Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(15): e2111445119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35377804

RESUMO

Volumetric muscle loss (VML) overwhelms the innate regenerative capacity of mammalian skeletal muscle (SkM), leading to numerous disabilities and reduced quality of life. Immune cells are critical responders to muscle injury and guide tissue resident stem cell­ and progenitor-mediated myogenic repair. However, how immune cell infiltration and intercellular communication networks with muscle stem cells are altered following VML and drive pathological outcomes remains underexplored. Herein, we contrast the cellular and molecular mechanisms of VML injuries that result in the fibrotic degeneration or regeneration of SkM. Following degenerative VML injuries, we observed the heightened infiltration of natural killer (NK) cells as well as the persistence of neutrophils beyond 2 wk postinjury. Functional validation of NK cells revealed an antagonistic role in neutrophil accumulation in part via inducing apoptosis and CCR1-mediated chemotaxis. The persistent infiltration of neutrophils in degenerative VML injuries was found to contribute to impairments in muscle stem cell regenerative function, which was also attenuated by transforming growth factor beta 1 (TGFß1). Blocking TGFß signaling reduced neutrophil accumulation and fibrosis and improved muscle-specific force. Collectively, these results enhance our understanding of immune cell­stem cell cross talk that drives regenerative dysfunction and provide further insight into possible avenues for fibrotic therapy exploration.


Assuntos
Células Matadoras Naturais , Músculo Esquelético , Doenças Musculares , Neutrófilos , Regeneração , Células Satélites de Músculo Esquelético , Animais , Fibrose , Células Matadoras Naturais/imunologia , Camundongos , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Doenças Musculares/imunologia , Doenças Musculares/patologia , Infiltração de Neutrófilos , Neutrófilos/imunologia , Regeneração/imunologia , Células Satélites de Músculo Esquelético/imunologia , Fator de Crescimento Transformador beta/metabolismo
2.
Aging Cell ; 20(2): e13312, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33511781

RESUMO

Recruited immune cells play a critical role in muscle repair, in part by interacting with local stem cell populations to regulate muscle regeneration. How aging affects their communication during myogenesis is unclear. Here, we investigate how aging impacts the cellular function of these two cell types after muscle injury during normal aging or after immune rejuvenation using a young to old (Y-O) or old to old (O-O) bone marrow (BM) transplant model. We found that skeletal muscle from old mice (20 months) exhibited elevated basal inflammation and possessed fewer satellite cells compared with young mice (3 months). After cardiotoxin muscle injury (CTX), old mice exhibited a blunted inflammatory response compared with young mice and enhanced M2 macrophage recruitment and IL-10 expression. Temporal immune and cytokine responses of old mice were partially restored to a young phenotype following reconstitution with young cells (Y-O chimeras). Improved immune responses in Y-O chimeras were associated with greater satellite cell proliferation compared with O-O chimeras. To identify how immune cell aging affects myoblast function, conditioned media (CM) from activated young or old macrophages was applied to cultured C2C12 myoblasts. CM from young macrophages inhibited myogenesis while CM from old macrophages reduced proliferation. These functional differences coincided with age-related differences in macrophage cytokine expression. Together, this study examines the infiltration and proliferation of immune cells and satellite cells after injury in the context of aging and, using BM chimeras, demonstrates that young immune cells retain cell autonomy in an old host to increase satellite cell proliferation.


Assuntos
Senescência Celular/imunologia , Desenvolvimento Muscular/imunologia , Células Satélites de Músculo Esquelético/imunologia , Animais , Cardiotoxinas/farmacologia , Senescência Celular/efeitos dos fármacos , Camundongos , Desenvolvimento Muscular/efeitos dos fármacos , Células Satélites de Músculo Esquelético/efeitos dos fármacos
3.
Sci Rep ; 10(1): 11119, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32632224

RESUMO

Skeletal muscle stem (satellite) cells transplanted into host mouse muscles contribute to muscle regeneration. Irradiation of host muscle enhances donor stem cell engraftment by promoting the proliferation of transplanted donor cells. We hypothesised that, similar to other systems, cells damaged by radiation might be effecting this donor cell proliferation. But we found no difference in the percentage of dying (TUNEL+) cells in immunodeficient dystrophic mouse muscles at the times after the irradiation dose that enhances donor cell engraftment. Similarly, irradiation did not significantly increase the number of TUNEL+ cells in non-dystrophic immunodeficient mouse muscles and it only slightly enhanced donor satellite cell engraftment in this mouse strain, suggesting either that the effector cells are present in greater numbers within dystrophic muscle, or that an innate immune response is required for effective donor cell engraftment. Donor cell engraftment within non-irradiated dystrophic host mouse muscles was not enhanced if they were transplanted with either satellite cells, or myofibres, derived from irradiated dystrophic mouse muscle. But a mixture of cells from irradiated muscle transplanted with donor satellite cells promoted donor cell engraftment in a few instances, suggesting that a rare, yet to be identified, cell type within irradiated dystrophic muscle enhances the donor stem cell-mediated regeneration. The mechanism by which cells within irradiated host muscle promote donor cell engraftment remains elusive.


Assuntos
Inflamação/terapia , Músculo Esquelético/citologia , Músculo Esquelético/efeitos da radiação , Distrofia Muscular Animal/terapia , Regeneração , Células Satélites de Músculo Esquelético/efeitos da radiação , Células Satélites de Músculo Esquelético/transplante , Animais , Diferenciação Celular , Proliferação de Células , Feminino , Raios gama , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular Animal/imunologia , Células Satélites de Músculo Esquelético/imunologia
4.
Curr Osteoporos Rep ; 18(4): 350-356, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32500480

RESUMO

PURPOSE OF REVIEW: The age-related loss of skeletal muscle and bone tissue decreases functionality and increases the risk for falls and injuries. One contributing factor of muscle and bone loss over time is chronic low-grade inflammation. Exercise training is an effective countermeasure for decreasing the loss of muscle and bone tissue, possibly by enhancing immune system response. Herein, we discuss key interactions between the immune system, muscle, and bone in relation to exercise perturbations, and we identify that there is substantial "cross-talk" between muscle and bone and the immune system in response to exercise. RECENT FINDINGS: Recent advances in our understanding of the "cross-talk" between muscle and bone and the immune system indicate that exercise is likely to mediate many of the beneficial effects on muscle and bone via their interactions with the immune system. The age-related loss of muscle and bone tissue may be partially explained by an impaired immune system via chronic low-grade inflammation. Exercise training has a beneficial effect on immune system function and aging muscle and bone. Theoretically, the "cross-talk" between the immune system, muscle, and bone in response to exercise enhances aging musculoskeletal health.


Assuntos
Envelhecimento/imunologia , Osso e Ossos/imunologia , Citocinas/imunologia , Exercício Físico/fisiologia , Sistema Imunitário/imunologia , Músculo Esquelético/imunologia , Humanos , Macrófagos/imunologia , Células Satélites de Músculo Esquelético/imunologia
5.
Nat Commun ; 10(1): 1364, 2019 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-30910999

RESUMO

The mechanisms linking muscle injury and regeneration are not fully understood. Here we report an unexpected role for ZEB1 regulating inflammatory and repair responses in dystrophic and acutely injured muscles. ZEB1 is upregulated in the undamaged and regenerating myofibers of injured muscles. Compared to wild-type counterparts, Zeb1-deficient injured muscles exhibit enhanced damage that corresponds with a retarded p38-MAPK-dependent transition of their macrophages towards an anti-inflammatory phenotype. Zeb1-deficient injured muscles also display a delayed and poorer regeneration that is accounted by the retarded anti-inflammatory macrophage transition and their intrinsically deficient muscle satellite cells (MuSCs). Macrophages in Zeb1-deficient injured muscles show lower phosphorylation of p38 and its forced activation reverts the enhanced muscle damage and poorer regeneration. MuSCs require ZEB1 to maintain their quiescence, prevent their premature activation following injury, and drive efficient regeneration in dystrophic muscles. These data indicate that ZEB1 protects muscle from damage and is required for its regeneration.


Assuntos
Músculo Esquelético/metabolismo , Distrofias Musculares/genética , RNA Mensageiro/genética , Regeneração/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Cromonas/farmacologia , Modelos Animais de Doenças , Flavonoides/farmacologia , Regulação da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/imunologia , Laminina/genética , Laminina/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Morfolinas/farmacologia , Músculo Esquelético/imunologia , Músculo Esquelético/lesões , Distrofias Musculares/imunologia , Distrofias Musculares/patologia , Fenótipo , Fosforilação , RNA Mensageiro/imunologia , Regeneração/imunologia , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia , Transdução de Sinais , Homeobox 1 de Ligação a E-box em Dedo de Zinco/deficiência , Homeobox 1 de Ligação a E-box em Dedo de Zinco/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
6.
Int J Mol Sci ; 19(8)2018 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-30072615

RESUMO

Signal transducer and activator of transcription 3 (STAT3) signaling plays critical roles in regulating skeletal muscle mass, repair, and diseases. In this review, we discuss the upstream activators of STAT3 in skeletal muscles, with a focus on interleukin 6 (IL6) and transforming growth factor beta 1 (TGF-ß1). We will also discuss the double-edged effect of STAT3 activation in the muscles, including the role of STAT3 signaling in muscle hypertrophy induced by exercise training or muscle wasting in cachectic diseases and muscular dystrophies. STAT3 is a critical regulator of satellite cell self-renewal after muscle injury. STAT3 knock out affects satellite cell myogenic progression by impairing proliferation and inducing premature differentiation. Recent studies in STAT3 signaling demonstrated its direct role in controlling myogenic capacity of myoblasts and satellite cells, as well as the potential benefit in using STAT3 inhibitors to treat muscle diseases. However, prolonged STAT3 activation in muscles has been shown to be responsible for muscle wasting by activating protein degradation pathways. It is important to balance the extent of STAT3 activation and the duration and location (cell types) of the STAT3 signaling when developing therapeutic interventions. STAT3 signaling in other tissues and organs that can directly or indirectly affects skeletal muscle health are also discussed.


Assuntos
Interleucina-6/imunologia , Músculo Esquelético/patologia , Doenças Musculares/patologia , Fator de Transcrição STAT3/imunologia , Fator de Crescimento Transformador beta1/imunologia , Animais , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/fisiopatologia , Interleucina-6/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Doenças Musculares/imunologia , Doenças Musculares/metabolismo , Doenças Musculares/fisiopatologia , Fator de Transcrição STAT3/metabolismo , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
7.
FEBS J ; 284(4): 517-524, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27479876

RESUMO

Skeletal muscle regeneration results from the activation and differentiation of myogenic stem cells, called satellite cells, located beneath the basal lamina of the muscle fibers. Inflammatory and immune cells have a crucial role in the regeneration process. Acute muscle injury causes an immediate transient wave of neutrophils followed by a more persistent infiltration of M1 (proinflammatory) and M2 (anti-inflammatory/proregenerative) macrophages. New studies show that injured muscle is also infiltrated by a specialized population of regulatory T (Treg) cells, which control both the inflammatory response, by promoting the M1-to-M2 switch, and the activation of satellite cells. Treg cells accumulate in injured muscle in response to specific cytokines, such as IL-33, and promote muscle growth by releasing growth factors, such as amphiregulin. Muscle repair during aging is impaired due to reduced number of Treg cells and can be enhanced by IL-33 supplementation. Migration of Treg cells could also contribute to explain the effect of heterochronic parabiosis, whereby muscle regeneration of aged mice can be improved by a parabiotically linked young partners. In mdx dystrophin-deficient mice, a model of human Duchenne muscular dystrophy, muscle injury, and inflammation is mitigated by expansion of the Treg-cell population but exacerbated by Treg-cell depletion. These findings support the notion that immunological mechanisms are not only essential in the response to pathogenic microbes and tumor cells but also have a wider homeostatic role in tissue repair, and open new perspectives for boosting muscle growth in chronic muscle disease and during aging.


Assuntos
Distrofia Muscular de Duchenne/imunologia , Regeneração/imunologia , Células Satélites de Músculo Esquelético/imunologia , Linfócitos T Reguladores/imunologia , Envelhecimento , Anfirregulina/genética , Anfirregulina/imunologia , Animais , Movimento Celular , Regulação da Expressão Gênica , Humanos , Interleucina-33/genética , Interleucina-33/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/imunologia , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Neutrófilos/imunologia , Neutrófilos/patologia , Células Satélites de Músculo Esquelético/patologia , Linfócitos T Reguladores/patologia
8.
Cell Death Dis ; 6: e1830, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26203859

RESUMO

Recent evidence has revealed the importance of reciprocal functional interactions between different types of mononuclear cells in coordinating the repair of injured muscles. In particular, signals released from the inflammatory infiltrate and from mesenchymal interstitial cells (also known as fibro-adipogenic progenitors (FAPs)) appear to instruct muscle stem cells (satellite cells) to break quiescence, proliferate and differentiate. Interestingly, conditions that compromise the functional integrity of this network can bias muscle repair toward pathological outcomes that are typically observed in chronic muscular disorders, that is, fibrotic and fatty muscle degeneration as well as myofiber atrophy. In this review, we will summarize the current knowledge on the regulation of this network in physiological and pathological conditions, and anticipate the potential contribution of its cellular components to relatively unexplored conditions, such as aging and physical exercise.


Assuntos
Eosinófilos/metabolismo , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Adipócitos/citologia , Adipócitos/imunologia , Adipócitos/metabolismo , Comunicação Celular , Diferenciação Celular , Eosinófilos/citologia , Eosinófilos/imunologia , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Homeostase , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/patologia , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/imunologia
9.
Biomed Res Int ; 2014: 438675, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25028653

RESUMO

Skeletal muscle is able to restore contractile functionality after injury thanks to its ability to regenerate. Following muscle necrosis, debris is removed by macrophages, and muscle satellite cells (MuSCs), the muscle stem cells, are activated and subsequently proliferate, migrate, and form muscle fibers restoring muscle functionality. In most muscle dystrophies (MDs), MuSCs fail to properly proliferate, differentiate, or replenish the stem cell compartment, leading to fibrotic deposition. However, besides MuSCs, interstitial nonmyogenic cells and inflammatory cells also play a key role in orchestrating muscle repair. A complete understanding of the complexity of these mechanisms should allow the design of interventions to attenuate MDs pathology without disrupting regenerative processes. In this review we will focus on the contribution of immune cells in the onset and progression of MDs, with particular emphasis on Duchenne muscular dystrophy (DMD). We will briefly summarize the current knowledge and recent advances made in our understanding of the involvement of different innate immune cells in MDs and will move on to critically evaluate the possible role of cell populations within the acquired immune response. Revisiting previous observations in the light of recent evidence will likely change our current view of the onset and progression of the disease.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Linfócitos/imunologia , Macrófagos/imunologia , Músculo Esquelético/imunologia , Distrofia Muscular de Duchenne/imunologia , Regeneração/imunologia , Animais , Humanos , Linfócitos/patologia , Macrófagos/patologia , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/patologia
10.
Mol Ther ; 22(5): 1008-17, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24569833

RESUMO

Stem cell therapy is a promising strategy for treatment of muscular dystrophies. In addition to muscle fiber formation, reconstitution of functional stem cell pool by donor cells is vital for long-term treatment. We show here that some CD133(+) cells within human muscle are located underneath the basal lamina of muscle fibers, in the position of the muscle satellite cell. Cultured hCD133(+) cells are heterogeneous and multipotent, capable of forming myotubes and reserve satellite cells in vitro. They contribute to extensive muscle regeneration and satellite cell formation following intramuscular transplantation into irradiated and cryodamaged tibialis anterior muscles of immunodeficient Rag2-/γ chain-/C5-mice. Some donor-derived satellite cells expressed the myogenic regulatory factor MyoD, indicating that they were activated. In addition, when transplanted host muscles were reinjured, there was significantly more newly-regenerated muscle fibers of donor origin in treated than in control, nonreinjured muscles, indicating that hCD133(+) cells had given rise to functional muscle stem cells, which were able to activate in response to injury and contribute to a further round of muscle regeneration. Our findings provide new evidence for the location and characterization of hCD133(+) cells, and highlight that these cells are highly suitable for future clinical application.


Assuntos
Antígenos CD/genética , Terapia Baseada em Transplante de Células e Tecidos , Glicoproteínas/genética , Distrofias Musculares/terapia , Peptídeos/genética , Transplante de Células-Tronco , Antígeno AC133 , Animais , Antígenos CD/biossíntese , Glicoproteínas/biossíntese , Humanos , Camundongos , Distrofias Musculares/genética , Proteína MyoD/biossíntese , Mioblastos/citologia , Mioblastos/imunologia , Mioblastos/metabolismo , Regeneração/genética , Regeneração/imunologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/metabolismo
11.
PLoS One ; 8(8): e70971, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951058

RESUMO

BACKGROUND: Systemic elevations in PAI-1 suppress the fibrinolytic pathway leading to poor collagen remodelling and delayed regeneration of tibialis anterior (TA) muscles in type-1 diabetic Akita mice. However, how impaired collagen remodelling was specifically attenuating regeneration in Akita mice remained unknown. Furthermore, given intrinsic differences between muscle groups, it was unclear if the reparative responses between muscle groups were different. PRINCIPAL FINDINGS: Here we reveal that diabetic Akita muscles display differential regenerative responses with the TA and gastrocnemius muscles exhibiting reduced regenerating myofiber area compared to wild-type mice, while soleus muscles displayed no difference between animal groups following injury. Collagen levels in TA and gastrocnemius, but not soleus, were significantly increased post-injury versus controls. At 5 days post-injury, when degenerating/necrotic regions were present in both animal groups, Akita TA and gastrocnemius muscles displayed reduced macrophage and satellite cell infiltration and poor myofiber formation. By 10 days post-injury, necrotic regions were absent in wild-type TA but persisted in Akita TA. In contrast, Akita soleus exhibited no impairment in any of these measures compared to wild-type soleus. In an effort to define how impaired collagen turnover was attenuating regeneration in Akita TA, a PAI-1 inhibitor (PAI-039) was orally administered to Akita mice following cardiotoxin injury. PAI-039 administration promoted macrophage and satellite cell infiltration into necrotic areas of the TA and gastrocnemius. Importantly, soleus muscles exhibit the highest inducible expression of MMP-9 following injury, providing a mechanism for normative collagen degradation and injury recovery in this muscle despite systemically elevated PAI-1. CONCLUSIONS: Our findings suggest the mechanism underlying how impaired collagen remodelling in type-1 diabetes results in delayed regeneration is an impairment in macrophage infiltration and satellite cell recruitment to degenerating areas; a phenomena that occurs differentially between muscle groups.


Assuntos
Diabetes Mellitus Experimental/imunologia , Macrófagos/imunologia , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Células Satélites de Músculo Esquelético/imunologia , Animais , Diabetes Mellitus Experimental/patologia , Fibrose , Ácidos Indolacéticos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/efeitos dos fármacos , Necrose , Regeneração , Células Satélites de Músculo Esquelético/efeitos dos fármacos
12.
Exerc Immunol Rev ; 18: 42-97, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22876722

RESUMO

Exercise-induced muscle damage is an important topic in exercise physiology. However several aspects of our understanding of how muscles respond to highly stressful exercise remain unclear In the first section of this review we address the evidence that exercise can cause muscle damage and inflammation in otherwise healthy human skeletal muscles. We approach this concept by comparing changes in muscle function (i.e., the force-generating capacity) with the degree of leucocyte accumulation in muscle following exercise. In the second section, we explore the cytokine response to 'muscle-damaging exercise', primarily eccentric exercise. We review the evidence for the notion that the degree of muscle damage is related to the magnitude of the cytokine response. In the third and final section, we look at the satellite cell response to a single bout of eccentric exercise, as well as the role of the cyclooxygenase enzymes (COX1 and 2). In summary, we propose that muscle damage as evaluated by changes in muscle function is related to leucocyte accumulation in the exercised muscles. 'Extreme' exercise protocols, encompassing unaccustomed maximal eccentric exercise across a large range of motion, generally inflict severe muscle damage, inflammation and prolonged recovery (> 1 week). By contrast, exercise resembling regular athletic training (resistance exercise and downhill running) typically causes mild muscle damage (myofibrillar disruptions) and full recovery normally occurs within a few days. Large variation in individual responses to a given exercise should, however be expected. The link between cytokine and satellite cell responses and exercise-induced muscle damage is not so clear The systemic cytokine response may be linked more closely to the metabolic demands of exercise rather than muscle damage. With the exception of IL-6, the sources of systemic cytokines following exercise remain unclear The satellite cell response to severe muscle damage is related to regeneration, whereas the biological significance of satellite cell proliferation after mild damage or non-damaging exercise remains uncertain. The COX enzymes regulate satellite cell activity, as demonstrated in animal models; however the roles of the COX enzymes in human skeletal muscle need further investigation. We suggest using the term 'muscle damage' with care. Comparisons between studies and individuals must consider changes in and recovery of muscle force-generating capacity.


Assuntos
Citocinas/imunologia , Exercício Físico , Leucócitos/imunologia , Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , Regeneração/imunologia , Células Satélites de Músculo Esquelético/imunologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Ciclo-Oxigenase 1/imunologia , Ciclo-Oxigenase 2/imunologia , Humanos , Inflamação/imunologia , Interleucina-6/imunologia , Leucócitos/metabolismo , Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/efeitos dos fármacos
13.
Curr Opin Pharmacol ; 12(3): 372-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22401932

RESUMO

Skeletal muscle diseases heavily impair the strength and the movement of patients. Muscles loose their adaptive capacity, undergoing atrophy or wasting, due to a number of pathological insults. Metabolic changes, such as those occurring during aging, contribute to the progressive reduction of myofiber size and decline of skeletal muscle performance that is typically observed in the elderly. The nicotinamide adenine dinucleotide (NAD)-dependent deacetylase SIRT1 has been involved in the protection against metabolic disorders, against cancers and in the enhancement of life span. Here we discuss the current evidence placing SIRT1 at the crossroad between energy homeostasis, fiber strength, and regeneration from damage in the skeletal muscle. Furthermore, we underline how cell type specific targeting of SIRT1 could be beneficial in the treatment of skeletal muscle diseases.


Assuntos
Músculo Esquelético/fisiopatologia , Doenças Musculares/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Envelhecimento/metabolismo , Animais , Proliferação de Células , Progressão da Doença , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Músculo Esquelético/imunologia , Músculo Esquelético/fisiologia , Doenças Musculares/imunologia , Doenças Musculares/fisiopatologia , Regeneração , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/fisiologia , Sirtuína 1/química
14.
J Appl Physiol (1985) ; 112(10): 1773-82, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22383511

RESUMO

Skeletal muscle is one of the most sensitive tissues to mechanical loading, and unloading inhibits the regeneration potential of skeletal muscle after injury. This study was designed to elucidate the specific effects of unloading stress on the function of immunocytes during muscle regeneration after injury. We examined immunocyte infiltration and muscle regeneration in cardiotoxin (CTX)-injected soleus muscles of tail-suspended (TS) mice. In CTX-injected TS mice, the cross-sectional area of regenerating myofibers was smaller than that of weight-bearing (WB) mice, indicating that unloading delays muscle regeneration following CTX-induced skeletal muscle damage. Delayed infiltration of macrophages into the injured skeletal muscle was observed in CTX-injected TS mice. Neutrophils and macrophages in CTX-injected TS muscle were presented over a longer period at the injury sites compared with those in CTX-injected WB muscle. Disturbance of activation and differentiation of satellite cells was also observed in CTX-injected TS mice. Further analysis showed that the macrophages in soleus muscles were mainly Ly-6C-positive proinflammatory macrophages, with high expression of tumor necrosis factor-α and interleukin-1ß, indicating that unloading causes preferential accumulation and persistence of proinflammatory macrophages in the injured muscle. The phagocytic and myotube formation properties of macrophages from CTX-injected TS skeletal muscle were suppressed compared with those from CTX-injected WB skeletal muscle. We concluded that the disturbed muscle regeneration under unloading is due to impaired macrophage function, inhibition of satellite cell activation, and their cooperation.


Assuntos
Macrófagos/imunologia , Músculo Esquelético/imunologia , Atrofia Muscular/imunologia , Regeneração , Animais , Antígenos de Diferenciação/metabolismo , Antígenos Ly/metabolismo , Biomarcadores/metabolismo , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Regulação da Expressão Gênica , Elevação dos Membros Posteriores , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/genética , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Atrofia Muscular/etiologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Atrofia Muscular/fisiopatologia , Infiltração de Neutrófilos , Fagocitose , Fenótipo , Proteínas Ligases SKP Culina F-Box/genética , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/patologia , Fatores de Tempo , Proteínas com Motivo Tripartido , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/genética
15.
J Nutr Biochem ; 23(9): 1072-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22079208

RESUMO

Acute skeletal muscle damage results in fiber disruption, oxidative stress and inflammation. We investigated cell-specific contributions to the regeneration process after contusion-induced damage (rat gastrocnemius muscle) with or without chronic grape seed-derived proanthocyanidolic oligomer (PCO) administration. In this placebo-controlled study, male Wistar rats were subjected to PCO administration for 2 weeks, after which they were subjected to a standardised contusion injury. Supplementation was continued after injury. Immune and satellite cell responses were assessed, as well as oxygen radical absorption capacity and muscle regeneration. PCO administration resulted in a rapid satellite cell response with an earlier peak in activation (Pax7⁺, CD56⁺, at 4 h post-contusion) vs. placebo groups (PLA) (P<.001: CD56⁺ on Day 5 and Pax7⁺ on Day 7). Specific immune-cell responses in PLA followed expected time courses (neutrophil elevation on Day 1; sustained macrophage elevation from Days 3 to 5). PCO dramatically decreased neutrophil elevation to nonsignificant, while macrophage responses were normal in extent, but significantly earlier (peak between Days 1 and 3) and completely resolved by Day 5. Anti-inflammatory cytokine, IL-10, increased significantly only in PCO (Day 3). Muscle fiber regeneration (MHC(f) content and central nuclei) started earlier and was complete by Day 14 in PCO, but not in PLA. Thus, responses by three crucial cell types involved in muscle recovery were affected by in vivo administration of a specific purified polyphenol in magnitude (neutrophil), time course (macrophages), or time course and activation state (satellite cell), explaining faster effective regeneration in the presence of proanthocyanidolic oligomers.


Assuntos
Antioxidantes/uso terapêutico , Contusões/reabilitação , Suplementos Nutricionais , Extrato de Sementes de Uva/uso terapêutico , Músculo Esquelético/fisiologia , Polifenóis/uso terapêutico , Proantocianidinas/uso terapêutico , Regeneração , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Antioxidantes/análise , Antioxidantes/metabolismo , Antígeno CD56/metabolismo , Contusões/dietoterapia , Contusões/imunologia , Contusões/patologia , Citocinas/sangue , Citocinas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Músculo Esquelético/imunologia , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Cadeias Pesadas de Miosina/metabolismo , Infiltração de Neutrófilos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Fatores de Transcrição Box Pareados/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia
16.
Yakugaku Zasshi ; 131(9): 1329-32, 2011.
Artigo em Japonês | MEDLINE | ID: mdl-21881307

RESUMO

Skeletal muscle has great regenerative potential that depends on muscle stem cells, called satellite cells. In uninjured muscle, satellite cells reside beneath the basal lamina and are maintained in quiescent and undifferentiated state. This state is considered a requisite for sustaining the satellite cell pool, but the molecular mechanism is still unknown. In our previous study, we developed a novel monoclonal antibody that specifically recognized muscle satellite cells in skeletal muscle. Using this monoclonal antibody, we purified satellite cells and performed genome-wide transcriptome analysis. In these analyses, we found that satellite cells expressed Hesr1/Hey1 and Hesr3/HeyL genes known as down stream target of Notch signaling. Although each single knock out mice did not indicate obvious phenotype in skeletal muscle, Hesr1/Hesr3 double knock out mice showed remarkably decreased number of satellite cells. Intriguingly, dKO satellite cells were not kept in quiescent and differentiated state in adult skeletal muscle. This dysregulated state of satellite cells lead to gradually decreased number of satellite cells and age-dependent regeneration defect. These results indicate that Hesr1/3 is essential for muscle stem cell biology and will facilitate this research field.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Ciclo Celular/fisiologia , Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/fisiologia , Animais , Anticorpos Monoclonais , Separação Celular , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Camundongos , Doenças Musculares/genética , Receptores Notch/fisiologia , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/patologia , Transdução de Sinais/genética
17.
Am J Physiol Regul Integr Comp Physiol ; 300(3): R716-23, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191000

RESUMO

The proinflammatory cytokine TNF-α is known to have a direct action on skeletal muscle in mammals. However, little is known regarding the potential effects of cytokines on nonimmune tissues, particularly in skeletal muscle, in fish. The aim of this study was to investigate the effects of recombinant trout TNF-α (rtTNF-α) on skeletal muscle carbohydrate metabolism in rainbow trout (Oncorhynchus mykiss). We used a primary cell culture of muscle cells from rainbow trout to show that rtTNF-α stimulates glucose uptake in myoblasts and myotubes at concentrations that do not affect the viability of the cells, requiring de novo protein synthesis as shown by the impairment of rtTNF-α-stimulated glucose uptake by cycloheximide. With the use of specific inhibitors, we show that rtTNF-α-stimulated glucose uptake is mediated by the p38MAPK, NF-κB, and JNK pathways. Additionally, we provide evidence that the stimulatory effects of rtTNF-α on glucose uptake in trout skeletal muscle cells may be caused, at least in part, by an increase in the amount of GLUT4 at the plasma membrane. Incubation of trout muscle cells with conditioned medium from LPS-stimulated trout macrophages, enriched in TNF-α, increased glucose uptake. Our results indicate that recombinant, as well as native trout TNF-α, directly stimulates glucose uptake in trout muscle cells and provide evidence, for the first time in nonmammalian vertebrates, for a potential regulatory role of TNF-α in skeletal muscle metabolism.


Assuntos
Desoxiglucose/metabolismo , Proteínas de Peixes/metabolismo , Músculo Esquelético/metabolismo , Oncorhynchus mykiss/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Transporte Biológico , Membrana Celular/metabolismo , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/imunologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/imunologia , Comunicação Parácrina , Inibidores de Proteínas Quinases/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Proteínas Recombinantes/metabolismo , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/imunologia , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
PLoS One ; 5(5): e10900, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20531935

RESUMO

HLA class-I expression is weak in embryonic stem cells but increases rapidly during lineage progression. It is unknown whether all three classical HLA class-I antigens follow the same developmental program. In the present study, we investigated allele-specific expression of HLA-A, -B, and -C at the mRNA and protein levels on human mesenchymal stem cells from bone marrow and adipose tissue as well as striated muscle satellite cells and lymphocytes. Using multicolour flow cytometry, we found high cell surface expression of HLA-A on all stem cells and PBMC examined. Surprisingly, HLA-B was either undetectable or very weakly expressed on all stem cells protecting them from complement-dependent cytotoxicity (CDC) using relevant human anti-B and anti-Cw sera. IFNgamma stimulation for 48-72 h was required to induce full HLA-B protein expression. Quantitative real-time RT-PCR showed that IFNgamma induced a 9-42 fold increase of all six HLA-A,-B,-C gene transcripts. Interestingly, prior to stimulation, gene transcripts for all but two alleles were present in similar amounts suggesting that post-transcriptional mechanisms regulate the constitutive expression of HLA-A,-B, and -C. Locus-restricted expression of HLA-A, -B and -C challenges our current understanding of the function of these molecules as regulators of CD8(+) T-cell and NK-cell function and should lead to further inquiries into their expression on other cell types.


Assuntos
Membrana Celular/imunologia , Antígenos HLA-B/genética , Células-Tronco Mesenquimais/imunologia , Células Satélites de Músculo Esquelético/imunologia , Alelos , Citotoxicidade Imunológica/imunologia , Regulação para Baixo/genética , Citometria de Fluxo , Regulação da Expressão Gênica , Antígenos HLA-A/genética , Antígenos HLA-A/imunologia , Antígenos HLA-B/imunologia , Antígenos HLA-C/genética , Antígenos HLA-C/imunologia , Humanos , Interferon gama/imunologia , Espaço Intracelular/imunologia , Cinética , Células-Tronco Mesenquimais/citologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Satélites de Músculo Esquelético/citologia
20.
Hum Mol Genet ; 18(11): 1976-89, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19286669

RESUMO

Skeletal muscle requires an efficient and active membrane repair system to overcome the rigours of frequent contraction. Dysferlin is a component of that system and absence of dysferlin causes muscular dystrophy (dysferlinopathy) characterized by adult onset muscle weakness, high serum creatine kinase levels and a prominent inflammatory infiltrate. We have observed that dysferlinopathy patient biopsies show an excess of immature fibres and therefore investigated the role of dysferlin in muscle regeneration. Using notexin-induced muscle damage, we have shown that regeneration is attenuated in a mouse model of dysferlinopathy, with delayed removal of necrotic fibres, an extended inflammatory phase and delayed functional recovery. Satellite cell activation and myoblast fusion appear normal, but there is a reduction in early neutrophil recruitment in regenerating and also needle wounded muscle in dysferlin-deficient mice. Primary mouse dysferlinopathy myoblast cultures show reduced cytokine release upon stimulation, indicating that the secretion of chemotactic molecules is impaired. We suggest an extension to the muscle membrane repair model, where in addition to fusing patch repair vesicles with the sarcolemma dysferlin is also involved in the release of chemotactic agents. Reduced neutrophil recruitment results in incomplete cycles of regeneration in dysferlinopathy which combines with the membrane repair deficit to ultimately trigger dystrophic pathology. This study reveals a novel pathomechanism affecting muscle regeneration and maintenance in dysferlinopathy and highlights enhancement of the neutrophil response as a potential therapeutic avenue in these disorders.


Assuntos
Proteínas de Membrana/deficiência , Proteínas Musculares/deficiência , Músculo Esquelético/fisiopatologia , Distrofias Musculares/fisiopatologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Disferlina , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/genética , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , Distrofias Musculares/imunologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Neutrófilos/imunologia , Células Satélites de Músculo Esquelético/imunologia , Células Satélites de Músculo Esquelético/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...