.


Assuntos
Proteína HMGB2/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Ligação a RNA/metabolismo , Regeneração , Células Satélites de Músculo Esquelético/metabolismo , Animais , Animais Recém-Nascidos , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Linhagem Celular , Núcleo Celular/metabolismo , Proliferação de Células , Ciclina A2/genética , Ciclina A2/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Biochem Biophys Res Commun ; 503(2): 970-976, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29932923

RESUMO

MicroRNAs are a class of highly conserved ∼20 nucleotides non-coding RNAs that post-transcriptionally regulate gene expression. Many miRNAs were studied in the development of skeletal muscle, such as miR-1, miR-206, and miR-133. In our previous study, miR-127-3p was found highly expressed in porcine fetal skeletal muscle, whereas the detailed functions of miR-127-3p in muscle development is still unclear. In this study, we detected that miR-127-3p also highly expressed in skeletal muscle, cardiac muscle of adult mice and proliferative C2C12 cell lines. Overexpression of miR-127-3p almost has no effects on differentiation of C2C12 cell lines. However, miR-127-3p significantly inhibited the cell proliferation of C2C12 cells. Moreover, we identified KMT5a as a target gene that was down-regulated in both mRNA and protein level when miR-127-3p mimics were introduced. Furthermore, KMT5a overexpression in miR-127-3p treated cells rescued the influence of miR-127-3p on C2C12 proliferation. In brief, our data reveals that miR-127-3p regulates the proliferation of myocytes through KMT5a.


Assuntos
Proliferação de Células , Regulação para Baixo , Histona-Lisina N-Metiltransferase/genética , MicroRNAs/genética , Células Musculares/citologia , Animais , Linhagem Celular , Células HEK293 , Humanos , Camundongos , Células Musculares/metabolismo , Regulação para Cima
5.
Biochim Biophys Acta ; 1859(11): 1459-1469, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27452504

RESUMO

Activating transcription factor 4 (ATF4), which is highly expressed in 3T3-L1 adipocytes after adipogenic induction, is essential for adipocytes differentiation. ATF4 also plays a vital role in regulating fatty acids biosynthesis, whereas the detailed mechanism of this process is still unclear. Here we demonstrated that siRNA-based ATF4 depletion in 3T3-L1 adipocytes significantly reduced the accumulation of fatty acids and triglycerides. Moreover, SREBP1c protein, which is an important transcription factor of lipogenesis, appreciably decreased while Srebp1c mRNA increased. Then we identified that ATF4 could maintain SREBP1c protein stability by directly activating the expression of USP7 which deubiquitinates SREBP1c and increases its protein content in cell. Besides, USP7 could restore the synthesis of fatty acids and triglycerides in the absence of ATF4. On the other hand, we found that ATF4 might inhibit the transcription of Srebp1c through TRB3, which is repressed by IBMX and DEX during early adipogenesis. Thus, our data indicate that ATF4 regulates SREBP1c expression to control fatty acids synthesis.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Adipócitos/citologia , Diferenciação Celular , Ácidos Graxos/biossíntese , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Células 3T3-L1 , Animais , Camundongos , Transcrição Gênica/fisiologia , Peptidase 7 Específica de Ubiquitina , Proteases Específicas de Ubiquitina/metabolismo
6.
BMC Dev Biol ; 15: 42, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26559169

RESUMO

BACKGROUND: The investigation of skeletal muscle development is of importance in stock farming and biomedicine. It is still ambiguous that whether animals are born with the full set of skeletal muscle fibers or if the number of myofibers continues to increase postnatally. RESULTS: Here, an inducible lineage-tracing system was employed to monitor the changes of myofiber number in various skeletal muscles during development. We confirm that the total myofiber number of longissimus dorsi, gastrocnemius and rectus femoris is determined prenatally. However, tibialis anterior and extensor digitorum longus have a different development pattern, and their myofiber number still increases in the first postnatal week and then remains stable afterwards. CONCLUSIONS: Our results highlight different development time frames of anatomically distinct skeletal muscles.


Assuntos
Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/citologia , Animais , Doxiciclina , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Músculo Quadríceps/crescimento & desenvolvimento , Coloração e Rotulagem , beta-Galactosidase/genética
7.
Cell Death Differ ; 27(5): 1644-1659, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31685980

RESUMO

Zinc finger protein 422 (Zfp422) is a widely expressed zinc finger protein that serves as a transcriptional factor to regulate downstream gene expression, but until now, little is known about its roles in myogenesis. We found here that Zfp422 plays a critical role in skeletal muscle development and regeneration. It highly expresses in mouse skeletal muscle during embryonic development. Specific knockout of Zfp422 in skeletal muscle impaired embryonic muscle formation. Satellite cell-specific Zfp422 deletion severely inhibited muscle regeneration. Myoblast differentiation and myotube formation were suppressed in Zfp422-deleted C2C12 cells, isolated primary myoblasts, and satellite cells. Chromatin Immunoprecipitation Sequencing (ChIP-Seq) revealed that Zfp422 regulated ephrin type-A receptor 7 (EphA7) expression by binding an upstream 169-bp DNA sequence, which was proved to be an enhancer of EphA7. Knocking EphA7 down in C2C12 cells or deleting Zfp422 in myoblasts will inhibit cell apoptosis which is required for myoblast differentiation. These results indicate that Zfp422 is essential for skeletal muscle differentiation and fusion, through regulating EphA7 expression to maintain proper apoptosis.


Assuntos
Apoptose , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Músculo Esquelético/citologia , Mioblastos/citologia , Mioblastos/metabolismo , Proteínas Nucleares/metabolismo , Receptor EphA7/metabolismo , Animais , Apoptose/genética , Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular/genética , Músculo Esquelético/embriologia , Proteínas Nucleares/genética , Regeneração , Células Satélites de Músculo Esquelético/citologia
8.
Cell Prolif ; 53(2): e12744, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31840352

RESUMO

OBJECTIVES: Mixed lineage leukaemia protein-1 (MLL1) mediates histone 3 lysine 4 (H3K4) trimethylation (me3) and plays vital roles during early embryonic development and hematopoiesis. In our previous study, we found its expression was positively correlated with embryonic myogenic ability in pigs, indicating its potential roles in mammalian muscle development. The present work aimed to explore the roles and regulation mechanisms of MLL1 in myogenesis. MATERIALS AND METHODS: The expression of MLL1 in C2C12 cells was experimentally manipulated using small interfering RNAs (siRNA). 5-ethynyl-2'-deoxyuridine (EdU) assay, cell cycle assay, immunofluorescence, qRT-PCR and Western blot were performed to assess myoblast proliferation and differentiation. Chromatin immunoprecipitation assay was conducted to detect H3K4me3 enrichment on myogenic factor 5 (Myf5) promoter. A cardiotoxin (CTX)-mediated muscle regeneration model was used to investigate the effects of MLL1 on myogenesis in vivo. RESULTS: MLL1 was highly expressed in proliferating C2C12 cells, and expression decreased after differentiation. Knocking down MLL1 suppressed myoblast proliferation and impaired myoblast differentiation. Furthermore, knockdown of MLL1 resulted in the arrest of cell cycle in G1 phase, with decreased expressions of Myf5 and Cyclin D1. Mechanically, MLL1 transcriptionally regulated Myf5 by mediating H3K4me3 on its promoter. In vivo data implied that MLL1 was required for Pax7-positive satellite cell proliferation and muscle repair. CONCLUSION: MLL1 facilitates proliferation of myoblasts and Pax7-positive satellite cells by epigenetically regulating Myf5 via mediating H3K4me3 on its promoter.


Assuntos
Epigênese Genética/genética , Histona-Lisina N-Metiltransferase/genética , Desenvolvimento Muscular/genética , Proteína de Leucina Linfoide-Mieloide/genética , Fator Regulador Miogênico 5/genética , Animais , Pontos de Checagem do Ciclo Celular/genética , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/genética , Feminino , Fase G1/genética , Histonas/genética , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos/fisiologia , Fator de Transcrição PAX7/genética , Regiões Promotoras Genéticas/genética , Células Satélites de Músculo Esquelético/fisiologia
9.
J Anim Sci ; 97(5): 1967-1978, 2019 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-31222274

RESUMO

Pig is one of the major dietary protein sources for human consumption, from which muscle is the largest protein origin. However, molecular mechanisms concerning early porcine embryonic muscle development distinctions between pig breeds are still unclear. In this study, an integrated analysis of transcriptome and miRNAome was conducted using longissimus dorsi muscle of 4 early embryonic stages around the primary myofiber formation time (18-, 21-, 28-, and 35-d post coitus) from 2 pig breeds (Landrace [LR] and Wuzhishan [WZS]) differing in meat mass. The global miRNA/mRNA expression profile showed that WZS prepared for myogenic developmental processes earlier than LR. After identifying and analyzing the interaction network of top 100 up-/down-regulated miRNA and their target genes, we were able to find 3 gene clusters: chromatin modification-related (Chd2, H3f3a, Chd6, and Mll1), myogenesis-related (Pax3, Pbx1, Mef2a, and Znf423), and myosin component-related (Mylk, Myo5a, Mylk4, Myh9, and Mylk2) gene clusters. These genes may involve in miRNA-gene myogenic regulatory network that plays vital role in regulating distinct early porcine embryonic myogenic processes between LR and WZS. In summary, our study reveals an epigenetic-mediated myogenic regulatory axial that will help us to decipher molecular mechanisms concerning early porcine embryonic muscle development distinctions between pig breeds.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento/genética , MicroRNAs/genética , RNA Mensageiro/genética , Suínos/genética , Transcriptoma , Animais , Desenvolvimento Embrionário/genética , Feminino , Perfilação da Expressão Gênica/veterinária , Redes Reguladoras de Genes , Masculino , Desenvolvimento Muscular/genética , Especificidade da Espécie , Suínos/embriologia , Suínos/crescimento & desenvolvimento
10.
Sci Rep ; 7: 41608, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28148961

RESUMO

Differentiation of myoblasts is essential in the development and regeneration of skeletal muscles to form multinucleated, contractile muscle fibers. However, the process of myoblast differentiation in mammals is complicated and requires to be further investigated. In this study, we found Palmdelphin (Palmd), a cytosolic protein, promotes myoblast differentiation. Palmd is predominantly expressed in the cytosol of myoblasts and is gradually up-regulated after differentiation. Knockdown of Palmd by small interfering RNA (siRNA) in C2C12 markedly inhibits myogenic differentiation, suggesting a specific role of Palmd in the morphological changes of myoblast differentiation program. Overexpression of Palmd in C2C12 enhances myogenic differentiation. Remarkably, inhibition of Palmd results in impaired myotube formation during muscle regeneration after injury. These findings reveal a new cytosolic protein that promotes mammalian myoblast differentiation and provide new insights into the molecular regulation of muscle formation.


Assuntos
Diferenciação Celular/genética , Proteínas de Membrana/genética , Desenvolvimento Muscular/genética , Mioblastos/citologia , Mioblastos/metabolismo , Regeneração , Animais , Linhagem Celular , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Proteínas de Membrana/metabolismo , Camundongos , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA