Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 300(2): 105602, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38159852

RESUMO

In humans, skeletal muscles comprise nearly 40% of total body mass, which is maintained throughout adulthood by a balance of muscle protein synthesis and breakdown. Cellular amino acid (AA) levels are critical for these processes, and mammalian cells contain transporter proteins that import AAs to maintain homeostasis. Until recently, the control of transporter regulation has largely been studied at the transcriptional and posttranslational levels. However, here, we report that the RNA-binding protein YBX3 is critical to sustain intracellular AAs in mouse skeletal muscle cells, which aligns with our recent findings in human cells. We find that YBX3 directly binds the solute carrier (SLC)1A5 AA transporter messenger (m)RNA to posttranscriptionally control SLC1A5 expression during skeletal muscle cell differentiation. YBX3 regulation of SLC1A5 requires the 3' UTR. Additionally, intracellular AAs transported by SLC1A5, either directly or indirectly through coupling to other transporters, are specifically reduced when YBX3 is depleted. Further, we find that reduction of the YBX3 protein reduces proliferation and impairs differentiation in skeletal muscle cells, and that YBX3 and SLC1A5 protein expression increase substantially during skeletal muscle differentiation, independently of their respective mRNA levels. Taken together, our findings suggest that YBX3 regulates AA transport in skeletal muscle cells, and that its expression is critical to maintain skeletal muscle cell proliferation and differentiation.


Assuntos
Sistema ASC de Transporte de Aminoácidos , Fibras Musculares Esqueléticas , Proteínas de Ligação a RNA , Animais , Humanos , Camundongos , Sistema ASC de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Regulação da Expressão Gênica/genética , Células NIH 3T3 , Células HCT116 , Proliferação de Células/genética , Diferenciação Celular/genética
2.
J Proteome Res ; 23(8): 3444-3459, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39024330

RESUMO

Ferroptosis adversely affects the viability, differentiation, and metabolic integrity of C2C12 myoblasts, contributing to the decline in skeletal muscle health. The intricate mechanisms behind this process are not fully understood. In this study, we induced ferroptosis in myoblasts using targeted inducers and found a marked decrease in specific redox metabolites, particularly taurine. Taurine supplementation effectively reversed the deleterious effects of ferroptosis, significantly increased cellular glutathione levels, reduced MDA and ROS levels, and rejuvenated impaired myogenic differentiation. Furthermore, taurine downregulated HO-1 expression and decreased intracellular Fe2+ levels, thereby stabilizing the labile iron pool. Using NMR metabolomic analysis, we observed that taurine profoundly promoted glycerophospholipid metabolism, which is critical for cell membrane repair, and enhanced mitochondrial bioenergetics, thereby increasing the energy reserves essential for muscle satellite cell regeneration. These results suggest that taurine is a potent ferroptosis inhibitor that attenuates key drivers of this process, strengthens oxidative defenses, and improves redox homeostasis. This combined effect protects cells from ferroptosis-induced damage. This study highlights the potential of taurine as a valuable ferroptosis inhibitor that protects skeletal muscle from ferroptosis-induced damage and provides a basis for therapeutic strategies to rejuvenate and facilitate the regeneration of aging skeletal muscle.


Assuntos
Ferroptose , Homeostase , Ferro , Mioblastos , Oxirredução , Taurina , Taurina/farmacologia , Ferroptose/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Mioblastos/citologia , Ferro/metabolismo , Animais , Camundongos , Homeostase/efeitos dos fármacos , Linhagem Celular , Espécies Reativas de Oxigênio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Glutationa/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Glicerofosfolipídeos/metabolismo
3.
Anim Biotechnol ; 34(7): 2514-2526, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35875894

RESUMO

MiR-424-5p was found to be a potential regulator in the proliferation, migration, and invasion of various cancer cells. However, the effects and functional mechanism of miR-424-5p in the process of myogenesis are still unclear. Previously, using microRNA (miRNA) sequencing and expression analysis, we discovered that miR-424-5p was expressed differentially in the different skeletal muscle growth periods of Xuhuai goats. We hypothesized that miR-424-5p might play an important role in skeletal muscle myogenesis. Then, we found that the proliferation ability of the mouse myoblast cell (C2C12 myoblast cell line) was significantly augmented, whereas the C2C12 differentiation was repressed after increasing the expression of miR-424-5p. Mechanistically, HSP90AA1 presented a close interrelation with miR-424-5p, which was predicted as a target gene in the progression of skeletal muscle myogenesis, using transcriptome sequencing, dual luciferase reporter gene detection, and qRT-PCR. The silencing of HSP90AA1 obviously increased C2C12 proliferation and diminished differentiation, which is consistent with the ability of miR-424-5p in C2C12. Altogether, our findings indicated the role of miR-424-5p as a novel potential regulator via HSP90AA1 during muscle myogenesis progression.


Assuntos
MicroRNAs , Animais , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proliferação de Células/genética , Diferenciação Celular/genética , Linhagem Celular , Desenvolvimento Muscular/genética , Cabras/genética , Músculo Esquelético/metabolismo
4.
Int J Mol Sci ; 23(22)2022 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-36430479

RESUMO

Lactate is a general compound fuel serving as the fulcrum of metabolism, which is produced from glycolysis and shuttles between different cells, tissues and organs. Lactate is usually accumulated abundantly in muscles during exercise. It remains unclear whether lactate plays an important role in the metabolism of muscle cells. In this research, we assessed the effects of lactate on myoblasts and clarified the underlying metabolic mechanisms through NMR-based metabonomic profiling. Lactate treatment promoted the proliferation and differentiation of myoblasts, as indicated by significantly enhanced expression levels of the proteins related to cellular proliferation and differentiation, including p-AKT, p-ERK, MyoD and myogenin. Moreover, lactate treatment profoundly regulated metabolisms in myoblasts by promoting the intake and intracellular utilization of lactate, activating the TCA cycle, and thereby increasing energy production. For the first time, we found that lactate treatment evidently promotes AMPK signaling as reflected by the elevated expression levels of p-AMPK and p-ACC. Our results showed that lactate as a metabolic regulator activates AMPK, remodeling the cellular metabolic profile, and thereby promoting the proliferation and differentiation of myoblasts. This study elucidates molecular mechanisms underlying the effects of lactate on skeletal muscle in vitro and may be of benefit to the exploration of lactate acting as a metabolic regulator.


Assuntos
Proteínas Quinases Ativadas por AMP , Ácido Láctico , Mioblastos , Proliferação de Células , Músculo Esquelético , Metaboloma
5.
Int J Mol Sci ; 23(19)2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-36232401

RESUMO

The sphingosine 1-phosphate (S1P) and endocannabinoid (ECS) systems comprehend bioactive lipids widely involved in the regulation of similar biological processes. Interactions between S1P and ECS have not been so far investigated in skeletal muscle, where both systems are active. Here, we used murine C2C12 myoblasts to investigate the effects of S1P on ECS elements by qRT-PCR, Western blotting and UHPLC-MS. In addition, the modulation of the mitochondrial membrane potential (ΔΨm), by JC-1 and Mitotracker Red CMX-Ros fluorescent dyes, as well as levels of protein controlling mitochondrial function, along with the oxygen consumption were assessed, by Western blotting and respirometry, respectively, after cell treatment with methanandamide (mAEA) and in the presence of S1P or antagonists to endocannabinoid-binding receptors. S1P induced a significant increase in TRPV1 expression both at mRNA and protein level, while it reduced the protein content of CB2. A dose-dependent effect of mAEA on ΔΨm, mediated by TRPV1, was evidenced; in particular, low doses were responsible for increased ΔΨm, whereas a high dose negatively modulated ΔΨm and cell survival. Moreover, mAEA-induced hyperpolarization was counteracted by S1P. These findings open new dimension to S1P and endocannabinoids cross-talk in skeletal muscle, identifying TRPV1 as a pivotal target.


Assuntos
Endocanabinoides , Corantes Fluorescentes , Animais , Ácidos Araquidônicos , Linhagem Celular , Endocanabinoides/metabolismo , Endocanabinoides/farmacologia , Corantes Fluorescentes/metabolismo , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/farmacologia , Camundongos , Mitocôndrias/metabolismo , Mioblastos/metabolismo , Alcamidas Poli-Insaturadas , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacologia , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
6.
J Cell Biochem ; 122(3-4): 442-455, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33399227

RESUMO

As a posttranscriptional regulatory factor, microRNA (miRNA) plays an important role in the formation of myotubes. However, little is known about the mechanism of miRNA regulating myotube morphogenesis. Here, we aimed to characterize the function of miR-455-5p in myotube morphogenesis by inducing differentiation in C2C12 myoblasts containing murine Mylip fragments with the miR-455-5p target sequence. We found that miR-455-5p overexpression promoted the differentiation and hypertrophy of myotubes, while miR-455-5p inhibition led to the failure of myotube differentiation and formation of short myotubes. Furthermore, we demonstrated that miR-455-5p directly targeted the Mylip 3'-untranslated region, which plays a key role in monitoring myotube morphogenesis. Interestingly, the expression and function of Mylip were opposite to those of miR-455-5p during myogenesis. Our data uncovered novel miR-455-5p targets and established a functional link between Mylip and myotube morphogenesis. Understanding the involvement of Mylip in myotube morphogenesis provides insight into the function of the gene regulatory network.


Assuntos
Diferenciação Celular/fisiologia , MicroRNAs/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Humanos , MicroRNAs/genética , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia
7.
J Biol Chem ; 294(39): 14279-14288, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31387946

RESUMO

Skeletal muscle myoblast differentiation involves elaborate signaling networks, including the activity of various ion channels and transporters. Several K+ and Ca2+ channels have been shown to affect myogenesis, but little is known about roles of Cl- channels in the associated processes. Here, we report that the leucine-rich repeat containing family 8 (LRRC8)/volume-regulated anion channel (VRAC) promotes mouse myoblast differentiation. All LRRC8 subunits of heteromeric VRAC were expressed during myotube formation of murine C2C12 myoblasts. Pharmacological VRAC inhibitors, siRNA-mediated knockdown of the essential VRAC subunit LRRC8A, or VRAC activity-suppressing overexpression of LRRC8A effectively reduced the expression of the myogenic transcription factor myogenin and suppressed myoblast fusion while not affecting myoblast proliferation. We found that inhibiting VRAC impairs plasma membrane hyperpolarization early during differentiation. At later times (more than 6 h after inducing differentiation), VRAC inhibition no longer suppressed myoblast differentiation, suggesting that VRAC acts upstream of K+ channel activation. Consequently, VRAC inhibition prevented the increase of intracellular steady-state Ca2+ levels that normally occurs during myogenesis. Our results may explain the mechanism for the thinning of skeletal muscle bundles observed in LRRC8A-deficient mice and highlight the importance of the LRRC8/VRAC anion channel in cell differentiation.


Assuntos
Diferenciação Celular , Potenciais da Membrana , Proteínas de Membrana/metabolismo , Mioblastos/metabolismo , Animais , Linhagem Celular , Proteínas de Membrana/genética , Camundongos , Mioblastos/citologia , Mioblastos/fisiologia , Miogenina/genética , Miogenina/metabolismo
8.
J Cell Physiol ; 235(7-8): 5702-5710, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31975415

RESUMO

Moderate exercise improves glycometabolic disorder and type 2 diabetes mellitus in menopausal females. So far, the effect of exercise-induced estrogen on muscular glycometabolism is not well defined. The current study was designed to explore the effect of mechanical stretch-induced estrogen on glycometabolism in mouse C2 C12 myoblasts. The mouse C2 C12 myoblasts in vitro were assigned randomly to the control (C), stretch (S), and stretch plus aromatase inhibitor anastrozole (SA) groups. Cells in the S group were stretched by the Flexcell FX-5000™ system (15% magnitude, 1 Hz frequency, and 6-hr duration) whereas those in the SA group were treated with 400 µg/ml anastrozole before the same stretching. Glucose uptake, estradiol levels, PFK-1 levels, and oxygen consumption rate were determined, and the expression of HK, PI3K, p-AKT, AKT, and GLUT4 proteins were semiquantified with western blot analysis. Compared to the control, the estradiol level, oxygen consumption rate, expression of HK, PI3K, and PFK-1 proteins, the ratio of p-AKT to AKT, and the ratio of GLUT4 in the cell membrane to that in the whole cell were higher in the S group. On the other hand, the estradiol level, glucose uptake, expression of PFK-1 and GLUT4 proteins, oxygen consumption rate, expression of HK protein, and the ratio of p-AKT/AKT were lower in the myoblasts in the SA group than those in the S group. The level of estradiol was positively correlated with glucose uptake (p < .01, r = .818). Therefore, mechanical stretch-induced estrogen increased the expression of glycometabolism-related enzymes and proteins in the mouse C2 C12 myoblasts.


Assuntos
Diabetes Mellitus Tipo 2/genética , Transportador de Glucose Tipo 4/genética , Estresse Mecânico , Anastrozol/farmacologia , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Estrogênios/genética , Estrogênios/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/genética , Glucose/metabolismo , Humanos , Insulina/metabolismo , Resistência à Insulina/genética , Camundongos , Mioblastos/metabolismo , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/efeitos dos fármacos
9.
J Cell Physiol ; 235(4): 3508-3518, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31549411

RESUMO

Muscle loss is a major problem for many in lifetime. Muscle and bone degeneration has also been observed in individuals exposed to microgravity and in unloading conditions. C2C12 myoblst cells are able to form myotubes, and myofibers and these cells have been employed for muscle regeneration purposes and in myogenic regeneration and transplantation studies. We exposed C2C12 cells in an random position machine to simulate microgravity and study the energy and the biochemical challenges associated with this treatment. Simulated microgravity exposed C2C12 cells maintain positive proliferation indices and delay the differentiation process for several days. On the other hand this treatment significantly alters many of the biochemical and the metabolic characteristics of the cell cultures including calcium homeostasis. Recent data have shown that these perturbations are due to the inhibition of the ryanodine receptors on the membranes of intracellular calcium stores. We were able to reverse this perturbations treating cells with thapsigargin which prevents the segregation of intracellular calcium ions in the mitochondria and in the sarco/endoplasmic reticula. Calcium homeostasis appear a key target of microgravity exposure. In conclusion, in this study we reported some of the effects induced by the exposure of C2C12 cell cultures to simulated microgravity. The promising information obtained is of fundamental importance in the hope to employ this protocol in the field of regenerative medicine.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Muscular/fisiologia , Regeneração/efeitos da radiação , Ausência de Peso/efeitos adversos , Animais , Sinalização do Cálcio/efeitos da radiação , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/efeitos da radiação , Humanos , Camundongos , Desenvolvimento Muscular/efeitos da radiação , Fibras Musculares Esqueléticas/efeitos da radiação , Mioblastos/metabolismo , Mioblastos/efeitos da radiação , Simulação de Ausência de Peso/efeitos adversos
10.
Biochem Biophys Res Commun ; 532(3): 482-488, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-32892951

RESUMO

Leucine-rich repeat containing family 8 (LRRC8) proteins form the volume-regulated anion channel (VRAC). Recently, they were shown to be required for normal differentiation and fusion of C2C12 myoblasts, by promoting membrane hyperpolarization and intracellular Ca2+ signals. However, the mechanism by which they are involved remained obscure. Here, using a FRET-based sensor for VRAC activity, we show temporary activation of VRAC within the first 2 h of myogenic differentiation. During this period, we also observed a significant decrease in the intracellular Cl- concentration that was abolished by the VRAC inhibitor carbenoxolone. However, lowering the intracellular Cl- concentration by extracellular Cl- depletion did not promote differentiation as judged by the percentage of myogenin-positive nuclei or total myogenin levels in C2C12 cells. Instead, it inhibited myosin expression and myotube formation. Together, these data suggest that VRAC is activated and mediates Cl- efflux early on during myogenic differentiation, and a moderate intracellular Cl- concentration is necessary for myoblast fusion.


Assuntos
Cloretos/metabolismo , Proteínas de Membrana/metabolismo , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Animais , Carbenoxolona/farmacologia , Diferenciação Celular/fisiologia , Fusão Celular , Linhagem Celular , Citosol/metabolismo , Transferência Ressonante de Energia de Fluorescência , Transporte de Íons/efeitos dos fármacos , Camundongos , Desenvolvimento Muscular/fisiologia , Mioblastos Esqueléticos/efeitos dos fármacos
11.
Xenobiotica ; 50(11): 1352-1358, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29171786

RESUMO

Genistein is a widely studied phytoestrogen. The effects of genistein on myoblasts were reported long ago, but the conclusions are controversial. In this study, we evaluated the effects of different concentrations of genistein on C2C12 myoblasts. Genistein treatment promoted myoblast proliferation in a dose-dependent manner in the concentration range of 0-2 µM/L, reaching its maximum effect at 2 µM/L. Proliferation then declined, and a concentration higher than 20 µM/L showed significant inhibition. In addition, genistein treatment promoted myoblast differentiation at a dose of 10 µM/L. However, at treatment concentrations higher than 10 µM/L, the effect on myoblast differentiation was rapidly inhibited as the concentration increased. Genistein treatment also down-regulated the expression of miR-222, resulting in increased expression of its target genes, MyoG, MyoD, and ERα and thereby promoting myoblast differentiation. Our results suggest that genistein has a dose-dependent and bidirectional regulation effect on myoblast proliferation and differentiation. We also found that genistein is a miRNA inducer, and it specifically affects the expression of miR-222 to regulate myoblast differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Genisteína , Mioblastos/efeitos dos fármacos , Fitoestrógenos , Humanos , Mioblastos/metabolismo , Mioblastos/fisiologia
12.
Biosci Biotechnol Biochem ; 83(2): 291-299, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30336746

RESUMO

The development of skeletal muscle is a complex process including myoblasts proliferation and differentiation. MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at post-transcriptional level. Increasing evidences indicate that miRNAs are important regulators in myogenic processes. Here, we reported that the expression of miR-10b-5p steadily decreased during myoblasts proliferation, but significantly increased during myoblasts differentiation. The over-expression of miR-10b-5p promoted myoblasts proliferation and blunted myofiber formation in C2C12 cells, while miR-10b-5p down-regulation showed an opposite result. At the same time, we observed that the down-regulation of nuclear factor of activated T-cells 5 (NFAT5) repressed the differentiation of C2C12 cells, and interestingly, miR-10b-5p could suppress NFAT5 expression. Luciferase activity assays confirmed that miR-10b-5p directly target the 3'-untranslated region (3'-UTR) of NFAT5. Overall, we proposed here a novel insight that miR-10b-5p regulates the proliferation and differentiation of C2C12 myoblasts, and the impact on myogenic differentiation is partly through targeting NFAT5. Abbreviations: NFAT5: nuclear factor of activated T-cells 5; Cyclin B: cycle protein B; Cyclin D1: cycle protein D1; Cyclin E: cycle protein E; CDK4: cyclin-dependent kinase 4; MyoD: myogenic differentiation antigen; MyoG: myogenin; Myf5: myogenic factor 5; MRF4: myogenic regulatory factor 4; MyHC: myosin heavy chain; AQP5: aquaporin-5; CACNA1C: calcium voltage-gated channel subunit alpha1 C; SRF: serum response factor; Pax7: paired box 7; KLF4: Kruppel-like factor 4; 3'-UTR: 3'-untranslated region; GM: growth medium; DM: differentiation medium.


Assuntos
Diferenciação Celular/genética , Proliferação de Células/genética , MicroRNAs/fisiologia , Mioblastos/citologia , Regiões 3' não Traduzidas , Animais , Linhagem Celular , Regulação para Baixo , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Fatores de Transcrição/genética
13.
Mol Biol (Mosk) ; 53(1): 44-52, 2019.
Artigo em Russo | MEDLINE | ID: mdl-30895952

RESUMO

MiR-222-3р has been implicated in tumor cell proliferation and has an important role in the differentiation and maturation of myogenic cells. However, its role in skeletal myoblast proliferation is still unclear. In this study, we found that miR-222-3р expression increases initially and then decreases during C2C12 myoblast proliferation. Using synthetic miRNA mimics and inhibitors in gain- or loss-of-function experiments, we snowed that miR-222-3р overexpression in C2C12 cells promotes myoblast proliferation and represses myofiber formation, while miR-222-3р downregulation has the opposite effect. Using a prediction program, BTG2 was identified as a possible target gene of miR-222-3р. During myogenesis, miR-222-3р mimics repress BTG2 expression, while miR-222-3р inhibitors promote BTG2 expression. Using dual-luciferase reporter assay, we further demonstrated that miR-222-3р specifically targets BTG2. Additionally, we show that siRNA-mediated downregulation of BTG2 expression in C2C12 myoblasts promotes the proliferation and suppresses differentiation. In conclusion, we provide a novel insight into the mechanism by which miR-222-3р regulates the proliferation and differentiation of C2C12 myoblasts by targeting BTG2. This information contributes to our understanding of the role of miRNAs in skeletal muscle development.


Assuntos
Diferenciação Celular , Proteínas Imediatamente Precoces/genética , MicroRNAs/genética , Desenvolvimento Muscular , Mioblastos/citologia , Proteínas Supressoras de Tumor/genética , Animais , Linhagem Celular , Proliferação de Células , Camundongos
14.
Biochim Biophys Acta Mol Cell Res ; 1864(4): 697-707, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28130125

RESUMO

Skeletal muscle cell proliferation and differentiation are tightly regulated. Epigenetic regulation is a major component of the regulatory mechanism governing these processes. Histone modification is part of the epigenetic code used for transcriptional regulation of chromatin through the establishment of an active or repressive state for genes involved in myogenesis in a temporal manner. Here, we uncovered the function of SET domain containing 2 (Setd2), an essential histone 3 lysine 36 trimethyltransferase, in regulating the proliferation and differentiation of myoblasts. Setd2 was silenced in the skeletal muscle myoblast cell line, C2C12, using the CRISPR/CAS9 system. The mutant cells exhibited defect in myotube formation. The myotube formation marker, myosin heavy chain (MHC), was downregulated earlier in Setd2 silenced cells compared to wild-type myoblasts during differentiation. The deficiency in Setd2 also resulted in repression of Myogenin (MyoG) expression, a key myogenic regulator during differentiation. In addition to the myoblast differentiation defect, decreased proliferation rate with significantly reduced levels of histone 3 phosphorylation, indicative of cell proliferation defect, were observed in the Setd2 silenced cells; suggesting an impaired proliferation phenotype. Furthermore, compromised G1/S- and G2/M-phase transition and decreased expression levels of major regulators of cell cycle G1/S checkpoints, cyclin D1, CDK4, CDK6, and cyclin E2 were detected in Setd2 silenced cells. Consistent with the cell cycle arrested phenotype, cyclin-dependent kinase inhibitor p21 was upregulated in Setd2 silenced cells. Together, this study demonstrates an essential role of Setd2 in myoblast proliferation and differentiation, and uncovers Setd2-mediated molecular mechanism through regulating MyoG and p21.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Miogenina/genética , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Pontos de Checagem do Ciclo Celular , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Cromatina/química , Cromatina/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Edição de Genes , Inativação Gênica , Histona-Lisina N-Metiltransferase/deficiência , Histonas/metabolismo , Camundongos , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Miogenina/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Fosforilação
15.
FASEB J ; 31(7): 2963-2972, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28356344

RESUMO

Yes-associated protein (YAP) is a transcriptional coactivator in the Hippo pathway that regulates cell proliferation, differentiation, and apoptosis. The MEK5/ERK5 MAPK cascade is essential for the early step of myogenesis. In this study, we generated C2C12 stable cell lines that expressed YAP (C2C12-YAP cells) and found that ERK5 and MEK5 were activated in C2C12-YAP cells compared with control C2C12 (C2C12-vector) cells. C2C12-YAP stable cells also differentiated into myotubes better than C2C12-vector cells, and expressed elevated levels of myogenin, a transcription factor that regulates myogenesis, as well as elevated levels of myosin heavy chain, a skeletal muscle marker. Western blot analysis revealed that Src and c-Abl (Abelson murine leukemia viral oncogene homolog 1) activation were enhanced in C2C12-YAP cells. Conversely, treatment of inhibitors of c-Abl, Src, or MEK5 inhibited activation of MEK5 and ERK5 and myogenesis of C2C12 myoblasts. Specific interactions between YAP and proteins in the ERK5 pathway, such as MEK kinase 3 (MEKK3) and ERK5, were illustrated by coimmunoprecipitation experiments. MEKK3 contains the PPGY motif (aa 178-181), which may interact with YAP. Site-directed mutagenesis experiments revealed that expression of MEKK3 Y181F mutant inhibited MEK5/ERK5 activation and myogenic differentiation. These results suggest that YAP promotes muscle differentiation by activating the Abl/Src/MEKK3/MEK5/ERK5 kinase cascade.-Chen, T.-H., Chen, C.-Y., Wen, H.-C., Chang, C.-C., Wang, H.-D., Chuu, C.-P., Chang, C.-H. YAP promotes myogenic differentiation via the MEK5-ERK5 pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Regulação da Expressão Gênica/fisiologia , MAP Quinase Quinase 5/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Linhagem Celular , Citoplasma , Genes abl , MAP Quinase Quinase 5/genética , MAP Quinase Quinase Quinase 3/genética , MAP Quinase Quinase Quinase 3/metabolismo , Camundongos , Proteína Quinase 7 Ativada por Mitógeno/genética , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Fosfoproteínas/genética , Transporte Proteico , Proteínas de Sinalização YAP , Quinases da Família src
16.
Biochem Biophys Res Commun ; 479(1): 97-102, 2016 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-27629357

RESUMO

Low frequency pulsed electromagnetic field (PEMF) has been shown to affect the activity of various cell types and promote them proliferation. However, its effect on skeletal muscle cells remains to be determined. In our study, we confirmed that PEMF (100 Hz, 1 mT) could promote C2C12 myoblasts proliferation by using Cell Counting Kit-8 (CCK-8) and 5-Ethynyl-2'-deoxyuridine (EdU) assays, yet hardly any distinction was found in the rate of cell apoptosis between PEMF and control groups by flow cytometry (Annexin V-FITC/PI double staining method). To further study the mechanism of action of PEMF, Western blot was utilized to detect the mitogen-activated protein kinase (MAPK) signaling pathways. After exposing C2C12 myoblasts to PEMF, we found the phosphorylation level of extracellular signal-regulated kinase (ERK) was significantly increased, while p38 MAPK and c-Jun N-terminal kinase (JNK) pathways were not affected. Pretreating the cells with the ERK kinase1/2 (MEK1/2) inhibitor U0126 obviously inhibited the proliferation of C2C12 cells. Taken together, our research for the first time demonstrated that PEMF promoted C2C12 myoblasts proliferation via activating MAPK/ERK pathway.


Assuntos
Proliferação de Células/fisiologia , Campos Eletromagnéticos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mioblastos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Western Blotting , Butadienos/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Mioblastos/citologia , Nitrilas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Cell Mol Life Sci ; 72(23): 4633-51, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26077601

RESUMO

Although cholesterol is essential for membrane fluidity and deformability, the level of its lateral heterogeneity at the plasma membrane of living cells is poorly understood due to lack of appropriate probe. We here report on the usefulness of the D4 fragment of Clostridium perfringens toxin fused to mCherry (theta*), as specific, non-toxic, sensitive and quantitative cholesterol-labeling tool, using erythrocyte flat membrane. By confocal microscopy, theta* labels cholesterol-enriched submicrometric domains in coverslip-spread but also gel-suspended (non-stretched) fresh erythrocytes, suggesting in vivo relevance. Cholesterol domains on spread erythrocytes are stable in time and space, restricted by membrane:spectrin anchorage via 4.1R complexes, and depend on temperature and sphingomyelin, indicating combined regulation by extrinsic membrane:cytoskeleton interaction and by intrinsic lipid packing. Cholesterol domains partially co-localize with BODIPY-sphingomyelin-enriched domains. In conclusion, we show that theta* is a useful vital probe to study cholesterol organization and demonstrate that cholesterol forms submicrometric domains in living cells.


Assuntos
Colesterol/metabolismo , Membrana Eritrocítica/metabolismo , Microdomínios da Membrana/metabolismo , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Compostos de Boro/química , Compostos de Boro/metabolismo , Linhagem Celular , Membrana Eritrocítica/química , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Microdomínios da Membrana/química , Camundongos , Mioblastos/metabolismo , Esfingomielinas/química , Esfingomielinas/metabolismo , Temperatura
18.
J Cell Mol Med ; 19(2): 396-407, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25283340

RESUMO

In this paper, we investigated the isoform-specific roles of certain protein kinase C (PKC) isoforms in the regulation of skeletal muscle growth. Here, we provide the first intriguing functional evidence that nPKCδ (originally described as an inhibitor of proliferation in various cells types) is a key player in promoting both in vitro and in vivo skeletal muscle growth. Recombinant overexpression of a constitutively active nPKCδ in C2C12 myoblast increased proliferation and inhibited differentiation. Conversely, overexpression of kinase-negative mutant of nPKCδ (DN-nPKCδ) markedly inhibited cell growth. Moreover, overexpression of nPKCδ also stimulated in vivo tumour growth and induced malignant transformation in immunodeficient (SCID) mice whereas that of DN-nPKCδ suppressed tumour formation. The role of nPKCδ in the formation of rhabdomyosarcoma was also investigated where recombinant overexpression of nPKCδ in human rhabdomyosarcoma RD cells also increased cell proliferation and enhanced tumour formation in mouse xenografts. The other isoforms investigated (PKCα, ß, ε) exerted only minor (mostly growth-inhibitory) effects in skeletal muscle cells. Collectively, our data introduce nPKCδ as a novel growth-promoting molecule in skeletal muscles and invite further trials to exploit its therapeutic potential in the treatment of skeletal muscle malignancies.


Assuntos
Proliferação de Células/fisiologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Músculo Esquelético/metabolismo , Proteína Quinase C-delta/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Camundongos
19.
FASEB J ; 27(11): 4532-46, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23913862

RESUMO

In view of its multiple detrimental effects, transforming growth factor ß1 (TGFß1) is recognized as critical negative regulator of skeletal muscle repair. Apoptosis of skeletal muscle precursor cells driven by TGFß1 contributes to the negative role exerted by the cytokine in tissue repair, although the underlying molecular mechanisms are still elusive. Herein we report the identification of a new signaling pathway, relying on Rho kinase-2 stimulation, subsequent to SMAD-dependent S1P4 up-regulation and transactivation via sphingosine kinase (SK)-2, that accounts for TGFß1-induced apoptosis in cultured myoblasts. S1P4-specific gene silencing reduced by almost 50% activation of caspase-3 and poly-ADP ribosyl transferase cleavage elicited by TGFß1. Moreover, the selective S1P4 antagonist CYM50358 also reduced the TGFß1 proapoptotic effects. By employing pharmacological and molecular biological approaches, the involvement of SK2 and ROCK2 in the transmission of the TGFß1 apoptotic action was also demonstrated. These results reinforce the notion that the SK/S1P axis plays a fundamental role in TGFß1 mode of action in skeletal muscle cells and, by disclosing a novel mechanism by which TGFß1 exerts its harmful action, pinpoint new molecular targets that in principle could be beneficial in the treatment of several skeletal muscle disorders or aging-dependent muscle atrophy.


Assuntos
Apoptose , Mioblastos/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/farmacologia , Quinases Associadas a rho/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular , Camundongos , Mioblastos/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Lisoesfingolipídeo/genética , Receptores de Esfingosina-1-Fosfato
20.
Polymer (Guildf) ; 55(1): 445-452, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-24443593

RESUMO

Cell patches are widely used for healing injuries on the surfaces or interfaces of tissues such as those of epidermis and myocardium. Here we report a novel type of porous scaffolds made of poly(D,L-lactic-co-glycolic acid) for fabricating cell patches. The scaffolds have a single layer of spherical pores arranged in a unique hexagonal pattern and are therefore referred to as "scaffolds with a hexagonal array of interconnected pores (SHAIPs)". SHAIPs contain both uniform pores and interconnecting windows that can facilitate the exchange of biomacromolecules, ensure homogeneous cell seeding, and promote cell migration. As a proof-of-concept demonstration, we have created skeletal muscle patches with a thickness of approximately 150 µm using SHAIPs. The myoblasts seeded in the scaffolds maintained high viability and were able to differentiate into multi-nucleated myotubes. Moreover, neovasculature could efficiently develop into the patches upon subcutaneous implantation in vivo.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA