Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Pharm Bull ; 43(11): 1785-1791, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33132324

RESUMO

Mesenchymal stem cells (MSCs) are capable of repairing skeletal muscle via paracrine mechanisms. This regenerative effect of MSCs on skeletal muscle is based on promoting the proliferation and differentiation of myogenic cells and inhibiting the inflammatory response of immune cells. However, it is unclear whether MSCs affect the inflammatory response of skeletal muscle cells. In this study, we evaluated the paracrine effect of mouse MSCs on the inflammatory response of lipopolysaccharide (LPS)-stimulated C2C12 mouse myoblasts. Interleukin (IL)-6 production from LPS-stimulated C2C12 cells was significantly increased by coculture with MSCs or culture in conditioned medium of MSCs. This increased IL-6 production from C2C12 cells was not significantly suppressed by inhibiting mitogen-activated protein kinase pathways, but it was significantly suppressed by pretreatment with nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) inhibitors. In addition, IL-6 and inducible nitric oxide synthase (iNOS) mRNA expression was increased significantly in C2C12 cells cocultured with MSCs, while tumor necrosis factor (TNF)-α and IL-1ß mRNA expression was decreased. Furthermore, conditioned medium of C2C12 cells cocultured with MSCs exerted remarkable anti-inflammatory effects on LPS-stimulated mouse macrophages.


Assuntos
Sistema de Sinalização das MAP Quinases/imunologia , Células-Tronco Mesenquimais/metabolismo , Mioblastos Esqueléticos/imunologia , Comunicação Parácrina/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular , Proliferação de Células , Técnicas de Cocultura , Meios de Cultivo Condicionados/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/imunologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Mioblastos Esqueléticos/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
Inflammation ; 41(1): 199-212, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28971270

RESUMO

The objective of this study is to investigate the role of Calmodulin-dependent protein kinase IV (CaMKIV) in Cardiotoxin (CTX)-induced mice muscle inflammation. CTX injection i.m. was performed to induce B6 mice acute tibialis anterior (TA) muscle injury. The mice were then injected i.p. with the recombinant CaMKIV protein or its antagonist KN-93. Immunoblotting was used to assess Calmodulin (CaM) and CaMKIV levels. Immunofluorescence was used to detect intramuscular infiltration or major histocompatibility complex (MHC)-I expression in damaged muscle. The extent of infiltration was evaluated by fluorescent intensity analysis. Cytokines/chemokines levels were determined by qPCR. CaMKIV gene knockdown in C2C12 cells was performed in order to evaluate the effects of CaMKIV on immuno-behavior of muscle cells. CTX administration induced a strong up-regulation of CaM and p-CaMKIV levels in infiltrated mononuclear cells and regenerated myofibers. In vivo adding of the recombinant CaMKIV protein enhanced intramuscular infiltration of monocytes/macrophages in damaged muscle and increased the number of proinflammatory Ly-6C+F4/80+ macrophage cells. CaMKIV protein treatment induced a striking up-regulation of mRNA levels of IL-1, IL-6, MCP-1, and MCP-3 in CD45+ cells sorted from damaged muscle; increased the infiltration of CD8+ T cells; and induced the up-regulation of MHC-I in partial regenerated myofibers, which was rarely observed in muscle damage alone. Additionally, CaMKIV protein treatment diminished the regulatory T cells (Tregs) number and led to the damaged TA muscle repair delay. In vitro CaMKIV gene knockdown reversed IFN-γ-induced up-regulation of MHC-I/II and TLR3 in the differentiated C2C12 myotubes. CaMKIV can act as an immunostimulation molecule and enhances the acute muscle inflammatory responses.


Assuntos
Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/metabolismo , Desenvolvimento Muscular , Músculo Esquelético/enzimologia , Mioblastos Esqueléticos/enzimologia , Miosite/enzimologia , Regeneração , Doença Aguda , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular , Quimiocinas/genética , Quimiocinas/metabolismo , Quimiotaxia de Leucócito , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Monócitos/metabolismo , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Mioblastos Esqueléticos/imunologia , Mioblastos Esqueléticos/patologia , Miosite/induzido quimicamente , Miosite/genética , Miosite/imunologia , Transdução de Sinais , Fatores de Tempo
3.
J Physiol Biochem ; 73(1): 49-57, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27718123

RESUMO

Excessive exercise induces an inflammatory response caused by oxidative stress, which delays recovery of damaged muscle fibers. The reduction of inflammatory response is important for skeletal muscle homeostasis. Peroxisome proliferator-activated receptor gamma (PPARγ) is an anti-inflammatory molecule, but the role of PPARγ in skeletal muscle as anti-inflammatory activity is not clear. Thus, this study examined the anti-inflammatory role of PPARγ against H2O2-induced oxidative stress in skeletal muscle. Sprague Dawley (SD) rats were exercised on a treadmill to induce oxidative stress. In vitro oxidative stress was evaluated in differentiated C2C12 cells stimulated using 200 µM H2O2. Inflammation-related molecules were determined by immunohistochemistry and Western blot analysis. Expressions of the inflammatory molecules tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), cyclooxygenase-2 (COX-2), and matrix metalloproteinase-2 (MMP-2) in muscles of the acute exercise group were highly increased. PPARγ was also highly expressed in these muscles. These inflammatory molecules were also markedly increased in C2C12 cells with H2O2 stimulation. However, PPARγ overexpression in C2C12 transfected by Ad/PPARγ dramatically reduced the inflammatory molecules. PPARγ also enhanced the anti-oxidants molecules like Cu/Zn-SOD, Mn-SOD, and hemeoxygenase-1 by reducing the generation of ROS, even in the presence of H2O2. PPARγ displayed dual anti-inflammatory and anti-oxidant roles by inhibiting the mitogen-activated protein kinase (MAPK) pathway and translocation of nuclear transcriptional factor-κB (NF-κB) from the cytosol to the nucleus. These results demonstrate a potential role of PPARγ in protecting muscle fibers against oxidative stress caused by excessive acute exercise due to its anti-inflammatory and anti-oxidant activity exerted by inhibition of the MAPK/NF-κB pathway.


Assuntos
Sistema de Sinalização das MAP Quinases , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Miosite/metabolismo , NF-kappa B/antagonistas & inibidores , PPAR gama/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Linhagem Celular , Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/metabolismo , Humanos , Mediadores da Inflamação/agonistas , Mediadores da Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metaloproteinase 2 da Matriz/química , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Atividade Motora , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/imunologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/imunologia , Miosite/induzido quimicamente , Miosite/etiologia , Miosite/imunologia , NF-kappa B/metabolismo , Oxidantes/toxicidade , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/agonistas , PPAR gama/genética , Esforço Físico , Distribuição Aleatória , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
4.
N Biotechnol ; 33(5 Pt A): 514-23, 2016 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-26773739

RESUMO

Biological processes that are mediated by cell-cell interactions in heterogeneous populations are best approached by methods that have single cell resolution. Most of these methods rely on the preparation, from solid tissues, of cell suspensions by enzymatic digestion, followed by analysis of single cell reactivity to an antibody panel that allows the discrimination of cell populations and characterization of their activation state. Thus for any specific biological problem, both efficient and at the same time mild, protocols for cell separation, together with tissue specific panels of antibodies, need to be developed and optimized. Here we characterize an antibody panel that permits the discrimination of mononuclear muscle cell populations by mass cytometry and use it to characterize the cell populations obtained by three different cell extraction procedures from muscle fibers. We show that our panel of antibodies, albeit limited and incomplete, is sufficient to discriminate most of the mononuclear muscle cell populations and that each cell extraction method yields heterogeneous cell populations with a different relative abundance of the distinct cell types.


Assuntos
Separação Celular/métodos , Músculo Esquelético/citologia , Animais , Anticorpos , Biotecnologia , Diferenciação Celular , Citometria de Fluxo/métodos , Espectrometria de Massas/métodos , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/imunologia , Músculo Esquelético/imunologia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/imunologia , Análise de Célula Única/métodos
5.
Hum Mol Genet ; 23(10): 2604-17, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24368419

RESUMO

Although the cause of Duchenne muscular dystrophy (DMD) is known, the specific factors that initiate and perpetuate disease progression are not well understood. We hypothesized that leaky dystrophin-deficient skeletal muscle releases endogenous danger signals (TLR ligands), which bind to Toll-like receptors (TLRs) on muscle and immune cells and activate downstream processes that facilitate degeneration and regeneration in dystrophic skeletal muscle. Here, we demonstrate that dystrophin-deficient mouse muscle cells show increased expression of several cell-surface and endosomal TLRs. In vitro screening identified ssRNA as a relevant endogenous TLR7 ligand. TLR7 activation led to myd88-dependent production of pro-inflammatory cytokines in dystrophin-deficient muscle cells, and cause significant degeneration/regeneration in vivo in mdx mouse muscle. Also, knockout of the central TLR adaptor protein, myd88 in mdx mice significantly improved skeletal and cardiac muscle function. Likewise, proof-of-concept experiments showed that treating young mdx mice with a TLR7/9 antagonist significantly reduced skeletal muscle inflammation and increased muscle force, suggesting that blocking this pathway may have therapeutic potential for DMD.


Assuntos
Glicoproteínas de Membrana/fisiologia , Músculo Esquelético/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Miocárdio/metabolismo , Receptor 7 Toll-Like/fisiologia , Receptor Toll-Like 9/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Distrofina/deficiência , Feminino , Humanos , Masculino , Glicoproteínas de Membrana/agonistas , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Mioblastos Esqueléticos/imunologia , Mioblastos Esqueléticos/metabolismo , Miocárdio/patologia , Fenótipo , Receptor 7 Toll-Like/agonistas
6.
Mol Cell Biol ; 34(4): 606-18, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24298022

RESUMO

The myostatin/activin type II receptor (ActRII) pathway has been identified to be critical in regulating skeletal muscle size. Several other ligands, including GDF11 and the activins, signal through this pathway, suggesting that the ActRII receptors are major regulatory nodes in the regulation of muscle mass. We have developed a novel, human anti-ActRII antibody (bimagrumab, or BYM338) to prevent binding of ligands to the receptors and thus inhibit downstream signaling. BYM338 enhances differentiation of primary human skeletal myoblasts and counteracts the inhibition of differentiation induced by myostatin or activin A. BYM338 prevents myostatin- or activin A-induced atrophy through inhibition of Smad2/3 phosphorylation, thus sparing the myosin heavy chain from degradation. BYM338 dramatically increases skeletal muscle mass in mice, beyond sole inhibition of myostatin, detected by comparing the antibody with a myostatin inhibitor. A mouse version of the antibody induces enhanced muscle hypertrophy in myostatin mutant mice, further confirming a beneficial effect on muscle growth beyond myostatin inhibition alone through blockade of ActRII ligands. BYM338 protects muscles from glucocorticoid-induced atrophy and weakness via prevention of muscle and tetanic force losses. These data highlight the compelling therapeutic potential of BYM338 for the treatment of skeletal muscle atrophy and weakness in multiple settings.


Assuntos
Receptores de Activinas Tipo II/imunologia , Ativinas/metabolismo , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Hipertrofia/metabolismo , Mioblastos Esqueléticos/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animais , Anticorpos Bloqueadores/metabolismo , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Atrofia/imunologia , Atrofia/metabolismo , Diferenciação Celular/fisiologia , Humanos , Hipertrofia/patologia , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mioblastos Esqueléticos/imunologia , Transdução de Sinais/fisiologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo
7.
J Immunol ; 191(1): 407-14, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23729446

RESUMO

The outstanding regeneration ability of skeletal muscle is based on stem cells that become activated and develop to myoblasts after myotrauma. Proliferation and growth of myoblasts result in self-renewal of skeletal muscle. In this article, we show that myotrauma causes a hypoxic microenvironment leading to accumulation of the transcription factor hypoxia-inducible factor-1 (HIF-1) in skeletal muscle cells, as well as invading myeloid cells. To evaluate the impact of HIF-1 in skeletal muscle injury and repair, we examined mice with a conditional HIF-1α knockout targeted to skeletal muscle or myeloid cells in a model of soft tissue trauma. No differences in acute trauma size were detected between control and HIF-1α knockout mice. However, muscles of myeloid HIF-1α knockout mice showed a significant delay in myoblast proliferation and growth of regenerating myofibers, in association with decreased expression of cyclooxygenase-2 in HIF-1α-deficient myeloid cells. Moreover, the removal of necrotic cell debris and the regeneration of endothelial cell structure were impaired in myeloid HIF-1α knockout mice that showed delayed invasion of macrophages to the injury site. Our findings for the first time, to our knowledge, demonstrate that myeloid HIF-1α is required for adequate skeletal muscle regeneration.


Assuntos
Regulação para Baixo/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Músculo Esquelético/imunologia , Células Mieloides/imunologia , Regeneração/imunologia , Regulação para Cima/imunologia , Animais , Regulação para Baixo/genética , Membro Posterior , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/citologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Mioblastos Esqueléticos/imunologia , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patologia , Regeneração/genética , Regulação para Cima/genética , Cicatrização/genética , Cicatrização/imunologia
8.
Transplantation ; 91(4): 425-31, 2011 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-21200367

RESUMO

BACKGROUND: The limitations of syngenic cell therapy include patient safety and quality control of the source cells. Therefore, it is important to develop and assess procedures using allogenic cells. We investigated the impact of allogenic skeletal myoblast (SMB) transplantation on acute myocardial infarction with respect to immune response, donor cell survival, and therapeutic efficacy. METHODS: Female Lewis rats underwent proximal left anterior descending coronary artery ligation. Fifteen minutes later, they underwent major histocompatibility (MHC)-matched Lewis SMB transplantation (group S) and MHC-mismatched ACI SMB transplantation (group A), or treated with buffer injection as a control (group C). RESULTS: Flow cytometry showed that the SMBs expressed MHC antigens and B7 signal molecules in vitro. In group A, transcription levels of interleukin-2 receptor and interferon-γ were significantly increased 7 days after transplantation, and the area surrounding the donor SMBs was intensely infiltrated with CD4- and CD8-positive cells. Estimation of the number of donor cells in the recipient left ventricular chamber revealed that except for day 0, group A had fewer donor SMBs, which disappeared faster, compared with group S. Echocardiography demonstrated that the ejection fraction (EF) of group A was lower than that of group S. CONCLUSION: MHC-mismatched allogenic SMB transplantation in infarcted myocardium induces the immune response and acceleration of donor cell clearance, decreasing the therapeutic effect. Donor cell survival and inflammation may play important roles in the therapeutic mechanism of SMB transplantation therapy for acute myocardial infarction.


Assuntos
Mioblastos Esqueléticos/transplante , Infarto do Miocárdio/terapia , Animais , Antígeno B7-1/imunologia , Antígeno B7-2/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Interferon gama/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Mioblastos Esqueléticos/imunologia , Infarto do Miocárdio/imunologia , Ratos , Ratos Endogâmicos Lew , Receptores de Interleucina-2/imunologia , Volume Sistólico/imunologia , Transplante Homólogo
9.
J Neuroimmunol ; 215(1-2): 73-83, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19766326

RESUMO

This study explores the potential of non-neural progenitor cells for CNS cell therapy. Muscle progenitor cells (MPCs), transplanted either intraventricularly or intraperitonealy, incorporated into the CNS of EAE-induced but not of naïve mice. Some of the migrating MPCs expressed the neuronal marker beta-III-Tubulin and gained neuronal morphology. Co-treatment of transplanted mice with the immunomodulatory agent glatiramer acetate (GA, Copaxone) resulted in improved MPCs incorporation and differentiation towards the neuronal pathway. The therapeutic potential of myogenic progenitor cells was demonstrated by amelioration of clinical symptoms and reduced mortality in EAE mice, as well as by expression of IL-10, TGF-beta, and the neurotrophin-BDNF.


Assuntos
Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/terapia , Transplante de Células-Tronco Mesenquimais , Mioblastos Esqueléticos/imunologia , Mioblastos Esqueléticos/transplante , Animais , Biomarcadores/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Células Cultivadas , Sistema Nervoso Central/citologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Acetato de Glatiramer , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mioblastos Esqueléticos/efeitos dos fármacos , Peptídeos/administração & dosagem , Ratos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/imunologia
10.
Am J Physiol Endocrinol Metab ; 296(6): E1300-10, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19336660

RESUMO

Obesity is characterized by adipose tissue expansion as well as macrophage infiltration of adipose tissue. This results in an increase in circulating inflammatory cytokines and nonesterified fatty acids, factors that cause skeletal muscle insulin resistance. Whether obesity also results in skeletal muscle inflammation is not known. In this study, we quantified macrophages immunohistochemically in vastus lateralis biopsies from eight obese and eight lean subjects. Our study demonstrates that macrophages infiltrate skeletal muscle in obesity, and we developed an in vitro system to study this mechanistically. Myoblasts were isolated from vastus lateralis biopsies and differentiated in culture. Coculture of differentiated human myotubes with macrophages in the presence of palmitic acid, to mimic an obese environment, revealed that macrophages in the presence of palmitic acid synergistically augment cytokine and chemokine expression in myotubes, decrease IkappaB-alpha protein expression, increase phosphorylated JNK, decrease phosphorylated Akt, and increase markers of muscle atrophy. These results suggest that macrophages alter the inflammatory state of muscle cells in an obese milieu, inhibiting insulin signaling. Thus in obesity both adipose tissue and skeletal muscle inflammation may contribute to insulin resistance.


Assuntos
Ácidos Graxos não Esterificados/metabolismo , Resistência à Insulina/imunologia , Macrófagos/imunologia , Mioblastos Esqueléticos/imunologia , Miosite/imunologia , Obesidade/imunologia , Adulto , Comunicação Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Citocinas/genética , Citocinas/metabolismo , Ácidos Graxos não Esterificados/farmacologia , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Expressão Gênica/imunologia , Humanos , Insulina/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/imunologia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Miosite/metabolismo , Miosite/patologia , Obesidade/metabolismo , Ácido Palmítico/farmacologia , Transdução de Sinais/imunologia , Adulto Jovem
11.
J Appl Physiol (1985) ; 106(1): 130-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18974369

RESUMO

We tested the hypothesis that cytokines derived from differentiated skeletal muscle cells in culture induce neutrophil chemotaxis after mechanical strain. Flexible-bottom plates with cultured human muscle cells attached were exposed to mechanical strain regimens (ST) of 0, 10, 30, 50, or 70 kPa of negative pressure. Conditioned media were tested for the ability to induce chemotaxis of human blood neutrophils in vitro and for a marker of muscle cell injury (lactate dehydrogenase). Conditioned media promoted neutrophil chemotaxis in a manner that was related both to the degree of strain and to the magnitude of muscle cell injury (ST 70 > ST 50 > ST 30). Protein profiling using a multiplex cytokine assay revealed that mechanical strain increased the presence of IL-8, granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor, monocyte chemotactic protein (MCP)-1, and IL-6 in conditioned media. We also detected 14 other cytokines in conditioned media from control cultures that did not respond to mechanical strain. Neutralization of IL-8 and GM-CSF completely inhibited the chemotactic response for ST 30 and ST 50 and reduced the chemotactic response for ST 70 by 40% and 47%, respectively. Neutralization of MCP-1 or IL-6 did not reduce chemotaxis after ST 70. This study enhances our understanding of the immunobiology of skeletal muscle by revealing that skeletal muscle cell-derived IL-8 and GM-CSF promote neutrophil chemotaxis after injurious mechanical strain.


Assuntos
Quimiotaxia de Leucócito , Citocinas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-8/metabolismo , Mioblastos Esqueléticos/imunologia , Neutrófilos/imunologia , Comunicação Parácrina , Sobrevivência Celular , Células Cultivadas , Quimiocina CCL2/metabolismo , Meios de Cultivo Condicionados/metabolismo , Feminino , Humanos , Interleucina-6/metabolismo , L-Lactato Desidrogenase/metabolismo , Mioblastos Esqueléticos/enzimologia , Mioblastos Esqueléticos/patologia , Análise Serial de Proteínas , Estresse Mecânico
12.
Ann Thorac Surg ; 86(6): 1841-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19021990

RESUMO

BACKGROUND: We sought to compare host immune cell kinetics, survival profile of donor skeletal myoblasts, and skeletal myoblast graft efficacy after autologous and allogeneic skeletal myoblast transplantation into a rat model of myocardial infarction. METHODS: One week after myocardial infarction, 128 animals were divided into four groups: group 1 (n = 24, receiving medium only), group 2 (n = 24, receiving medium and cyclosporine), group 3 (n = 40, autologous skeletal myoblast transplantation), and group 4 (n = 40, allogeneic skeletal myoblast transplantation with cyclosporine treatment). Rats were euthanized 10 minutes, 1 day, and 4, 7, and 28 days later. Host immune cell kinetics were assessed by immunohistochemical studies for macrophages, and CD4+ and CD8+ lymphocytes. Donor skeletal myoblast survival was confirmed by tracking prelabeled signals, and quantified by beta-gal assay. Heart function was evaluated by echocardiography. RESULTS: A transient immune cell infiltration was demonstrated in group 3, with macrophage infiltration on day 1 and day 4, CD8+ cell infiltration on day 4 and day 7, and CD4+ cell infiltration on day 4. In group 4, immunocyte infiltration was slightly more severe than that in group 3. Automyoblasts and allomyoblasts showed no significant difference of survival from day 1 to day 7 (p > 0.10); however, on day 28, automyoblasts showed better survival than allomyoblasts (p < 0.05). Transplantation of allomyoblasts increased systolic heart function and limited heart dilation after myocardial injury to a similar degree as automyoblasts (p > 0.10). CONCLUSIONS: The use of allomyoblasts is feasible and effective for cardiac repair with immunosuppressive treatment as compared with automyoblasts.


Assuntos
Transplante de Células/métodos , Ciclosporinas/farmacologia , Mioblastos Esqueléticos/transplante , Infarto do Miocárdio/cirurgia , Animais , Modelos Animais de Doenças , Feminino , Rejeição de Enxerto , Sobrevivência de Enxerto , Imuno-Histoquímica , Imunossupressores/farmacologia , Masculino , Mioblastos Esqueléticos/imunologia , Infarto do Miocárdio/patologia , Probabilidade , Distribuição Aleatória , Ratos , Ratos Wistar , Regeneração/fisiologia , Fatores de Risco , Sensibilidade e Especificidade , Transplante Autólogo , Transplante Homólogo , Remodelação Ventricular/fisiologia
13.
Tsitologiia ; 49(11): 944-51, 2007.
Artigo em Russo | MEDLINE | ID: mdl-18217362

RESUMO

The investigation of antigenic diversion of tumor cells resulting from the expression of heteroorganic antigens has been continued. Tumor-associated heteroorganic antigens with mol. weight 200-210 kDa (identified before as laminin), 105-130, 75-80 and 43 kDa were detected by anti-kidney serum in fractions of plasmatic membranes of cells of rat ascitic Zajdela hepatoma and cultured HTC hepatoma; the antigen 43 kDa was isolated on immunosorbent and identified by mass spectrometry as beta-actin. Anti-kidney serum revealed laminin in fractions of plasmatic membranes of cultured L8 and L6J1 myoblasts, and L6J1 myotubes; apparently, synthesis of laminin by hepatoma and myogenic cells is not connected with their proliferative activity. Besides, anti-kidney serum detected components 38, 42, 44, 48, 62, 78 and 120 kDa, expression of which on myogenic cells surface might be consequence of active cell proliferation and (or) differentiation.


Assuntos
Antígenos de Neoplasias/imunologia , Rim/imunologia , Neoplasias Hepáticas Experimentais/imunologia , Actinas/imunologia , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Antígenos de Neoplasias/genética , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Membrana Celular/imunologia , Proliferação de Células , Transformação Celular Neoplásica/imunologia , Técnica Indireta de Fluorescência para Anticorpo , Immunoblotting , Laminina/imunologia , Masculino , Dados de Sequência Molecular , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/imunologia , Especificidade de Órgãos , Ratos
14.
Cell Transplant ; 14(7): 457-67, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16285254

RESUMO

The development of an optimized animal model for the in vivo analysis of human muscle cells remains an important goal in the search of therapy for muscular dystrophy. Here we examined the efficiency of human myoblast xenografts in three distinct immunodeficient mouse models. We found that different conditioning regimes used to provoke host muscle regeneration (i.e., cardiotoxin versus cryodamage) had a marked impact on xenograft success. Tibialis anterior muscle of Rag2-, Rag-/gammac-, and Rag-/gammac-/C5- mice was treated by cardiotoxin or cryodamage, submitted to enzymatic digestion, and analyzed by cytofluorometry to quantitate inflammatory cells. Human myoblasts were injected into pretreated muscles from immunodeficient recipients and the cell engraftment evaluated by immunocytochemistry, 4-8 weeks after transplantation. Donor cell differentiation and dispersion within the host muscles was also investigated. Host regeneration in cardiotoxin-treated mice was accompanied by a higher inflammatory cell infiltration when compared to that induced by cryodamage. Accordingly, when compared to the cardiotoxin group, more human myogenic cells were found after cryodamage. When the distinct immunodeficient mice were compared, we found that the alymphoid strain lacking the complement component C5 (Rag-/gammac-/C5- mice) was the most efficient host for human muscle xenografts, when compared with C5(+)Rag-/gammac- mice or Rag- mice. Our results demonstrate that cryolesion-conditioned muscles of Rag-/gammac-/C5- mice provide the best environment for long-term in vivo human myoblast differentiation, opening the way for a novel approach to study the pathophysiology of human muscle disorders.


Assuntos
Diferenciação Celular , Modelos Animais , Mioblastos Esqueléticos/transplante , Regeneração/efeitos dos fármacos , Tíbia/citologia , Tíbia/fisiologia , Animais , Humanos , Recém-Nascido , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/imunologia , Regeneração/imunologia , Tíbia/imunologia
15.
Hum Gene Ther ; 15(7): 627-36, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15242523

RESUMO

Cellular cardiomyoplasty using skeletal myoblasts may be beneficial for infarct repair. One drawback to skeletal muscle cells is their lack of gap junction expression after differentiation, thus preventing electrical coupling to host cardiomyocytes. We sought to overexpress the gap junction protein connexin43 (Cx43) in differentiated skeletal myotubes, using retroviral, adenoviral, and plasmid-mediated gene transfer. All strategies resulted in overexpression of Cx43 in cultured myotubes, but expression of Cx43 from constitutive viral promoters caused significant death upon differentiation. Dye transfer studies showed that surviving myotubes contained functional gap junctions, however. Retrovirally transfected myoblasts did not express Cx43 after grafting into the heart, possibly due to promoter silencing. Adenovirally transfected myoblasts expressed abundant Cx43 after forming myotubes in cardiac grafts, but grafts showed signs of injury at 1 week and had died by 2 weeks. Interestingly, transfection of already differentiated myotubes with adenoviral Cx43 was nontoxic, implying a window of vulnerability during differentiation. To test this hypothesis, Cx43 was expressed from the muscle creatine kinase (MCK) promoter, which is active only after myocyte differentiation. The MCK promoter resulted in high levels of Cx43 expression in differentiated myotubes but did not cause cell death during differentiation. MCK-Cx43-transfected myoblasts formed viable cardiac grafts and, in some cases, Cx43-expressing myotubes were in close apposition to host cardiomyocytes, possibly allowing electrical coupling. Thus, high levels of Cx43 during skeletal muscle differentiation cause cell death. When, however, expression of Cx43 is delayed until after differentiation, using the MCK promoter, myotubes are viable and express gap junction proteins after grafting in the heart. This strategy may permit electrical coupling of skeletal and cardiac muscle for cardiac repair.


Assuntos
Conexina 43/genética , Mioblastos Esqueléticos/metabolismo , Adenoviridae/genética , Animais , Diferenciação Celular , Conexina 43/análise , Conexina 43/biossíntese , Creatina Quinase/genética , Creatina Quinase Forma MM , Junções Comunicantes/genética , Junções Comunicantes/metabolismo , Humanos , Isoenzimas/genética , Camundongos , Fibras Musculares Esqueléticas/imunologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/imunologia , Mioblastos Esqueléticos/transplante , Miocárdio/imunologia , Miocárdio/metabolismo , Regiões Promotoras Genéticas/genética , Ratos , Transfecção
16.
Exp Cell Res ; 291(1): 83-90, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14597410

RESUMO

Skeletal muscle-derived CD34+/45- (Sk-34) cells were identified as a new candidate for stem cells. However, the relationship between Sk-34 cells and side-population (SP) cells is unknown. Here, we demonstrate that Sk-34 cells prepared from murine skeletal muscles consist wholly of main-population (MP) cells. The Sk-34 cells included only a few SP cells (1:1000, SP:MP). Colony-forming units of Sk-34 cells of both SP and MP possessed the same potential to differentiate into adipocytes, endothelial, and myogenic cells and showed the same colony-forming activity (1.6%). In addition, the colony-forming units of the CD34-/45- (double negative: DN) population were found to begin CD34 expression and to possess the potential to differentiate into myogenic and endothelial cells. We also found that expression of CD34 antigen precedes MyoD expression during the myogenic process of DN cells. Furthermore, both Sk-34 and DN cell populations were mostly negative for CD73 (93-95%), whereas the CD45+ cell population was >25% positive for CD73, and this trend was also seen in bone marrow-derived CD45+ cells. These results indicate that the MP cell population is about 99.9% responsible for the reported in vitro myogenic-endothelial responses of skeletal muscle-derived cells.


Assuntos
Diferenciação Celular/imunologia , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/citologia , Músculo Esquelético/crescimento & desenvolvimento , Mioblastos Esqueléticos/citologia , Células-Tronco/citologia , 5'-Nucleotidase/metabolismo , Adipócitos/citologia , Adipócitos/imunologia , Adipócitos/metabolismo , Animais , Antígenos CD34/imunologia , Antígenos CD34/metabolismo , Divisão Celular/fisiologia , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Imuno-Histoquímica , Antígenos Comuns de Leucócito/imunologia , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/imunologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/imunologia , Proteína MyoD/metabolismo , Mioblastos Esqueléticos/imunologia , Mioblastos Esqueléticos/metabolismo , Transplante de Células-Tronco/métodos , Células-Tronco/imunologia , Células-Tronco/metabolismo
17.
J Neuroimmunol ; 137(1-2): 125-33, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12667656

RESUMO

We studied the production of interleukin (IL)-6 and CCL20/macrophage inflammatory protein-3 alpha (MIP-3 alpha) by human myoblasts and muscle samples in response to IL-17 alone or in combination with IL-1 beta. Both IL-17 and IL-1 beta induced IL-6 production by normal myoblasts and muscle samples. IL-17 had no effect on CCL20 production by myoblasts. Combination of IL-17 and IL-1 beta further increased IL-6 and CCL20 production by muscle samples but not that of CK. IL-17 induced also HLA class I, C-Fos, nuclear factor kappa B (NF-kappa B) and C-Jun expression by myoblasts but not that of HLA class II, CD40, vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1). Finally, immunostaining of dermatomyositis (DM) and polymyositis (PM) muscle biopsies showed IL-17 and CCL20 expression. Our study shows that low levels of cytokines produced by T cells (IL-17) and monocytes (IL-1 beta) can act in combination on skeletal myoblasts and muscle tissue.


Assuntos
Interleucina-17/fisiologia , Interleucina-1/fisiologia , Músculo Esquelético/imunologia , Miosite/imunologia , Linfócitos T/imunologia , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/fisiologia , Citocinas/biossíntese , Combinação de Medicamentos , Sinergismo Farmacológico , Humanos , Interleucina-1/farmacologia , Interleucina-17/farmacologia , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/imunologia , Mioblastos Esqueléticos/metabolismo , Miosite/metabolismo , Linfócitos T/metabolismo
18.
J Neuroimmunol ; 134(1-2): 151-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12507783

RESUMO

We have previously shown that interleukin (IL)-1 beta and other inflammatory cytokines are able to induce the expression of membrane and soluble intercellular adhesion molecule (ICAM)-1 on human myoblasts. In this paper we found that IL-10 and transforming growth factor (TGF)-beta 1 are able to prevent IL-1 beta-induced membrane and soluble ICAM-1 protein expression on human myoblasts, with different time courses. The effect of both cytokines is associated to a reduction in ICAM-1 mRNA. Our findings suggest that IL-10 and TGF-beta 1 are able to influence the inflammatory process in muscle tissue at least in part by means of control of membrane and soluble ICAM-1.


Assuntos
Rejeição de Enxerto/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Interleucina-10/metabolismo , Interleucina-1/imunologia , Mioblastos Esqueléticos/imunologia , Miosite/imunologia , Fator de Crescimento Transformador beta/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/fisiopatologia , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-1/metabolismo , Interleucina-10/imunologia , Interleucina-10/farmacologia , Células Musculares/efeitos dos fármacos , Células Musculares/imunologia , Células Musculares/metabolismo , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/metabolismo , Miosite/metabolismo , Miosite/fisiopatologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/imunologia , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1
19.
Int Immunol ; 14(8): 917-24, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12147628

RESUMO

In polymyositis (PM)/dermatomyositis (DM), various cytokines, especially macrophage-derived cytokines such as IL-1alpha, IL-1beta and tumor necrosis factor (TNF)-alpha, are expressed in the inflammatory foci. We previously reported that IL-15, a novel cytokine with a biological activity similar to that of IL-2, is expressed in muscle cells in PM/DM. In the present study, we set out to investigate the regulation of IL-15 in cultured myoblasts. Myoblasts constitutively produced a low level of IL-15 and the production was augmented by stimulation with IFN-gamma, IL-1alpha, IL-1beta, TNF-alpha or lipopolysaccharide (LPS) in a dose-dependent manner. These stimuli also enhanced the expression of IL-15 mRNA. About 30-40% of IL-15 was detected intracellularly, while the rest was released into the culture supernatant. Immunohistochemical staining revealed that intracellular IL-15 was localized in the perinuclear area of the cytoplasm in the myoblasts. Despite the considerable amounts of intracellular IL-15, the myoblasts predominantly expressed authentic IL-15 mRNA isoform. This isoform generates IL-15 with long signal peptide preprotein, which is all to be secreted. The biological activity of IL-15 secreted from the myoblasts was examined using an IL-15-dependent murine T cell line, CTLL-2. Culture supernatants of the myoblasts induced a proliferative response of CTLL-2 and this was specifically inhibited by anti-IL-15 antibody. These results suggest that inflammatory stimuli induce the production of IL-15 in the muscle cells in PM/DM, and IL-15 may contribute to the immunopathogenesis by augmenting recruitment and activation of the infiltrating T cells. Blocking of IL-15 production might be of therapeutic value in PM/DM.


Assuntos
Dermatomiosite/imunologia , Interleucina-15/biossíntese , Mioblastos Esqueléticos/imunologia , Polimiosite/imunologia , Animais , Estudos de Casos e Controles , Linhagem Celular , Células Cultivadas , Citocinas/farmacologia , Dermatomiosite/genética , Expressão Gênica , Humanos , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Interleucina-15/genética , Lipopolissacarídeos/farmacologia , Ativação Linfocitária , Camundongos , Mioblastos Esqueléticos/efeitos dos fármacos , Polimiosite/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA