Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Elife ; 102021 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-34448452

RESUMO

Skeletal muscles are composed of hundreds of multinucleated muscle fibers (myofibers) whose myonuclei are regularly positioned all along the myofiber's periphery except the few ones clustered underneath the neuromuscular junction (NMJ) at the synaptic zone. This precise myonuclei organization is altered in different types of muscle disease, including centronuclear myopathies (CNMs). However, the molecular machinery regulating myonuclei position and organization in mature myofibers remains largely unknown. Conversely, it is also unclear how peripheral myonuclei positioning is lost in the related muscle diseases. Here, we describe the microtubule-associated protein, MACF1, as an essential and evolutionary conserved regulator of myonuclei positioning and maintenance, in cultured mammalian myotubes, in Drosophila muscle, and in adult mammalian muscle using a conditional muscle-specific knockout mouse model. In vitro, we show that MACF1 controls microtubules dynamics and contributes to microtubule stabilization during myofiber's maturation. In addition, we demonstrate that MACF1 regulates the microtubules density specifically around myonuclei, and, as a consequence, governs myonuclei motion. Our in vivo studies show that MACF1 deficiency is associated with alteration of extra-synaptic myonuclei positioning and microtubules network organization, both preceding NMJ fragmentation. Accordingly, MACF1 deficiency results in reduced muscle excitability and disorganized triads, leaving voltage-activated sarcoplasmic reticulum Ca2+ release and maximal muscle force unchanged. Finally, adult MACF1-KO mice present an improved resistance to fatigue correlated with a strong increase in mitochondria biogenesis.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas dos Microfilamentos/metabolismo , Microtúbulos/metabolismo , Mitocôndrias Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Mioblastos Esqueléticos/metabolismo , Junção Neuromuscular/metabolismo , Biogênese de Organelas , Animais , Linhagem Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/ultraestrutura , Acoplamento Excitação-Contração , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Microtúbulos/genética , Microtúbulos/ultraestrutura , Mitocôndrias Musculares/genética , Mitocôndrias Musculares/ultraestrutura , Fadiga Muscular , Fibras Musculares Esqueléticas/ultraestrutura , Força Muscular , Mioblastos Esqueléticos/ultraestrutura , Junção Neuromuscular/genética , Junção Neuromuscular/ultraestrutura , Fatores de Tempo
2.
Am J Physiol Cell Physiol ; 321(4): C749-C759, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34406904

RESUMO

Recently, methods for creating three-dimensional (3-D) human skeletal muscle tissues from myogenic cell lines have been reported. Bioengineered muscle tissues are contractile and respond to electrical and chemical stimulation. In this study, we provide an electrophysiological analysis of healthy and dystrophic 3-D bioengineered skeletal muscle tissues, focusing on Duchenne muscular dystrophy (DMD). We enlist the 3-D in vitro model of DMD muscle tissue to evaluate muscle cell electrical properties uncoupled from presynaptic neural inputs, an understudied aspect of DMD. Our data show that previously reported electrophysiological aspects of DMD, including effects on membrane potential and membrane resistance, are replicated in the 3-D muscle tissue model. Furthermore, we test a potential therapeutic compound, poloxamer 188, and demonstrate capacity for improving the membrane potential in DMD muscle. Therefore, this study serves as a baseline for a new in vitro method to examine potential therapies for muscular disorders.


Assuntos
Distrofina/metabolismo , Potenciais da Membrana , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Mioblastos Esqueléticos/metabolismo , Engenharia Tecidual , Adolescente , Estudos de Casos e Controles , Técnicas de Cultura de Células , Linhagem Celular , Criança , Distrofina/genética , Impedância Elétrica , Humanos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/ultraestrutura , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Mutação , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/ultraestrutura , Poloxâmero/farmacologia , Sódio/metabolismo
3.
J Cell Physiol ; 235(5): 4667-4678, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31637727

RESUMO

Four and a half LIM domain protein 1 (FHL1) belongs to the FHL protein family and is predominantly expressed in skeletal and cardiac muscle. FHL1 acts as a scaffold during sarcomere assembly and plays a vital role in muscle growth and development. Autophagy is key to skeletal muscle development and regeneration, with its dysfunction associated with a range of muscular pathologies and disorders. In this study, we constructed FHL1-silenced or FHL1-overexpressed myoblasts to investigate its role in autophagy during the differentiation of chicken myoblasts into myotubules. Our data showed that FHL1 contributes to myoblast differentiation as measured through MyoG, MyoD, Myh3, and Mb mRNA expression, MyoG and MyHC protein expression and the morphological characteristics of myoblasts. The results showed that FHL1 silencing inhibited the expression of ATG5 and ATG7, meanwhile, immunofluorescence and immunoprecipitation showed that FHL1 and LC3 interacted to regulate the correct formation of autophagosomes. FHL1 inhibition increased cleaved caspase-3 and PARP abundance and promoted myoblast apoptosis. Furthermore, FHL1 rescued skeletal muscle atrophy through regulating the expression of Atrogin-1 and MuRF1. Taken together, these data suggested that FHL1 regulates chicken myoblast differentiation through its interaction with LC3.


Assuntos
Autofagia , Diferenciação Celular , Proteínas com Domínio LIM/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Desenvolvimento Muscular , Proteínas Musculares/metabolismo , Mioblastos Esqueléticos/metabolismo , Animais , Apoptose , Células Cultivadas , Galinhas , Regulação da Expressão Gênica , Proteínas com Domínio LIM/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Musculares/genética , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Mioblastos Esqueléticos/ultraestrutura , Transdução de Sinais
4.
PLoS One ; 13(4): e0196255, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29698438

RESUMO

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


Assuntos
Fatores de Transcrição MEF2/fisiologia , Músculo Esquelético/fisiologia , Mioblastos Esqueléticos/fisiologia , Mioblastos Esqueléticos/ultraestrutura , Adenoviridae/genética , Animais , Bovinos , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Citometria de Fluxo , Regulação da Expressão Gênica , Desenvolvimento Muscular , Proteínas Musculares/fisiologia , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo
5.
Oxid Med Cell Longev ; 2016: 5152029, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27610211

RESUMO

Creatine (Cr) is a nutritional supplement promoting a number of health benefits. Indeed Cr has been shown to be beneficial in disease-induced muscle atrophy, improve rehabilitation, and afford mild antioxidant activity. The beneficial effects are likely to derive from pleiotropic interactions. In accord with this notion, we previously demonstrated that multiple pleiotropic effects, including preservation of mitochondrial damage, account for the capacity of Cr to prevent the differentiation arrest caused by oxidative stress in C2C12 myoblasts. Given the importance of mitochondria in supporting the myogenic process, here we further explored the protective effects of Cr on the structure, function, and networking of these organelles in C2C12 cells differentiating under oxidative stressing conditions; the effects on the energy sensor AMPK, on PGC-1α, which is involved in mitochondrial biogenesis and its downstream effector Tfam were also investigated. Our results indicate that damage to mitochondria is crucial in the differentiation imbalance caused by oxidative stress and that the Cr-prevention of these injuries is invariably associated with the recovery of the normal myogenic capacity. We also found that Cr activates AMPK and induces an upregulation of PGC-1α expression, two events which are likely to contribute to the protection of mitochondrial quality and function.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Creatina/farmacologia , Mitocôndrias Musculares/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Mioblastos Esqueléticos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Citoproteção , DNA Mitocondrial/efeitos dos fármacos , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Proteínas de Grupo de Alta Mobilidade/metabolismo , Peróxido de Hidrogênio/toxicidade , Peroxidação de Lipídeos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/ultraestrutura , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/ultraestrutura , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteômica/métodos , Transdução de Sinais/efeitos dos fármacos
6.
FEBS Lett ; 590(3): 317-29, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26786059

RESUMO

The multifunctional protein Lmo7 has been implicated in some aspects of myogenesis in mammals. Here we studied the distribution and expression of Lmo7 and the effects of Lmo7 knockdown in primary cultures of chick skeletal muscle cells. Lmo7 was localized within the nuclei of myoblasts and at the perinuclear region of myotubes. Knockdown of Lmo7 using siRNA specific to chick reduces the number and width of myotubes and the number of MyoD positive-myoblasts. Both Wnt3a enriched medium and Bio, activators of the Wnt/beta-catenin pathway, could rescue the effects of the Lmo7 knockdown suggesting a crosstalk between the Wnt/beta-catenin and Lmo7-mediated signaling pathways. Our data shows a role of Lmo7 during the initial events of chick skeletal myogenesis, particularly in myoblast survival.


Assuntos
Proteínas Aviárias/metabolismo , Proteínas com Domínio LIM/metabolismo , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/metabolismo , Mioblastos Esqueléticos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas Aviárias/antagonistas & inibidores , Proteínas Aviárias/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Células Cultivadas , Embrião de Galinha , Citoplasma/metabolismo , Citoplasma/ultraestrutura , França , Proteínas de Fluorescência Verde/antagonistas & inibidores , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Recém-Nascido , Proteínas com Domínio LIM/antagonistas & inibidores , Proteínas com Domínio LIM/genética , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/ultraestrutura , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/ultraestrutura , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sarcolema/metabolismo , Sarcolema/ultraestrutura , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Via de Sinalização Wnt
7.
Endocrinology ; 157(1): 23-38, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26562261

RESUMO

Thyroid hormone (TH) and autophagy share similar functions in regulating skeletal muscle growth, regeneration, and differentiation. Although TH recently has been shown to increase autophagy in liver, the regulation and role of autophagy by this hormone in skeletal muscle is not known. Here, using both in vitro and in vivo models, we demonstrated that TH induces autophagy in a dose- and time-dependent manner in skeletal muscle. TH induction of autophagy involved reactive oxygen species (ROS) stimulation of 5'adenosine monophosphate-activated protein kinase (AMPK)-Mammalian target of rapamycin (mTOR)-Unc-51-like kinase 1 (Ulk1) signaling. TH also increased mRNA and protein expression of key autophagy genes, microtubule-associated protein light chain 3 (LC3), Sequestosome 1 (p62), and Ulk1, as well as genes that modulated autophagy and Forkhead box O (FOXO) 1/3a. TH increased mitochondrial protein synthesis and number as well as basal mitochondrial O2 consumption, ATP turnover, and maximal respiratory capacity. Surprisingly, mitochondrial activity and biogenesis were blunted when autophagy was blocked in muscle cells by Autophagy-related gene (Atg)5 short hairpin RNA (shRNA). Induction of ROS and 5'adenosine monophosphate-activated protein kinase (AMPK) by TH played a significant role in the up-regulation of Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A), the key regulator of mitochondrial synthesis. In summary, our findings showed that TH-mediated autophagy was essential for stimulation of mitochondrial biogenesis and activity in skeletal muscle. Moreover, autophagy and mitochondrial biogenesis were coupled in skeletal muscle via TH induction of mitochondrial activity and ROS generation.


Assuntos
Autofagia , Mitocôndrias Musculares/metabolismo , Dinâmica Mitocondrial , Músculo Esquelético/metabolismo , Tri-Iodotironina/metabolismo , Proteínas Quinases Ativadas por AMP/química , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Cinética , Masculino , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias Musculares/efeitos dos fármacos , Mitocôndrias Musculares/ultraestrutura , Dinâmica Mitocondrial/efeitos dos fármacos , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/ultraestrutura , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/ultraestrutura , Consumo de Oxigênio/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Tiroxina/metabolismo , Tiroxina/farmacologia , Fatores de Transcrição/agonistas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tri-Iodotironina/farmacologia
8.
Curr Pharm Des ; 21(25): 3665-72, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25613787

RESUMO

Apoptosis is essential for skeletal muscle development and homeostasis. It has been frequently involved in several muscle myopathies and sarcopenia, as well as in denervation, in disuse and acute strenuous or eccentric physical exercise. In this work skeletal muscle cell death, induced in vitro by a variety of physical triggers, has been investigated. C2C12 myoblasts and myotubes were exposed to UVB for 30 min, hyperthermia for 1 h at 43 °C, low pH for 3 h, hypothermia for 4h at 0 - 6°C, all followed by 2 - 4 h recovery. Their effects have been analysed by means of morpho- functional and molecular approaches. After UVB radiation, hyperthermia and acidosis, morphological apoptotic features and in situ DNA fragmentation appeared, more evident in myoblasts. Interestingly, apoptotic, non apoptotic and necrotic nuclei could be occasionally observed within the same myotube. Low pH induced apoptosis and necrosis, both characterized by swollen nuclei. In all these experimental conditions, the molecular investigations revealed a caspase pathway involvement in inducing cell death. Differently, hypothermia showed a scant and initial chromatin margination, in the presence of a diffused autophagic component. In this case, in situ DNA fragmentation and caspase activation have not been detected. Myoblasts and myotubes appeared sensitive to physical agents, some of which, induced apoptotic cell death. Moreover, hypothermia exposure seemed to enhance autophagic response, thus representing a way to delay trauma-correlated muscle inflammation. This study permits to highlight skeletal muscle cell behavior in response to physical agents, by adding important information to muscle cell death knowledge. UVB radiation and hyperthermia, usually used in clinical therapy, have also adverse effects on skeletal muscle such as myonuclei loss and cell death, contributing to muscle mass decrease. Acidosis occurs physiologically in muscular fatigue, reducing not only the athlete performance, but causing muscle cell damage or death too. Finally, hypothermia, stimulating the autophagic response, could have a key role in muscle injury prevention.


Assuntos
Apoptose , Temperatura Baixa , Temperatura Alta , Fibras Musculares Esqueléticas/ultraestrutura , Mioblastos Esqueléticos/ultraestrutura , Raios Ultravioleta , Animais , Apoptose/efeitos da radiação , Western Blotting , Técnicas de Cultura de Células , Linhagem Celular , Fragmentação do DNA/efeitos da radiação , Eletroforese em Gel de Ágar , Concentração de Íons de Hidrogênio , Marcação In Situ das Extremidades Cortadas , Camundongos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Fibras Musculares Esqueléticas/efeitos da radiação , Mioblastos Esqueléticos/efeitos da radiação
9.
Regen Med ; 7(1): 47-57, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22168497

RESUMO

AIM: To determine whether our novel approach of diazoxide-induced stem cell preconditioning might be extrapolated to human skeletal myoblasts to support their survival under lethal oxidant stress. METHODS & RESULTS: Using an in vitro model of H(2)O(2) treatment of human skeletal myoblasts, we report the ability of diazoxide-preconditioned human skeletal myoblasts to express cytokines and growth factors, which act in an autocrine and paracrine fashion to promote their own survival. Preconditioning of skeletal myoblasts was cytoprotective and significantly reduced their apoptotic index (p < 0.05). IL-11 gene and protein expression was significantly increased in preconditioned skeletal myoblasts. Transfection of skeletal myoblasts with IL-11-specific siRNA incurred their death under oxidant stress. The cytoprotective effect of diazoxide preconditioning was blocked by Erk1/2 inhibitor PD98059 (20-100 µM), which abrogated STAT-3 phosphorylation, thus confirming a possible involvement of Erk1/2/STAT3 signaling downstream of IL-11 in cell survival. We also investigated the time course of subcellular changes and signaling pathway of skeletal myoblasts apoptosis under oxidant stress before and after preconditioning. Apoptosis was induced in skeletal myoblasts with 100-500 µM H(2)O(2) for time points ranging from 1 to 24 h. Release of lactate dehydrogenase, disruption of the mitochondrial membrane potential and cytochrome-c translocation into cytoplasm were the earliest signs of apoptosis. Total Akt protein remained unchanged whereas marked reduction in pAkt was observed in the native skeletal myoblasts. Terminal dUTP nick end-labeling and annexin-V positivity were significantly increased after 4 h. Ultra-structure studies showed condensed chromatin, shriveled nuclei and swollen mitochondria. CONCLUSION: These data suggest that skeletal myoblasts undergo apoptosis under oxidant stress in a time-dependent manner and preconditioning of skeletal myoblasts significantly prevented their apoptosis via IL-11/STAT3 signaling.


Assuntos
Apoptose/efeitos dos fármacos , Interleucina-11/metabolismo , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Oxidantes/farmacologia , Fator de Transcrição STAT3/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Anexina A5/metabolismo , Western Blotting , Linhagem Celular , Citocromos c/metabolismo , Citoproteção/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Marcação In Situ das Extremidades Cortadas , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/ultraestrutura , Fosforilação/efeitos dos fármacos , Propídio/metabolismo , Interferência de RNA/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Coloração e Rotulagem , Fatores de Tempo
10.
J Cell Sci ; 124(Pt 24): 4213-20, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22193956

RESUMO

Myoblasts aggregate, differentiate and fuse to form skeletal muscle during both embryogenesis and tissue regeneration. For proper muscle function, long-range self-organization of myoblasts is required to create organized muscle architecture globally aligned to neighboring tissue. However, how the cells process geometric information over distances considerably longer than individual cells to self-organize into well-ordered, aligned and multinucleated myofibers remains a central question in developmental biology and regenerative medicine. Using plasma lithography micropatterning to create spatial cues for cell guidance, we show a physical mechanism by which orientation information can propagate for a long distance from a geometric boundary to guide development of muscle tissue. This long-range alignment occurs only in differentiating myoblasts, but not in non-fusing myoblasts perturbed by microfluidic disturbances or other non-fusing cell types. Computational cellular automata analysis of the spatiotemporal evolution of the self-organization process reveals that myogenic fusion in conjunction with rotational inertia functions in a self-reinforcing manner to enhance long-range propagation of alignment information. With this autocatalytic alignment feedback, well-ordered alignment of muscle could reinforce existing orientations and help promote proper arrangement with neighboring tissue and overall organization. Such physical self-enhancement might represent a fundamental mechanism for long-range pattern formation during tissue morphogenesis.


Assuntos
Desenvolvimento Muscular , Fibras Musculares Esqueléticas/citologia , Mioblastos Esqueléticos/citologia , Animais , Linhagem Celular , Células Cultivadas , Retroalimentação Fisiológica , Camundongos , Mioblastos Esqueléticos/ultraestrutura , Sarcômeros/ultraestrutura
11.
Tsitologiia ; 53(3): 227-34, 2011.
Artigo em Russo | MEDLINE | ID: mdl-21598685

RESUMO

The study of changes in the intracellular processes during differentiation of myoblasts into myotubules is of great importance for understanding several fundamental problems of cell biology. At first, this concerns the spatial organization of vacuolar apparatus that reflects the alterations in the properties of cell membranes, cytoskeleton elements and dynamics of vesicular transport in the course of differentiation. The distribution of acidic membrane organelles (lysosomes, late endosomes, Golgi cisternae) during the myotubule formation was revealed. It was shown that perinuclear localization of acidic organelles in myoblasts was replaced by diffuse distribution of these structures in the whole volume of myotubules. Using lipophilic fluorescent dyes, RH 414 and di-8-ANEPPS, the process of formation and dynamics of endocytic vesicles in myoblasts and myotubules was investigated. In the present work, semiconductive nanocrystals, quantum dots (QDs), conjugated with TAT-peptide, which belongs to cell-penetrating peptides, were used to characterize nonspecific endocytosis. It was shown that QDs--TAT complexes penetrate myoblasts but do not penetrate myotubules even after 24 h incubation, which might be connected with plasma membrane changes during the process of skeletal muscle differentiation.


Assuntos
Diferenciação Celular/fisiologia , Estruturas Celulares/ultraestrutura , Fibras Musculares Esqueléticas/citologia , Mioblastos Esqueléticos/citologia , Animais , Técnicas de Cultura de Células , Linhagem Celular , Microscopia Confocal , Fibras Musculares Esqueléticas/ultraestrutura , Mioblastos Esqueléticos/ultraestrutura , Ratos
12.
Biotechnol Prog ; 27(3): 891-5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21574266

RESUMO

Microelectrode array (MEA) technology holds tremendous potential in the fields of biodetection, lab-on-a-chip applications, and tissue engineering by facilitating noninvasive electrical interaction with cells in vitro. To date, significant efforts at integrating the cellular component with this detection technology have worked exclusively with neurons or cardiac myocytes. We investigate the feasibility of using MEAs to record from skeletal myotubes derived from primary myoblasts as a way of introducing a third electrogenic cell type and expanding the potential end applications for MEA-based biosensors. We find that the extracellular action potentials (EAPs) produced by spontaneously contractile myotubes have similar amplitudes to neuronal EAPs. It is possible to classify myotube EAPs by biological signal source using a shape-based spike sorting process similar to that used to analyze neural spike trains. Successful spike-sorting is indicated by a low within-unit variability of myotube EAPs. Additionally, myotube activity can cause simultaneous activation of multiple electrodes, in a similar fashion to the activation of electrodes by networks of neurons. The existence of multiple electrode activation patterns indicates the presence of several large, independent myotubes. The ability to identify these patterns suggests that MEAs may provide an electrophysiological basis for examining the process by which myotube independence is maintained despite rapid myoblast fusion during differentiation. Finally, it is possible to use the underlying electrodes to selectively stimulate individual myotubes without stimulating others nearby. Potential uses of skeletal myotubes grown on MEA substrates include lab-on-a-chip applications, tissue engineering, co-cultures with motor neurons, and neural interfaces.


Assuntos
Potenciais de Ação , Biotecnologia/métodos , Fibras Musculares Esqueléticas/fisiologia , Neurônios/fisiologia , Animais , Técnicas Biossensoriais , Eletrofisiologia , Humanos , Microeletrodos , Mioblastos Esqueléticos/ultraestrutura
13.
Medicina (Kaunas) ; 47(11): 607-15, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22286576

RESUMO

UNLABELLED: Acute myocardial infarction leads to the loss of functional cardiomyocytes and structural integrity. The adult heart cannot repair the damaged tissue due to inability of mature cardiomyocytes to divide and lack of stem cells. The aim of this study was to evaluate the efficiency of introduced autologous skeletal musclederived stem cells to recover the function of acutely infarcted rabbit heart in the early postoperative period. MATERIAL AND METHODS: As a model for myocardium restoration in vivo, experimental rabbit heart infarct was used. Autologic adult myogenic stem cells were isolated from skeletal muscle and propagated in culture. Before transplantation, the cells were labeled with 4',6-diamidino-2-phenylindole and then, during heart surgery, introduced into the rabbit acutely infarcted myocardium. Postoperative cardiac function was monitored by recording electrocardiograms and echocardiograms. At the end of the experiment, the efficiency of cell integration was evaluated histologically. RESULTS: Rabbit cardiac function recovered after 1 month after the induction of experimental infarction both in the control and experimental groups. Therefore, the first month after the infarction was the most significant for the assessment of cell transplantation efficacy. Transplanted cell integration into infarcted myocardium was time- and individual-dependent. Evaluation of changes in left ventricular ejection fraction after the induction of myocardial infarction revealed better recovery in the experimental group; however, the difference among animals in the experimental and control groups varied and was not significant. CONCLUSIONS. Autologous myogenic stem cells repopulated infarcted myocardium with different efficiency in each individual. This variability may account for the observed difference in postoperative cardiac recovery in a rabbit model.


Assuntos
Mioblastos Esqueléticos/transplante , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/cirurgia , Miocárdio , Recuperação de Função Fisiológica , Animais , Modelos Animais de Doenças , Mioblastos Esqueléticos/ultraestrutura , Infarto do Miocárdio/diagnóstico por imagem , Coelhos , Transplante Autólogo , Ultrassonografia
14.
Curr Pharm Des ; 16(8): 956-67, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20041822

RESUMO

Stem cells other than satellite cells that can give rise to primary myoblasts, which are able to form additional new fibers postnatally, are present in the interstitial spaces of skeletal muscle. These cells are sorted into CD34(+)/45(-) (Sk-34) and CD34(-)/45(-) (Sk-DN) cell fractions, and they are wholly (>99%) negative for Pax7 at initial isolation. Colony-forming units of these cells typically include non-adherent type myogenic cells, while satellite cells are known to be adherent in cell culture. In addition, both Pax7(-) and Pax7(+) cells are produced, depending on asymmetric cell division. A large number of myotubes are also formed in each colony, thus suggesting that putative Pax7(+) satellite cells also present in each colony. Interestingly, interstitial myogenic cells show basal lamina formation at early stages of myogenesis in response to various types of stimulation in compensatory enlarged muscle, a property that satellite cells do not possess in the parent fiber basal lamina cylinder. Basal lamina formation and production of satellite cells are essential before muscle fiber establishment in vivo. It is therefore likely that myogenic cells in skeletal muscle can be divided into two populations: 1) basal lamina-producing myogenic cells; and 2) basal lamina-non-producing myogenic cells. The latter population may be Pax7(+) satellite cells showing adherent capacity in cell culture, while the lamina-producing myogenic population derived from interstitial multipotent stem cells, which is predominant among Sk-34 and Sk-DN cells, plays a role in primary myoblast generation and shows non-adherent behavior in culture. Therefore, the physiological role of interstitial myogenic cells is as a source for new postnatal muscle fiber formation, and multinucleated muscle fibers (cells) are potentially formed clonally.


Assuntos
Líquido Extracelular/citologia , Hiperplasia/fisiopatologia , Fibras Musculares Esqueléticas/patologia , Mioblastos Esqueléticos/fisiologia , Transplante de Células-Tronco/métodos , Animais , Antígenos de Superfície/metabolismo , Membrana Basal/anatomia & histologia , Membrana Basal/ultraestrutura , Diferenciação Celular/fisiologia , Proliferação de Células , Humanos , Modelos Biológicos , Fibras Musculares Esqueléticas/fisiologia , Fibras Musculares Esqueléticas/ultraestrutura , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/transplante , Mioblastos Esqueléticos/ultraestrutura , Condicionamento Físico Animal/fisiologia
15.
BMC Biol ; 7: 41, 2009 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-19607661

RESUMO

BACKGROUND: Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant neuromuscular disorder associated with the partial deletion of integral numbers of 3.3 kb D4Z4 DNA repeats within the subtelomere of chromosome 4q. A number of candidate FSHD genes, adenine nucleotide translocator 1 gene (ANT1), FSHD-related gene 1 (FRG1), FRG2 and DUX4c, upstream of the D4Z4 array (FSHD locus), and double homeobox chromosome 4 (DUX4) within the repeat itself, are upregulated in some patients, thus suggesting an underlying perturbation of the chromatin structure. Furthermore, a mouse model overexpressing FRG1 has been generated, displaying skeletal muscle defects. RESULTS: In the context of myogenic differentiation, we compared the chromatin structure and tridimensional interaction of the D4Z4 array and FRG1 gene promoter, and FRG1 expression, in control and FSHD cells. The FRG1 gene was prematurely expressed during FSHD myoblast differentiation, thus suggesting that the number of D4Z4 repeats in the array may affect the correct timing of FRG1 expression. Using chromosome conformation capture (3C) technology, we revealed that the FRG1 promoter and D4Z4 array physically interacted. Furthermore, this chromatin structure underwent dynamic changes during myogenic differentiation that led to the loosening of the FRG1/4q-D4Z4 array loop in myotubes. The FRG1 promoter in both normal and FSHD myoblasts was characterized by H3K27 trimethylation and Polycomb repressor complex binding, but these repression signs were replaced by H3K4 trimethylation during differentiation. The D4Z4 sequences behaved similarly, with H3K27 trimethylation and Polycomb binding being lost upon myogenic differentiation. CONCLUSION: We propose a model in which the D4Z4 array may play a critical chromatin function as an orchestrator of in cis chromatin loops, thus suggesting that this repeat may play a role in coordinating gene expression.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , Desenvolvimento Muscular/genética , Distrofia Muscular Facioescapuloumeral/genética , Mioblastos Esqueléticos/metabolismo , Proteínas Nucleares/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Cromatina/química , Cromatina/metabolismo , Cromossomos Humanos Par 4/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Histona Desmetilases com o Domínio Jumonji , Masculino , Proteínas dos Microfilamentos , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapuloumeral/metabolismo , Mioblastos Esqueléticos/ultraestrutura , Proteínas Nucleares/genética , Oxirredutases N-Desmetilantes/metabolismo , Proteínas do Grupo Polycomb , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA , Proteínas Repressoras/metabolismo , Sequências de Repetição em Tandem/fisiologia
16.
J Mol Cell Cardiol ; 47(2): 335-45, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19465027

RESUMO

The possibility that resident myocardial progenitor cells may be re-activated by transplantation of exogenous stem cells into the post-infarcted heart has been suggested as a possible mechanism to explain the heart's functional improvement after stem cell therapy. Here we studied whether differentiation of mouse neonatal immature cardiomyocytes in vitro was influenced by mouse skeletal myoblasts C2C12, wild type or engineered to secrete the cardiotropic hormone relaxin. The cultured cardiomyocytes formed spontaneously beating clusters and temporally exhibited cardiac immunophenotypical (cKit, atrial natriuretic peptide, troponin T, connexin-43, HCN4) and electrical features (inward voltage-dependent Na(+), T- and L-type Ca(2+) currents, outward and inward K(+) currents, I(f) pacemaker current). These clusters were functionally connected through nanotubular structures and undifferentiated cardiac cells in the form of flattened stripes, bridging the clusters through connexin-43-containing gap junctions. These findings suggested the existence of long distance cell-to-cell communications among the cardiomyocyte aggregates involved in the intercellular transfer of Ca(2+) signals and organelles, likely required for coordination of myocardial differentiation. Co-presence of the myoblasts greatly increased cardiomyocyte differentiation and the amount of intercellular connections. In fact, these cells formed a structural support guiding elongation of nanotubules and stripe-like cells. The secretion of relaxin by the engineered myoblasts accelerated and enhanced the cardiomyogenic potential of the co-culture. These findings underscore the possibility that grafted myoblasts and cardiotropic factors, such as relaxin, may influence regeneration of resident immature cardiac cells, thus adding a tile to the mosaic of mechanisms involved in the functional benefits of cell transplantation for cardiac repair.


Assuntos
Comunicação Celular , Diferenciação Celular , Mioblastos Esqueléticos/metabolismo , Miócitos Cardíacos/citologia , Relaxina/metabolismo , Animais , Animais Recém-Nascidos , Membrana Celular/metabolismo , Células Cultivadas , Técnicas de Cocultura , Conexina 43/metabolismo , Fenômenos Eletrofisiológicos , Imunofenotipagem , Ativação do Canal Iônico , Camundongos , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/ultraestrutura , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Fatores de Tempo
17.
Am J Physiol Cell Physiol ; 295(1): C160-72, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18480300

RESUMO

Membrane-cytoskeleton interaction regulates transmembrane currents through stretch-activated channels (SACs); however, the mechanisms involved have not been tested in living cells. We combined atomic force microscopy, confocal immunofluorescence, and patch-clamp analysis to show that stress fibers (SFs) in C2C12 myoblasts behave as cables that, tensed by myosin II motor, activate SACs by modifying the topography and the viscoelastic (Young's modulus and hysteresis) and electrical passive (membrane capacitance, C(m)) properties of the cell surface. Stimulation with sphingosine 1-phosphate to elicit SF formation, the inhibition of Rho-dependent SF formation by Y-27632 and of myosin II-driven SF contraction by blebbistatin, showed that not SF polymerization alone but the generation of tensional forces by SF contraction were involved in the stiffness response of the cell surface. Notably, this event was associated with a significant reduction in the amplitude of the cytoskeleton-mediated corrugations in the cell surface topography, suggesting a contribution of SF contraction to plasma membrane stretching. Moreover, C(m), used as an index of cell surface area, showed a linear inverse relationship with cell stiffness, indicating participation of the actin cytoskeleton in plasma membrane remodeling and the ability of SF formation to cause internalization of plasma membrane patches to reduce C(m) and increase membrane tension. SF contraction also increased hysteresis. Together, these data provide the first experimental evidence for a crucial role of SF contraction in SAC activation. The related changes in cell viscosity may prevent SAC from abnormal activation.


Assuntos
Canais Iônicos/fisiologia , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Mioblastos Esqueléticos/fisiologia , Fibras de Estresse/fisiologia , Animais , Fenômenos Biomecânicos , Cálcio/fisiologia , Linhagem Celular , Membrana Celular/fisiologia , Citoesqueleto/fisiologia , Imunofluorescência , Ativação do Canal Iônico , Canais Iônicos/biossíntese , Mecanotransdução Celular , Camundongos , Microscopia de Força Atômica , Microscopia Confocal , Mioblastos Esqueléticos/ultraestrutura , Técnicas de Patch-Clamp , Estresse Mecânico , Tensão Superficial , Viscosidade
18.
J Cell Biol ; 180(4): 787-802, 2008 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-18299349

RESUMO

Nuclear factor kappaB (NF-kappaB) is involved in multiple skeletal muscle disorders, but how it functions in differentiation remains elusive given that both anti- and promyogenic activities have been described. In this study, we resolve this by showing that myogenesis is controlled by opposing NF-kappaB signaling pathways. We find that myogenesis is enhanced in MyoD-expressing fibroblasts deficient in classical pathway components RelA/p65, inhibitor of kappaB kinase beta (IKKbeta), or IKKgamma. Similar increases occur in myoblasts lacking RelA/p65 or IKKbeta, and muscles from RelA/p65 or IKKbeta mutant mice also contain higher fiber numbers. Moreover, we show that during differentiation, classical NF-kappaB signaling decreases, whereas the induction of alternative members IKKalpha, RelB, and p52 occurs late in myogenesis. Myotube formation does not require alternative signaling, but it is important for myotube maintenance in response to metabolic stress. Furthermore, overexpression or knockdown of IKKalpha regulates mitochondrial content and function, suggesting that alternative signaling stimulates mitochondrial biogenesis. Together, these data reveal a unique IKK/NF-kappaB signaling switch that functions to both inhibit differentiation and promote myotube homeostasis.


Assuntos
Quinase I-kappa B/metabolismo , Mitocôndrias/metabolismo , Desenvolvimento Muscular/genética , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , NF-kappa B/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Linhagem Celular , Células Cultivadas , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Quinase I-kappa B/genética , Camundongos , Camundongos Transgênicos , Mitocôndrias/ultraestrutura , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/ultraestrutura , Mioblastos Esqueléticos/ultraestrutura , Transdução de Sinais/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
19.
J Cell Physiol ; 209(2): 370-8, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16897754

RESUMO

Diacylglycerol kinases (DGKs) catalyze phosphorylation of diacylglycerol (DG) to yield phosphatidic acid (PA). Previous evidence has shown that the nucleus contains several DGK isoforms. In this study, we have analyzed the expression and subnuclear localization of DGK-zeta employing C2C12 mouse myoblasts. Immunocytochemistry coupled to confocal laser scanning microscopy showed that both endogenous and green fluorescent protein-tagged overexpressed DGK-zeta localized mostly to the nucleus. In contrast, overexpressed DGK-alpha, -beta, -delta, and -iota did not migrate to the nucleus. DGK-zeta was present in the nuclear speckle domains, as also revealed by immuno-electron microscopy analysis. Moreover, DGK-zeta co-localized and interacted with phosphoinositide-specific phospholipase Cbeta1 (PLCbeta1), that is involved in inositide-dependent signaling pathways important for the regulation of cell proliferation and differentiation. Furthermore, we report that DGK-zeta associated with nuclear matrix, the fundamental organizing principle of the nucleus where many cell functions take place, including DNA replication, gene expression, and protein phosphorylation. Nuclear DGK-zeta increased during myogenic differentiation of C2C12 cells, while DGK-zeta down-regulation by siRNA markedly impaired differentiation. Overall, our findings further support the importance of speckles and nuclear matrix in lipid-dependent signaling and suggest that nuclear DGK-zeta might play some fundamental role during myogenic differentiation of C2C12 cells.


Assuntos
Diferenciação Celular/fisiologia , Diacilglicerol Quinase/metabolismo , Mioblastos Esqueléticos/enzimologia , Matriz Nuclear/metabolismo , Amanitinas/farmacologia , Animais , Benzimidazóis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Citoplasma/metabolismo , Regulação para Baixo/efeitos dos fármacos , Heterocromatina/ultraestrutura , Isoenzimas/metabolismo , Camundongos , Desenvolvimento Muscular/efeitos dos fármacos , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/ultraestrutura , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase C beta , Fosfotransferases/metabolismo , Transporte Proteico/efeitos dos fármacos , Fosfolipases Tipo C/metabolismo
20.
Am J Pathol ; 169(3): 761-73, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936253

RESUMO

Bcl-2-associated athanogene 3 (BAG3) is a member of a conserved family of cyto-protective proteins that bind to and regulate Hsp70 family molecular chaperones. Here, we show that BAG3 is prominently expressed in striated muscle and colocalizes with Z-disks. Mice with homozygous disruption of the bag3 gene developed normally but deteriorated postnatally with stunted growth evident by 1 to 2 weeks of age and death by 4 weeks. BAG3-deficient animals developed a fulminant myopathy characterized by noninflammatory myofibrillar degeneration with apoptotic features. Knockdown of bag3 expression in cultured C2C12 myoblasts increased apoptosis on induction of differentiation, suggesting a need for bag3 for maintenance of myotube survival and confirming a cell autonomous role for bag3 in muscle. We conclude that although BAG3 is not required for muscle development, this co-chaperone appears to be critically important for maintenance of mature skeletal muscle.


Assuntos
Proteínas de Transporte/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Mioblastos Esqueléticos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Diferenciação Celular/genética , Linhagem Celular , Sobrevivência Celular/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Camundongos , Camundongos Knockout , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/ultraestrutura , Doenças Musculares/genética , Doenças Musculares/patologia , Mioblastos Esqueléticos/ultraestrutura , Ligação Proteica/genética , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA