Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
J Neuromuscul Dis ; 9(1): 1-23, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34542080

RESUMO

While skeletal muscle remodeling happens throughout life, diseases that result in its dysfunction are accountable for many deaths. Indeed, skeletal muscle is exceptionally capable to respond to stimuli modifying its homeostasis, such as in atrophy, hypertrophy, regeneration and repair. In particular conditions such as genetic diseases (muscular dystrophies), skeletal muscle's capacity to remodel is strongly affected and undergoes continuous cycles of chronic damage. This induces scarring, fatty infiltration, as well as loss of contractibility and of the ability to generate force. In this context, inflammation, primarily mediated by macrophages, plays a central pathogenic role. Macrophages contribute as the primary regulators of inflammation during skeletal muscle regeneration, affecting tissue-resident cells such as myogenic cells and endothelial cells, but also fibro-adipogenic progenitors, which are the main source of the fibro fatty scar. During skeletal muscle regeneration their function is tightly orchestrated, while in dystrophies their fate is strongly disturbed, resulting in chronic inflammation. In this review, we will discuss the latest findings on the role of macrophages in skeletal muscle diseases, and how they are regulated.


Assuntos
Inflamação/imunologia , Macrófagos/fisiologia , Distrofias Musculares/imunologia , Humanos
2.
Eur J Pharmacol ; 912: 174568, 2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-34656607

RESUMO

Muscular dystrophy is a well-known genetically heterogeneous group of rare muscle disorders. This progressive disease causes the breakdown of skeletal muscles over time and leads to grave weakness. This breakdown is caused by a diverse pattern of mutations in dystrophin and dystrophin associated protein complex. These mutations lead to the production of altered proteins in response to which, the body stimulates production of various cytokines and immune cells, particularly reactive oxygen species and NFκB. Immune cells display/exhibit a dual role by inducing muscle damage and muscle repair. Various anti-oxidants, anti-inflammatory and glucocorticoid drugs serve as potent therapeutics for muscular dystrophy. Along with the above mentioned therapeutics, induced pluripotent stem cells also serve as a novel approach paving a way for personalized treatment. These pluripotent stem cells allow regeneration of large numbers of regenerative myogenic progenitors that can be administered in muscular dystrophy patients which assist in the recovery of lost muscle fibers. In this review, we have summarized gene-editing, immunological and induced pluripotent stem cell based therapeutics for muscular dystrophy treatment.


Assuntos
Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas , Distrofias Musculares/genética , Distrofias Musculares/imunologia , Animais , Distrofina/genética , Humanos , Distrofias Musculares/metabolismo , Distrofias Musculares/terapia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo
3.
Front Immunol ; 12: 666879, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335568

RESUMO

Muscular dystrophies and inflammatory myopathies are heterogeneous muscular disorders characterized by progressive muscle weakness and mass loss. Despite the high variability of etiology, inflammation and involvement of both innate and adaptive immune response are shared features. The best understood immune mechanisms involved in these pathologies include complement cascade activation, auto-antibodies directed against muscular proteins or de-novo expressed antigens in myofibers, MHC-I overexpression in myofibers, and lymphocytes-mediated cytotoxicity. Intravenous immunoglobulins (IVIGs) administration could represent a suitable immunomodulator with this respect. Here we focus on mechanisms of action of immunoglobulins in muscular dystrophies and inflammatory myopathies highlighting results of IVIGs from pre-clinical and case reports evidences.


Assuntos
Autoimunidade , Imunoglobulinas/imunologia , Distrofias Musculares/imunologia , Miosite/imunologia , Autoanticorpos/imunologia , Humanos , Imunoglobulinas/metabolismo , Proteínas Musculares/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Miosite/metabolismo , Miosite/patologia
4.
Skelet Muscle ; 10(1): 18, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32498713

RESUMO

BACKGROUND: Laminin-α2-related congenital muscular dystrophy (LAMA2-CMD) is a devastating genetic disease caused by mutations in the LAMA2 gene. These mutations result in progressive muscle wasting and inflammation leading to delayed milestones, and reduced lifespan in affected patients. There is currently no cure or treatment for LAMA2-CMD. Preclinical studies have demonstrated that mouse laminin-111 can serve as an effective protein replacement therapy in a mouse model of LAMA2-CMD. METHODS: In this study, we generated a novel immunocompromised dyW mouse model of LAMA2-CMD to study the role the immune system plays in muscle disease progression. We used this immune-deficient dyW mouse model to test the therapeutic benefits of recombinant human laminin-111 and laminin-211 protein therapy on laminin-α2-deficient muscle disease progression. RESULTS: We show that immunodeficient laminin-α2 null mice demonstrate subtle differences in muscle regeneration compared to immunocompetent animals during early disease stages but overall exhibit a comparable muscle disease progression. We found human laminin-111 and laminin-211 could serve as effective protein replacement strategies with mice showing improvements in muscle pathology and function. We observed that human laminin-111 and laminin-211 exhibit differences on satellite and myoblast cell populations and differentially affect muscle repair. CONCLUSIONS: This study describes the generation of a novel immunodeficient mouse model that allows investigation of the role the immune system plays in LAMA2-CMD. This model can be used to assess the therapeutic potential of heterologous therapies that would elicit an immune response. Using this model, we show that recombinant human laminin-111 can serve as effective protein replacement therapy for the treatment of LAMA2-CMD.


Assuntos
Terapia Genética/métodos , Laminina/genética , Distrofias Musculares/terapia , Animais , Humanos , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
Bull Math Biol ; 81(10): 3976-3997, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31302876

RESUMO

Muscular dystrophy describes generalized progressive muscular weakness due to the wasting of muscle fibers. The progression of the disease is affected by known immunological and mechanical factors, and possibly other unknown mechanisms. This article introduces a new mathematical model, the FRiND model, to further elucidate these known immunological actions. We will perform stability and sensitivity analyses on this model. The models time course results will be verified by biological studies in the literature. This model could be the foundation for further understanding of immunological muscle repair.


Assuntos
Macrófagos/imunologia , Modelos Imunológicos , Distrofias Musculares/imunologia , Animais , Simulação por Computador , Humanos , Imunidade Inata , Inflamação/etiologia , Inflamação/imunologia , Inflamação/patologia , Macrófagos/patologia , Conceitos Matemáticos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Distrofias Musculares/etiologia , Distrofias Musculares/patologia , Regeneração/imunologia , Biologia de Sistemas
6.
Curr Opin Pharmacol ; 47: 40-45, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30901735

RESUMO

Mutations in the dystrophin and sarcoglycans genes result in muscular dystrophies causing severe disability and premature death and where no effective treatment is available. New therapeutic approaches targeting secondary disease mechanisms have a strong translational potential. Dystrophic muscle damage triggers release of ATP whilst loss of ecto-ATPase activity of sarcoglycan further elevates extracellular ATP (eATP) levels. Such a high eATP activates P2X7 purinoceptors on immune cells; these contribute to chronic inflammatory and immune responses that exacerbate the dystrophic pathology. Dystrophin mutations coincide with a significant P2X7 upregulation in Duchenne muscular dystrophy (DMD) muscle and alter receptor signalling in mouse dystrophic myoblasts and myofibers. P2X7 overexpression combined with the eATP-rich environment lead to cell dysfunction and death and ultimately to ineffective regeneration. P2X7 is therefore a therapeutic target for reducing damaging inflammation and supporting the repair of dystrophic muscles. Accordingly, genetic ablation and pharmacological inhibition of the eATP-P2X7 axis alleviated dystrophic phenotypes in mouse models of dystrophinopathy and sarcoglycanopathy. Thus, P2X7 inhibitors are good candidates for rapid re-purposing for the treatment of these highly debilitating diseases. Such a therapy is not constrained by causative mutations, so it would be suitable for all patients. Moreover, it appears effective in alleviating both muscle and non-muscle symptoms.


Assuntos
Distrofias Musculares/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animais , Humanos , Músculo Esquelético/metabolismo , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/imunologia , Antagonistas do Receptor Purinérgico P2X/uso terapêutico
7.
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
8.
Arch Immunol Ther Exp (Warsz) ; 66(5): 341-354, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29536116

RESUMO

Muscular dystrophies represent a group of diseases which may develop in several forms, and severity of the disease is usually associated with gene mutations. In skeletal muscle regeneration and in muscular dystrophies, both innate and adaptive immune responses are involved. The regenerative potential of mesenchymal stem/stromal cells (MSCs) of bone marrow origin was confirmed by the ability to differentiate into diverse tissues and by their immunomodulatory and anti-inflammatory properties by secretion of a variety of growth factors and anti-inflammatory cytokines. Skeletal muscle comprises different types of stem/progenitor cells such as satellite cells and non-satellite stem cells including MSCs, interstitial stem cells positive for stress mediator PW1 expression and negative for PAX7 called PICs (PW1+/PAX7- interstitial cells), fibro/adipogenic progenitors/mesenchymal stem cells, muscle side population cells and muscle resident pericytes, and all of them actively participate in the muscle regeneration process. In this review, we present biological properties of MSCs of bone marrow origin and a heterogeneous population of muscle-resident stem/progenitor cells, their interaction with the inflammatory environment of dystrophic muscle and potential implications for cellular therapies for muscle regeneration. Subsequently, we propose-based on current research results, conclusions, and our own experience-hypothetical mechanisms for modulation of the complete muscle regeneration process to treat muscular dystrophies.


Assuntos
Células da Medula Óssea/fisiologia , Inflamação/imunologia , Células-Tronco Mesenquimais/fisiologia , Desenvolvimento Muscular , Músculo Esquelético/fisiologia , Distrofias Musculares/imunologia , Células-Tronco/fisiologia , Animais , Regeneração Tecidual Guiada , Humanos , Imunidade , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Fator de Transcrição PAX7/metabolismo
9.
Skelet Muscle ; 7(1): 23, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-29078808

RESUMO

BACKGROUND: Chronic increases in the levels of the inflammatory cytokine interleukin-6 (IL-6) in serum and skeletal muscle are thought to contribute to the progression of muscular dystrophy. Dystrophin/utrophin double-knockout (dKO) mice develop a more severe and progressive muscular dystrophy than the mdx mice, the most common murine model of Duchenne muscular dystrophy (DMD). In particular, dKO mice have smaller body sizes and muscle diameters, and develop progressive kyphosis and fibrosis in skeletal and cardiac muscles. As mdx mice and DMD patients, we found that IL-6 levels in the skeletal muscle were significantly increased in dKO mice. Thus, in this study, we aimed to analyze the effects of IL-6 receptor (IL-6R) blockade on the muscle pathology of dKO mice. METHODS: Male dKO mice were administered an initial injection (200 mg/kg intraperitoneally (i.p.)) of either the anti-IL-6R antibody MR16-1 or an isotype-matched control rat IgG at the age of 14 days, and were then given weekly injections (25 mg/kg i.p.) until 90 days of age. RESULTS: Treatment of dKO mice with the MR16-1 antibody successfully inhibited the IL-6 pathway in the skeletal muscle and resulted in a significant reduction in the expression levels of phosphorylated signal transducer and activator of transcription 3 in the skeletal muscle. Pathologically, a significant increase in the area of embryonic myosin heavy chain-positive myofibers and muscle diameter, and reduced fibrosis in the quadriceps muscle were observed. These results demonstrated the therapeutic effects of IL-6R blockade on promoting muscle regeneration. Consistently, serum creatine kinase levels were decreased. Despite these improvements observed in the limb muscles, degeneration of the diaphragm and cardiac muscles was not ameliorated by the treatment of mice with the MR16-1 antibody. CONCLUSION: As no adverse effects of treatment with the MR16-1 antibody were observed, our results indicate that the anti-IL-6R antibody is a potential therapy for muscular dystrophy particularly for promoting skeletal muscle regeneration.


Assuntos
Anticorpos/administração & dosagem , Músculo Esquelético/efeitos dos fármacos , Distrofias Musculares/tratamento farmacológico , Receptores de Interleucina-6/imunologia , Regeneração/efeitos dos fármacos , Animais , Creatina Quinase/sangue , Modelos Animais de Doenças , Distrofina/genética , Fibrose/complicações , Inflamação/complicações , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofias Musculares/complicações , Distrofias Musculares/imunologia , Receptores de Interleucina-6/antagonistas & inibidores , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Utrofina/genética
10.
BMC Res Notes ; 10(1): 358, 2017 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-28755659

RESUMO

BACKGROUND: Collagen VI is a ubiquitously-expressed macromolecule that forms unique microfibrillar assemblies in the extracellular matrix. Mutations in the COL6A1, COL6A2 and COL6A3 genes result in congenital muscular dystrophy, arguing that collagen is critical for skeletal muscle development and function. Antibodies against collagen VI are important clinical and diagnostic tools in muscular dystrophy. They are used to confirm genetic findings by detecting abnormalities in the distribution, organization and overall levels of collagen VI in cells and tissues isolated from patients. METHODS: Many antibodies have been raised against tissue-purified collagen VI and individual collagen VI chains, however few have been properly validated for sensitivity and chain specificity. To address this deficiency, we compared the ability of 23 commercially-available antibodies to detect extracellular collagen VI by immunohistochemistry on frozen tissue sections. To determine chain specificity, immunoblot analyses were conducted on cell lysates isolated from cells transfected with cDNAs for each individual chain and cells expressing all three chains together. RESULTS: Our analyses identified 15 antibodies that recognized tissue collagen VI by immunohistochemistry at varying intensities and 20 that successfully detected collagen VI by immunoblotting. Three antibodies failed to recognize collagen VI by either method under the conditions tested. All chain-specific antibodies that worked by immunoblotting specifically recognized their correct chain, and no other chains. CONCLUSIONS: This series of side-by-side comparisons reveal at least two antibodies specific for each chain that work well for immunohistochemistry on frozen sections. This validation study expands the repertoire of antibodies available for muscular dystrophy studies caused by defects in collagen VI.


Assuntos
Anticorpos , Colágeno Tipo VI/imunologia , Distrofias Musculares/imunologia , Humanos , Imuno-Histoquímica
11.
Chembiochem ; 18(13): 1155-1171, 2017 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-28423240

RESUMO

About 2-3 million years ago, Alu-mediated deletion of a critical exon in the CMAH gene became fixed in the hominin lineage ancestral to humans, possibly through a stepwise process of selection by pathogen targeting of the CMAH product (the sialic acid Neu5Gc), followed by reproductive isolation through female anti-Neu5Gc antibodies. Loss of CMAH has occurred independently in some other lineages, but is functionally intact in Old World primates, including our closest relatives, the chimpanzee. Although the biophysical and biochemical ramifications of losing tens of millions of Neu5Gc hydroxy groups at most cell surfaces remains poorly understood, we do know that there are multiscale effects functionally relevant to both sides of the host-pathogen interface. Hominin CMAH loss might also contribute to understanding human evolution, at the time when our ancestors were starting to use stone tools, increasing their consumption of meat, and possibly hunting. Comparisons with chimpanzees within ethical and practical limitations have revealed some consequences of human CMAH loss, but more has been learned by using a mouse model with a human-like Cmah inactivation. For example, such mice can develop antibodies against Neu5Gc that could affect inflammatory processes like cancer progression in the face of Neu5Gc metabolic incorporation from red meats, display a hyper-reactive immune system, a human-like tendency for delayed wound healing, late-onset hearing loss, insulin resistance, susceptibility to muscular dystrophy pathologies, and increased sensitivity to multiple human-adapted pathogens involving sialic acids. Further studies in such mice could provide a model for other human-specific processes and pathologies involving sialic acid biology that have yet to be explored.


Assuntos
Genoma , Perda Auditiva/metabolismo , Oxigenases de Função Mista/genética , Distrofias Musculares/metabolismo , Neoplasias/metabolismo , Ácidos Neuramínicos/metabolismo , Animais , Autoanticorpos/biossíntese , Evolução Biológica , Suscetibilidade a Doenças , Expressão Gênica , Perda Auditiva/genética , Perda Auditiva/imunologia , Perda Auditiva/patologia , Humanos , Resistência à Insulina , Camundongos , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/imunologia , Distrofias Musculares/genética , Distrofias Musculares/imunologia , Distrofias Musculares/patologia , Mutação , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Ácidos Neuramínicos/química , Ácidos Neuramínicos/imunologia , Pan troglodytes
12.
Hum Gene Ther ; 28(6): 493-509, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28345428

RESUMO

Use of adeno-associated virus (AAV) to transduce genes into skeletal muscles can be associated with T-cell responses to viral capsid and/or to transgenic protein. Intramuscular mononuclear cell infiltrates primarily consisting of CD8+ T cells and also containing FOXP3+ regulatory T cells were present in rhesus macaque skeletal muscle treated with rAAVrh74.MCK.GALGT2 by vascular delivery. Administration of oral prednisone prior to AAV gene delivery and throughout the study reduced such infiltrates by 60% at 24 weeks post AAV delivery compared with AAV-treated animals not receiving prednisone, regardless of the presence of pre-existing AAV serum antibodies at the time of treatment. The majority of CD8+ T cells in AAV-treated muscles expressed activated caspase 3 and programmed cell death protein 1 (PD1), suggesting ongoing programmed cell death. AAV-transduced skeletal muscles also had elevated expression of programmed death ligand 2 (PDL2) on skeletal myofibers, and this increase in expression extended to muscles where transgene was not overexpressed. These data demonstrate that prednisone can reduce the extent of intramuscular T-cell infiltrates in AAV-treated muscles, which may aid in achieving long-term transgene expression, as may the induction of PDL2 expression on skeletal myofibers to promote PD1-mediated programmed T-cell death.


Assuntos
Dependovirus/genética , Vetores Genéticos/imunologia , Imunossupressores/farmacologia , Distrofias Musculares/terapia , Prednisona/farmacologia , Proteína 2 Ligante de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/genética , Administração Oral , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Morte Celular , Dependovirus/imunologia , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/química , Imunidade Celular/efeitos dos fármacos , Injeções Intra-Arteriais , Injeções Intramusculares , Macaca mulatta , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Distrofias Musculares/genética , Distrofias Musculares/imunologia , Distrofias Musculares/patologia , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/imunologia , Proteína 2 Ligante de Morte Celular Programada 1/agonistas , Proteína 2 Ligante de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/agonistas , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Transgenes
13.
J Neuromuscul Dis ; 3(4): 455-473, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27911334

RESUMO

In skeletal muscles, levels and activity of Matrix MetalloProteinases (MMPs) and Tissue Inhibitors of MetalloProteinases (TIMPs) have been involved in myoblast migration, fusion and various physiological and pathological remodeling situations including neuromuscular diseases. This has opened perspectives for the use of MMPs' overexpression to improve the efficiency of cell therapy in muscular dystrophies and resolve fibrosis. Alternatively, inhibition of individual MMPs in animal models of muscular dystrophies has provided evidence of beneficial, dual or adverse effects on muscle morphology or function. We review here the role played by MMPs/TIMPs in skeletal muscle inflammation and fibrosis, two major hurdles that limit the success of cell and gene therapy. We report and analyze the consequences of genetic or pharmacological modulation of MMP levels on the inflammation of skeletal muscles and their repair in light of experimental findings. We further discuss how the interplay between MMPs/TIMPs levels, cytokines/chemokines, growth factors and permanent low-grade inflammation favor cellular and molecular modifications resulting in fibrosis.


Assuntos
Inflamação/imunologia , Metaloproteinases da Matriz/imunologia , Músculo Esquelético/imunologia , Inibidores Teciduais de Metaloproteinases/imunologia , Animais , Terapia Baseada em Transplante de Células e Tecidos , Citocinas/imunologia , Fibrose , Terapia Genética , Humanos , Músculo Esquelético/patologia , Distrofias Musculares/imunologia , Distrofias Musculares/patologia , Distrofias Musculares/terapia
14.
Arthritis Res Ther ; 17: 238, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26338728

RESUMO

INTRODUCTION: Microchimeric cells have been studied for over a decade, with conflicting reports on their presence and role in autoimmune and other inflammatory diseases. To determine whether microchimeric cells were pathogenic or mediating tissue repair in inflammatory myopathies, we phenotyped and quantified microchimeric cells in juvenile idiopathic inflammatory myopathies (JIIM), muscular dystrophy (MD), and noninflammatory control muscle tissues. METHOD: Fluorescence immunophenotyping for infiltrating cells with sequential fluorescence in situ hybridization was performed on muscle biopsies from ten patients with JIIM, nine with MD and ten controls. RESULTS: Microchimeric cells were significantly increased in MD muscle (0.079 ± 0.024 microchimeric cells/mm(2) tissue) compared to controls (0.019 ± 0.007 cells/mm(2) tissue, p = 0.01), but not elevated in JIIM muscle (0.043 ± 0.015 cells/mm(2)). Significantly more CD4+ and CD8+ microchimeric cells were in the muscle of patients with MD compared with controls (mean 0.053 ± 0.020/mm(2) versus 0 ± 0/mm(2) p = 0.003 and 0.043 ± 0.023/mm(2) versus 0 ± 0/mm(2) p = 0.025, respectively). No differences in microchimeric cells between JIIM, MD, and noninflammatory controls were found for CD3+, Class II+, CD25+, CD45RA+, and CD123+ phenotypes, and no microchimeric cells were detected in CD20, CD83, or CD45RO populations. The locations of microchimeric cells were similar in all three conditions, with MD muscle having more microchimeric cells in perimysial regions than controls, and JIIM having fewer microchimeric muscle nuclei than MD. Microchimeric inflammatory cells were found, in most cases, at significantly lower proportions than autologous cells of the same phenotype. CONCLUSIONS: Microchimeric cells are not specific to autoimmune disease, and may not be important in muscle inflammation or tissue repair in JIIM.


Assuntos
Quimera/imunologia , Quimerismo , Distrofias Musculares/imunologia , Miosite/imunologia , Adolescente , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Criança , Pré-Escolar , Quimera/metabolismo , Feminino , Humanos , Células Híbridas/imunologia , Células Híbridas/metabolismo , Imunofenotipagem , Hibridização in Situ Fluorescente , Contagem de Linfócitos , Troca Materno-Fetal/genética , Troca Materno-Fetal/imunologia , Microscopia de Fluorescência , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Miosite/genética , Miosite/metabolismo , Gravidez
15.
Biomed Res Int ; 2014: 818107, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24959590

RESUMO

Muscular dystrophies (MDs) are a heterogeneous group of diseases, caused by mutations in different components of sarcolemma, extracellular matrix, or enzymes. Inflammation and innate or adaptive immune response activation are prominent features of MDs. Various therapies under development are directed toward rescuing the dystrophic muscle damage using gene transfer or cell therapy. Here we discussed current knowledge about involvement of immune system responses to experimental therapies in MDs.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Imunidade Inata , Distrofias Musculares/terapia , Células-Tronco/imunologia , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Terapia Genética , Humanos , Distrofias Musculares/imunologia , Distrofias Musculares/patologia , Mutação , Células-Tronco/citologia
16.
J Invest Dermatol ; 134(11): 2776-2783, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24940650

RESUMO

Plectin, a cytolinker of the plakin family, anchors the intermediate filament (IF) network formed by keratins 5 and 14 (K5/K14) to hemidesmosomes, junctional adhesion complexes in basal keratinocytes. Genetic alterations of these proteins cause epidermolysis bullosa simplex (EBS) characterized by disturbed cytoarchitecture and cell fragility. The mechanisms through which mutations located after the documented plectin IF-binding site, composed of the plakin-repeat domain (PRD) B5 and the linker, as well as mutations in K5 or K14, lead to EBS remain unclear. We investigated the interaction of plectin C terminus, encompassing four domains, the PRD B5, the linker, the PRD C, and the C extremity, with K5/K14 using different approaches, including a rapid and sensitive fluorescent protein-binding assay, based on enhanced green fluorescent protein-tagged proteins (FluoBACE). Our results demonstrate that all four plectin C-terminal domains contribute to its association with K5/K14 and act synergistically to ensure efficient IF binding. The plectin C terminus predominantly interacted with the K5/K14 coil 1 domain and bound more extensively to K5/K14 filaments compared with monomeric keratins or IF assembly intermediates. These findings indicate a multimodular association of plectin with K5/K14 filaments and give insights into the molecular basis of EBS associated with pathogenic mutations in plectin, K5, or K14 genes.


Assuntos
Queratina-14/química , Queratina-5/química , Plectina/química , Sítios de Ligação , Linhagem Celular Tumoral , Epidermólise Bolhosa Simples/imunologia , Células HEK293 , Humanos , Queratinas/química , Distrofias Musculares/imunologia , Mutação , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Técnicas do Sistema de Duplo-Híbrido
17.
Curr Opin Neurol ; 26(5): 554-60, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23995277

RESUMO

PURPOSE OF REVIEW: Necrotizing myopathy exhibits a specific histological pattern, characterized by significant necrosis, with simultaneous muscle fiber regeneration, but without or with little inflammation. This histological pattern may be observed in acquired myopathies but also in muscular dystrophy. Acquired necrotizing myopathy can be secondary to drugs or toxics agents, and if not, autoimmune mechanisms have to be suspected. Necrotizing autoimmune myopathy (NAM) is now recognized as a subgroup of idiopathic inflammatory myopathies. This review aims to further describe the different acquired myopathies, with special focus on NAM since specific autoantibodies have been discovered. RECENT FINDINGS: Usually, acquired myopathy has an acute onset, but a slower progression may sometimes be observed that could lead to erroneous diagnosis of muscular dystrophy. NAM may be associated with specific autoantibodies against signal recognition particle, or, as more recently described, against 3-hydroxy-3-methylglutaryl-coenzyme A reductase, especially in statin-exposed patients. Their presence permits affirmation that muscular necrosis is immune-mediated. The antibody titer is correlated to the disease activity. SUMMARY: NAMs are now considered as a new entity, treatable by immunosuppressants, and should not be misdiagnosed as a muscular dystrophy.


Assuntos
Distrofias Musculares/tratamento farmacológico , Miosite/tratamento farmacológico , Autoanticorpos/imunologia , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/tratamento farmacológico , Humanos , Imunossupressores/uso terapêutico , Distrofias Musculares/imunologia , Distrofias Musculares/patologia , Miosite/diagnóstico , Miosite/etiologia , Miosite/patologia
18.
Eur J Pharmacol ; 715(1-3): 296-303, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23707352

RESUMO

Inflammation plays a crucial role in muscle remodeling and repair after acute and chronic damage, in particular in muscular dystrophies, a heterogeneous group of genetic diseases leading to muscular degeneration. Defect of nitric oxide (NO) generation is a key pathogenic event in muscular dystrophies, thus NO donors have been explored as new therapeutics for this disease. We have investigated the immune-modulating effect of one of such drugs, molsidomine, able to slow the progression of muscular dystrophy in the α-Sarcoglican-null mice, a model for the limb girdle muscular dystrophy 2D, sharing several hallmarks of muscle degeneration with other muscular dystrophies. α-Sarcoglican-null mice were treated with molsidomine and drug effects on the inflammatory infiltrates and on muscle repair were assessed at selected time points. We found that molsidomine treatment modulates effectively the characteristics of the inflammatory infiltrate within dystrophic muscles, enhancing its healing function. Initially molsidomine amplified macrophage recruitment, promoting a more efficient clearance of cell debris and effective tissue regeneration. At a later stage molsidomine decreased significantly the extent of the inflammatory infiltrate, whose persistence exacerbates muscle damage: most of the remaining macrophages displayed characteristics of the transitional population, associated with reduced fibrosis and increased preservation of the muscle tissue. The dual action of molsidomine, the already known NO donation and the immunomodulatory function we now identified, suggests that it has a unique potential in tissue healing during chronic muscle damage. This, alongside its already approved use in human, makes molsidomine a drug with a significant therapeutic potential in muscular dystrophies.


Assuntos
Imunidade Inata/efeitos dos fármacos , Molsidomina/metabolismo , Molsidomina/farmacologia , Distrofias Musculares/imunologia , Óxido Nítrico/metabolismo , Animais , Modelos Animais de Doenças , Fibrose , Humanos , Inflamação/imunologia , Inflamação/patologia , Camundongos , Distrofias Musculares/patologia , Fenótipo
19.
FEBS J ; 280(17): 4165-76, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23527661

RESUMO

Cells encounter many physical, chemical and biological stresses that perturb plasma membrane integrity, warranting an immediate membrane repair response to regain cell homeostasis. Failure to respond properly to such perturbation leads to individual cell death, which may also produce systemic influence by triggering sterile immunological responses. In this review, we discuss recent progress on understanding the mechanisms underlying muscle cell membrane repair and the potential mediators of innate immune activation when the membrane repair system is defective, specifically focusing on pathology associated with dysferlin deficiency.


Assuntos
Membrana Celular/imunologia , Imunidade Inata , Proteínas de Membrana/fisiologia , Músculo Esquelético/imunologia , Distrofias Musculares/imunologia , Animais , Membrana Celular/metabolismo , Disferlina , Camundongos , Camundongos Knockout , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia
20.
Neurol India ; 61(6): 614-21, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24441329

RESUMO

BACKGROUND: In inflammatory myopathies muscle biopsy is a crucial diagnostic test. Misinterpretation between inflammatory myopathies and muscular dystrophies with inflammation is known. MATERIALS AND METHODS: Thirty-one patients clinically and pathologically diagnosed to have polymyositis and dermatomyositis and 16 patients of muscular dystrophy with inflammation were studied for MHC-I, MHC-II, CD4 and CD8 expression in skeletal muscle tissue. RESULTS: MHC-I upregulation was noted in all samples of PM and DM. Interstitial and perivascular inflammation in PM were predominantly CD8+ cells, in dermatomyositis, interstitial and perimysial perivascular inflammatory cells were CD4+ T cells and CD8+ T cells were seen around endomysial vessels. Interestingly MHC-I upregulation was seen in all 16 cases of muscular dystrophy with presence of inflammation. CONCLUSION: The pattern of MHC-I and II expression appeared to be similar in both inflammatory myopathies as well as in muscular dystrophies with inflammation and hence differentiating them on MHC - I expression was difficult.


Assuntos
Complexo Principal de Histocompatibilidade/imunologia , Distrofias Musculares/diagnóstico , Miosite/diagnóstico , Adolescente , Adulto , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Criança , Pré-Escolar , Diagnóstico Diferencial , Feminino , Humanos , Imuno-Histoquímica , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/imunologia , Distrofias Musculares/metabolismo , Miosite/imunologia , Miosite/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA