Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 7(1): 5160, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28698572

RESUMO

Loss of skeletal muscle mass and function occurs with increasing age. Calorie restriction (CR) increases the lifespan of C57Bl/6 mice, but not in the shorter-lived DBA/2 strain. There is some evidence that calorie restriction reduces or delays many of the age-related defects that occur in rodent skeletal muscle. We therefore investigated the effect of short (2.5 month) and longer term (8.5 and 18.5 months) CR on skeletal muscle in male and female C57Bl/6 and DBA/2 mice. We found that short-term CR increased the satellite cell number and collagen VI content of muscle, but resulted in a delayed regenerative response to injury.Consistent with this, the in vitro proliferation of satellite cells derived from these muscles was reduced by CR. The percentage of stromal cells, macrophages, hematopoietic stem cells and fibroadipogenic cells in the mononucleated cell population derived from skeletal muscle was reduced by CR at various stages. But overall, these changes are neither consistent over time, nor between strain and sex. The fact that changes induced by CR do not persist with time and the dissimilarities between the two mouse strains, combined with sex differences, urge caution in applying CR to improve skeletal muscle function across the lifespan in humans.


Assuntos
Restrição Calórica/efeitos adversos , Colágeno Tipo VI/metabolismo , Músculo Esquelético/citologia , Animais , Peso Corporal , Restrição Calórica/métodos , Proliferação de Células , Células Cultivadas , Metabolismo Energético , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Caracteres Sexuais , Fatores de Tempo
2.
Front Immunol ; 7: 667, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28127296

RESUMO

Immune senescence is a natural consequence of aging and may contribute to frailty and loss of homeostasis in later life. Calorie restriction increases healthy life-span in C57BL/6J (but not DBA/2J) mice, but whether this is related to preservation of immune function, and how it interacts with aging, is unclear. We compared phenotypic and functional characteristics of natural killer (NK) cells and T cells, across the lifespan, of calorie-restricted (CR) and control C57BL/6 and DBA/2 mice. Calorie restriction preserves a naïve T cell phenotype and an immature NK cell phenotype as mice age. The splenic T cell populations of CR mice had higher proportions of CD11a-CD44lo cells, lower expression of TRAIL, KLRG1, and CXCR3, and higher expression of CD127, compared to control mice. Similarly, splenic NK cells from CR mice had higher proportions of less differentiated CD11b-CD27+ cells and correspondingly lower proportions of highly differentiated CD11b+CD27-NK cells. Within each of these subsets, cells from CR mice had higher expression of CD127, CD25, TRAIL, NKG2A/C/E, and CXCR3 and lower expression of KLRG1 and Ly49 receptors compared to controls. The effects of calorie restriction on lymphoid cell populations in lung, liver, and lymph nodes were identical to those seen in the spleen, indicating that this is a system-wide effect. The impact of calorie restriction on NK cell and T cell maturation is much more profound than the effect of aging and, indeed, calorie restriction attenuates these age-associated changes. Importantly, the effects of calorie restriction on lymphocyte maturation were more marked in C57BL/6 than in DBA/2J mice indicating that delayed lymphocyte maturation correlates with extended lifespan. These findings have implications for understanding the interaction between nutritional status, immunity, and healthy lifespan in aging populations.

3.
Stem Cell Res ; 14(1): 20-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25460248

RESUMO

Duchenne muscular dystrophy is an inherited disorder that is characterized by progressive skeletal muscle weakness and wasting, with a failure of muscle maintenance/repair mediated by satellite cells (muscle stem cells). The function of skeletal muscle stem cells resident in dystrophic muscle may be perturbed by being in an increasing pathogenic environment, coupled with constant demands for repairing muscle. To investigate the contribution of satellite cell exhaustion to this process, we tested the functionality of satellite cells isolated from the mdx mouse model of Duchenne muscular dystrophy. We found that satellite cells derived from young mdx mice contributed efficiently to muscle regeneration within our in vivo mouse model. To then test the effects of long-term residence in a dystrophic environment, satellite cells were isolated from aged mdx muscle. Surprisingly, they were as functional as those derived from young or aged wild type donors. Removing satellite cells from a dystrophic milieu reveals that their regenerative capacity remains both intact and similar to satellite cells derived from healthy muscle, indicating that the host environment is critical for controlling satellite cell function.


Assuntos
Músculos/fisiologia , Regeneração , Células Satélites de Músculo Esquelético/transplante , Envelhecimento , Animais , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Nus , Músculos/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo
4.
Curr Gene Ther ; 14(4): 276-88, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25039614

RESUMO

Lentiviral vectors (LVs) represent suitable candidates to mediate gene therapy for muscular dystrophies as they infect dividing and non-dividing cells and integrate their genetic material into the host genome, thereby theoretically mediating longterm expression. We evaluated the ability of LVs where a GFP reporter gene was under the control of five different promoters, to transduce and mediate expression in myogenic and non-myogenic cells in vitro and in skeletal muscle fibres and stem (satellite) cells in vivo. We further analysed lentivirally-transduced satellite cell-derived myoblasts following their transplantation into dystrophic, immunodeficient mouse muscles. The spleen focus-forming virus promoter mediated the highest gene expression in all cell types; the CBX3-HNRPA2B1 ubiquitously-acting chromatin opening element (UCOE) promoter was also active in all cells, whereas the human desmin promoter in isolation or fused with UCOE had lower activity in non-muscle cells. Surprisingly, the human skeletal muscle actin promoter was also active in immune cells. The human desmin promoter mediated robust, persistent reporter gene expression in myogenic cells in vitro, and satellite cells and muscle fibres in vivo. The human desmin promoter combined with UCOE did not significantly increase transgene expression. Therefore, our data indicate that the desmin promoter is suitable for the development of therapeutic purposes.


Assuntos
Desmina/genética , Terapia Genética/métodos , Vetores Genéticos , Células Satélites de Músculo Esquelético/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Regulação da Expressão Gênica , Humanos , Camundongos Endogâmicos mdx , Músculo Esquelético/citologia , Mioblastos Esqueléticos/citologia , Especificidade de Órgãos , Regiões Promotoras Genéticas , Vírus Formadores de Foco no Baço/genética , Transgenes
5.
J Am Heart Assoc ; 2(4): e000284, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23963759

RESUMO

BACKGROUND: Dilated cardiomyopathy (DCM) is a leading cause of chronic morbidity and mortality in muscular dystrophy (MD) patients. Current pharmacological treatments are not yet able to counteract chronic myocardial wastage, thus novel therapies are being intensely explored. MicroRNAs have been implicated as fine regulators of cardiomyopathic progression. Previously, miR-669a downregulation has been linked to the severe DCM progression displayed by Sgcb-null dystrophic mice. However, the impact of long-term overexpression of miR-669a on muscle structure and functionality of the dystrophic heart is yet unknown. METHODS AND RESULTS: Here, we demonstrate that intraventricular delivery of adeno-associated viral (AAV) vectors induces long-term (18 months) miR-669a overexpression and improves survival of Sgcb-null mice. Treated hearts display significant decrease in hypertrophic remodeling, fibrosis, and cardiomyocyte apoptosis. Moreover, miR-669a treatment increases sarcomere organization, reduces ventricular atrial natriuretic peptide (ANP) levels, and ameliorates gene/miRNA profile of DCM markers. Furthermore, long-term miR-669a overexpression significantly reduces adverse remodeling and enhances systolic fractional shortening of the left ventricle in treated dystrophic mice, without significant detrimental consequences on skeletal muscle wastage. CONCLUSIONS: Our findings provide the first evidence of long-term beneficial impact of AAV-mediated miRNA therapy in a transgenic model of severe, chronic MD-associated DCM.


Assuntos
Cardiomiopatia Dilatada/terapia , Terapia Genética/métodos , MicroRNAs/metabolismo , Distrofias Musculares/complicações , Animais , Apoptose , Fator Natriurético Atrial/metabolismo , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Doença Crônica , Dependovirus , Modelos Animais de Doenças , Fibrose , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/terapia , Camundongos , Camundongos Knockout , MicroRNAs/genética , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Contração Miocárdica , Miocárdio/metabolismo , Miocárdio/patologia , Recuperação de Função Fisiológica , Sarcoglicanas/deficiência , Sarcoglicanas/genética , Sarcômeros/metabolismo , Sarcômeros/patologia , Índice de Gravidade de Doença , Fatores de Tempo , Função Ventricular Esquerda , Remodelação Ventricular
6.
Methods Mol Biol ; 1035: 179-90, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23959991

RESUMO

Skeletal muscle tissue has a remarkable capability of regenerating in pathological conditions or after injury. The principal muscle stem cells, satellite cells, are responsible for this prompt and efficient process. Normally quiescent in their niches underneath the basal lamina of each muscle fiber, satellite cells become activated to repair or form new fibers. Ideally, healthy donor stem cells could be transplanted to regenerate the skeletal muscle tissue to repair a genetic defect. However, to be efficient, cell grafting requires modulation of the host muscle environment to allow homing of, and regeneration by, donor satellite cells. Here, we provide methods to modulate the host mouse muscle environment in order to destroy or preserve the muscle niche before transplanting donor satellite cells. We also describe methods to investigate donor-derived muscle regeneration and self-renewal.


Assuntos
Músculo Esquelético/fisiopatologia , Células Satélites de Músculo Esquelético/transplante , Animais , Diferenciação Celular , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/terapia , Regeneração , Medicina Regenerativa , Nicho de Células-Tronco , Condicionamento Pré-Transplante
7.
PLoS One ; 8(1): e54599, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23349935

RESUMO

Skeletal muscle has a remarkable capability of regeneration following injury. Satellite cells, the principal muscle stem cells, are responsible for this process. However, this regenerative capacity is reduced in muscular dystrophies or in old age: in both these situations, there is a net loss of muscle fibres. Promoting skeletal muscle muscle hypertrophy could therefore have potential applications for treating muscular dystrophies or sarcopenia. Here, we observed that muscles of dystrophic mdx nude host mice that had been acutely injured by myotoxin and grafted with a single myofibre derived from a normal donor mouse exhibited increased muscle area. Transplantation experiments revealed that the hypertrophic effect is mediated by the grafted fibre and does not require either an imposed injury to the host muscle, or the contribution of donor cells to the host muscle. These results suggest the presence of a crucial cross-talk between the donor fibre and the host muscle environment.


Assuntos
Músculo Esquelético/transplante , Distrofia Muscular Animal/terapia , Regeneração , Células Satélites de Músculo Esquelético/transplante , Fatores Etários , Animais , Venenos de Crotalídeos/toxicidade , Hipertrofia , Camundongos , Camundongos Endogâmicos mdx , Camundongos Nus , Músculo Esquelético/citologia , Músculo Esquelético/lesões , Distrofia Muscular Animal/induzido quimicamente , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Regeneração/genética , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/citologia
8.
Stem Cells ; 30(10): 2330-41, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22887880

RESUMO

The dystrophin-associated glycoprotein complex (DGC) is found at the muscle fiber sarcolemma and forms an essential structural link between the basal lamina and internal cytoskeleton. In a set of muscular dystrophies known as the dystroglycanopathies, hypoglycosylation of the DGC component α-dystroglycan results in reduced binding to basal lamina components, a loss in structural stability, and repeated cycles of muscle fiber degeneration and regeneration. The satellite cells are the key stem cells responsible for muscle repair and reside between the basal lamina and sarcolemma. In this study, we aimed to determine whether pathological changes associated with the dystroglycanopathies affect satellite cell function. In the Large(myd) mouse dystroglycanopathy model, satellite cells are present in significantly greater numbers but display reduced proliferation on their native muscle fibers in vitro, compared with wild type. However, when removed from their fiber, proliferation in culture is restored to that of wild type. Immunohistochemical analysis of Large(myd) muscle reveals alterations to the basal lamina and interstitium, including marked disorganization of laminin, upregulation of fibronectin and collagens. Proliferation and differentiation of wild-type satellite cells is impaired when cultured on substrates such as collagen and fibronectin, compared with laminins. When engrafted into irradiated tibialis anterior muscles of mdx-nude mice, wild-type satellite cells expanded on laminin contribute significantly more to muscle regeneration than those expanded on fibronectin. These results suggest that defects in α-dystroglycan glycosylation are associated with an alteration in the satellite cell niche, and that regenerative potential in the dystroglycanopathies may be perturbed.


Assuntos
Membrana Basal/metabolismo , Distroglicanas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Animal/metabolismo , Sarcolema/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Animais , Membrana Basal/patologia , Diferenciação Celular , Proliferação de Células , Colágeno/química , Colágeno/metabolismo , Modelos Animais de Doenças , Fibronectinas/química , Fibronectinas/metabolismo , Glicosilação , Humanos , Laminina/química , Laminina/metabolismo , Camundongos , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Distrofia Muscular Animal/patologia , Ligação Proteica , Sarcolema/patologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/transplante
9.
PLoS One ; 7(5): e37950, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22662253

RESUMO

Satellite cells are myogenic cells found between the basal lamina and the sarcolemma of the muscle fibre. Satellite cells are the source of new myofibres; as such, satellite cell transplantation holds promise as a treatment for muscular dystrophies. We have investigated age and sex differences between mouse satellite cells in vitro and assessed the importance of these factors as mediators of donor cell engraftment in an in vivo model of satellite cell transplantation. We found that satellite cell numbers are increased in growing compared to adult and in male compared to female adult mice. We saw no difference in the expression of the myogenic regulatory factors between male and female mice, but distinct profiles were observed according to developmental stage. We show that, in contrast to adult mice, the majority of satellite cells from two week old mice are proliferating to facilitate myofibre growth; however a small proportion of these cells are quiescent and not contributing to this growth programme. Despite observed changes in satellite cell populations, there is no difference in engraftment efficiency either between satellite cells derived from adult or pre-weaned donor mice, male or female donor cells, or between male and female host muscle environments. We suggest there exist two distinct satellite cell populations: one for muscle growth and maintenance and one for muscle regeneration.


Assuntos
Músculo Esquelético/metabolismo , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/metabolismo , Fatores Etários , Animais , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Nus , Fibras Musculares Esqueléticas/metabolismo , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Células Satélites de Músculo Esquelético/transplante , Fatores Sexuais
10.
Stem Cells ; 30(9): 1971-84, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22730231

RESUMO

Stem cell transplantation is already in clinical practice for certain genetic diseases and is a promising therapy for dystrophic muscle. We used the mdx mouse model of Duchenne muscular dystrophy to investigate the effect of the host satellite cell niche on the contribution of donor muscle stem cells (satellite cells) to muscle regeneration. We found that incapacitation of the host satellite cells and preservation of the muscle niche promote donor satellite cell contribution to muscle regeneration and functional reconstitution of the satellite cell compartment. But, if the host niche is not promptly refilled, or is filled by competent host satellite cells, it becomes nonfunctional and donor engraftment is negligible. Application of this regimen to aged host muscles also promotes efficient regeneration from aged donor satellite cells. In contrast, if the niche is destroyed, yet host satellite cells remain proliferation-competent, donor-derived engraftment is trivial. Thus preservation of the satellite cell niche, concomitant with functional impairment of the majority of satellite cells within dystrophic human muscles, may improve the efficiency of stem cell therapy.


Assuntos
Sobrevivência de Enxerto/fisiologia , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Células Satélites de Músculo Esquelético/transplante , Animais , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Células Satélites de Músculo Esquelético/citologia , Transplante de Células-Tronco/métodos
11.
PLoS Curr ; 3: RRN1294, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22333991

RESUMO

Satellite cells, normally quiescent underneath the myofibre basal lamina, are skeletal muscle stem cells responsible for postnatal muscle growth, repair and regeneration. Since their scarcity and small size have limited study on transverse muscle sections, techniques to isolate individual myofibres, bearing their attendant satellite cells, were developed. Studies on mouse myofibres have generated much information on satellite cells, but the limited availability and small size of human muscle biopsies have hampered equivalent studies of satellite cells on human myofibres. Here, we identified satellite cells on fragments of human and mouse myofibres, using a method applicable to small muscle biopsies.

12.
Nucleic Acid Ther ; 21(4): 293-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21851223

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal X-linked inherited disease caused by mutations in the dystrophin gene and consequent lack of dystrophin in the skeletal, cardiac, and smooth musculature and in the nervous system. Patients die during their mid-twenties because of severe muscle loss and life-threatening respiratory and cardiac complications. The splicing modulation approach mediated by antisense oligonucleotides can restore the production of a partially functional quasi-dystrophin in skeletal muscles. We recently showed that a chronic, 12-month treatment with phosphorodiamidate morpholino oligomers efficiently restored dystrophin in widespread skeletal muscles and led to normal locomotor activity indistinguishable from that of dystrophin-expressing C57 mice. However, no detectable dystrophin expression was observed in the hearts of treated mice. In the present study, histological analyses show a more severe cardiac pathology compared with untreated animals in the face of enhanced locomotor behavior. This observation implies that the increase in locomotor activity of treated mdx mice may have a paradoxical detrimental effect on the dystrophic heart. In the context of skeletal muscle-centric therapies for DMD, our data suggest that particular vigilance should be instigated to monitor emergence of accelerated cardiac dysfunction.


Assuntos
Distrofina/genética , Éxons/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Morfolinos/uso terapêutico , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/tratamento farmacológico , Miocárdio/patologia , Animais , Cardiomiopatias/metabolismo , Cardiomiopatias/prevenção & controle , Relação Dose-Resposta a Droga , Distrofina/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Masculino , Camundongos , Camundongos Endogâmicos mdx , Morfolinos/administração & dosagem , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miocárdio/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo
13.
Hum Gene Ther ; 22(11): 1379-88, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21453126

RESUMO

Duchenne muscular dystrophy is a severe X-linked inherited muscle wasting disorder caused by mutations in the dystrophin gene. Adeno-associated virus (AAV) vectors have been extensively used to deliver genes efficiently for dystrophin expression in skeletal muscles. To overcome limited packaging capacity of AAV vectors (<5 kb), truncated recombinant microdystrophin genes with deletions of most of rod and carboxyl-terminal (CT) domains of dystrophin have been developed. We have previously shown the efficiency of mRNA sequence-optimized microdystrophin (ΔR4-23/ΔCT, called MD1) with deletion of spectrin-like repeat domain 4 to 23 and CT domain in ameliorating the pathology of dystrophic mdx mice. However, the CT domain of dystrophin is thought to recruit part of the dystrophin-associated protein complex, which acts as a mediator of signaling between extracellular matrix and cytoskeleton in muscle fibers. In this study, we extended the ΔR4-23/ΔCT microdystrophin by incorporating helix 1 of the coiled-coil motif in the CT domain of dystrophin (MD2), which contains the α1-syntrophin and α-dystrobrevin binding sites. Intramuscular injection of AAV2/9 expressing CT domain-extended microdystrophin showed efficient dystrophin expression in tibialis anterior muscles of mdx mice. The presence of the CT domain of dystrophin in MD2 increased the recruitment of α1-syntrophin and α-dystrobrevin at the sarcolemma and significantly improved the muscle resistance to lengthening contraction-induced muscle damage in the mdx mice compared with MD1. These results suggest that the incorporation of helix 1 of the coiled-coil motif in the CT domain of dystrophin to the microdystrophins will substantially improve their efficiency in restoring muscle function in patients with Duchenne muscular dystrophy.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Dependovirus/genética , Proteínas Associadas à Distrofina/metabolismo , Distrofina/genética , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Motivos de Aminoácidos , Animais , Distrofina/metabolismo , Terapia Genética , Vetores Genéticos , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia
14.
J Histochem Cytochem ; 58(11): 941-55, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20644208

RESUMO

Satellite cells are quiescent cells located under the basal lamina of skeletal muscle fibers that contribute to muscle growth, maintenance, repair, and regeneration. Mouse satellite cells have been shown to be muscle stem cells that are able to regenerate muscle fibers and self-renew. As human skeletal muscle is also able to regenerate following injury, we assume that the human satellite cell is, like its murine equivalent, a muscle stem cell. In this review, we compare human and mouse satellite cells and highlight their similarities and differences. We discuss gaps in our knowledge of human satellite cells, compared with that of mouse satellite cells, and suggest ways in which we may advance studies on human satellite cells, particularly by finding new markers and attempting to re-create the human satellite cell niche in vitro.


Assuntos
Células Satélites de Músculo Esquelético , Animais , Proliferação de Células , Senescência Celular , Exercício Físico/fisiologia , Humanos , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Regeneração , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia
15.
Dev Biol ; 337(1): 29-41, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19835858

RESUMO

Skeletal muscles of body and limb are derived from somites, but most head muscles originate from cranial mesoderm. The resident stem cells of muscle are satellite cells, which have the same embryonic origin as the muscle in which they reside. Here, we analysed satellite cells with a different ontology, comparing those of the extensor digitorum longus (EDL) of the limb with satellite cells from the masseter of the head. Satellite cell-derived myoblasts from MAS and EDL muscles had distinct gene expression profiles and masseter cells usually proliferated more and differentiated later than those from EDL. When transplanted, however, masseter-derived satellite cells regenerated limb muscles as efficiently as those from EDL. Clonal analysis showed that functional properties differed markedly between satellite cells: ranging from clones that proliferated extensively and gave rise to both differentiated and self-renewed progeny, to others that divided minimally before differentiating completely. Generally, masseter-derived clones were larger and took longer to differentiate than those from EDL. This distribution in cell properties was preserved in both EDL-derived and masseter-derived satellite cells from old mice, although clones were generally less proliferative. Satellite cells, therefore, are a functionally heterogeneous population, with many occupants of the niche exhibiting stem cell characteristics in both somite-derived and branchiomeric muscles.


Assuntos
Células Satélites de Músculo Esquelético/fisiologia , Somitos/citologia , Envelhecimento/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/citologia , Mioblastos/metabolismo , Regeneração , Células-Tronco/fisiologia
16.
Neuromuscul Disord ; 20(1): 6-15, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20034794

RESUMO

Stem cell therapy holds promise for treating muscle diseases. Although satellite cells regenerate skeletal muscle, they only have a local effect after intra-muscular transplantation. Alternative cell types, more easily obtainable and systemically-deliverable, were therefore sought. Human synovial stem cells (hSSCs) have been reported to regenerate muscle fibres and reconstitute the satellite cell pool. We therefore determined if these cells are able to regenerate skeletal muscle after intra-muscular injection into cryodamaged muscles of Rag2-/gamma chain-/C5-mice. We found that hSSCs possess only limited capacity to undergo myogenic differentiation in vitro or to contribute to muscle regeneration in vivo. However, this is enhanced by over-expression of human MyoD1. Interestingly, hSSCs express extracellular matrix components laminin alpha2 and collagen VI within grafted muscles. Therefore, despite their limited capacity to regenerate skeletal muscle, hSSCs could play a role in treating muscular dystrophies secondary to defects in extracellular matrix proteins.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Músculo Esquelético/fisiologia , Regeneração , Membrana Sinovial/citologia , Adolescente , Animais , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Criança , Temperatura Baixa , Colágeno Tipo VI/metabolismo , Humanos , Laminina/metabolismo , Camundongos , Músculo Esquelético/lesões , Distrofia Muscular de Duchenne/fisiopatologia , Distrofia Muscular de Duchenne/cirurgia , Proteína MyoD/metabolismo , Membrana Sinovial/fisiologia , Transplante Heterólogo
17.
Stem Cells ; 27(10): 2478-87, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19575422

RESUMO

Changes that occur in the skeletal muscle environment with the progress of muscular dystrophies may affect stem cell function and result in impaired muscle regeneration. It has previously been suggested that the success of stem cell transplantation could therefore be dependent both on the properties of the cell itself and on the host muscle environment. Here we engrafted young and mature adult mdx-nude mice, which are the genetic homolog of Duchenne muscular dystrophy, with a small number of satellite cells freshly isolated from young, normal donor mice. We found that the donor satellite cells contributed to muscle regeneration and self-renewal as efficiently within mature adult, as in young, dystrophic host muscle. Donor-derived satellite cells also contributed to robust regeneration after further injury, showing that they were functional despite the more advanced dystrophic muscle environment. These findings provide evidence that muscle tissue in a later stage of dystrophy may be effectively treated by stem cells.


Assuntos
Sobrevivência de Enxerto/fisiologia , Distrofia Muscular Animal/cirurgia , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Células Satélites de Músculo Esquelético/transplante , Transplante de Células-Tronco/métodos , Fatores Etários , Animais , Comunicação Celular/fisiologia , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Células Satélites de Músculo Esquelético/citologia , Resultado do Tratamento
18.
Exp Cell Res ; 315(6): 915-27, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19371636

RESUMO

Skeletal muscle regeneration relies on satellite cells, a population of myogenic precursors. Inflammation also plays a determinant role in the process, as upon injury, macrophages are attracted by the damaged myofibers and the activated satellite cells and act as key elements of dynamic muscle supportive stroma. Yet, it is not known how macrophages interact with the more profound stem cells of the satellite cell niche. Here we show that in the presence of a murine macrophage conditioned medium (mMCM) a subpopulation of multipotent cells could be selected and expanded from adult rat muscle. These cells were small, round, poorly adhesive, slow-growing and showed mesenchymal differentiation plasticity. At the same time, mMCM showed clear myogenic capabilities, as experiments with satellite cells mechanically isolated from suspensions of single myofibers showed that the macrophagic factors inhibited their tendency to shift towards adipogenesis. In vivo, intramuscular administrations of concentrated mMCM in a rat model of extensive surgical ablation dramatically improved muscle regeneration. Altogether, these findings suggest that macrophagic factors could be of great help in developing therapeutic protocols with myogenic stem cells.


Assuntos
Meios de Cultivo Condicionados/química , Macrófagos/metabolismo , Células-Tronco Multipotentes/fisiologia , Desenvolvimento Muscular/fisiologia , Músculo Esquelético , Células Satélites de Músculo Esquelético/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células , Humanos , Macrófagos/citologia , Masculino , Camundongos , Células-Tronco Multipotentes/citologia , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Ratos , Ratos Wistar , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/citologia
19.
Tissue Eng Part A ; 15(9): 2447-57, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19292666

RESUMO

An in vitro muscle-like structure with parallel-oriented contractile myotubes is needed as a model of muscle tissue regeneration. For this purpose, it is necessary to reproduce a controllable microscale environment mimicking the in vivo cues. In this work we focused on the application of topological and electrical stimuli on muscle precursor cell (MPC) culture to influence MPC orientation and induce myotube alignment. The two stimulations were tested both independently and together. A structural and topological template was achieved using micropatterned poly-(L-lactic acid) membranes. Electrical stimulation, consisting of square pulses of 70 mV/cm amplitude each 30 s, was applied to the MPC culture. The effect of different pulse durations on cultures was evaluated by galvanotaxis analysis. The highest cell displacement rate toward the cathode was observed for 3 ms pulse stimulation, which was then applied in combination with topological stimuli. Topological and electrical stimuli had an additive effect in enhancing differentiation of cultured MPC, shown by high Troponin I protein production and, in parallel, Myogenin and Desmin genes, down- and upregulation respectively.


Assuntos
Diferenciação Celular , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Engenharia Tecidual/métodos , Animais , Diferenciação Celular/efeitos dos fármacos , Separação Celular , Células Cultivadas , Desmina/genética , Desmina/metabolismo , Estimulação Elétrica , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Láctico/farmacologia , Membranas Artificiais , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Dióxido de Nitrogênio/metabolismo , Poliésteres , Polímeros/farmacologia , Ratos , Ratos Wistar , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Propriedades de Superfície/efeitos dos fármacos , Troponina I/metabolismo
20.
PLoS One ; 4(2): e4475, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19221588

RESUMO

Pax3 and Pax7 are paired-box transcription factors with roles in developmental and adult regenerative myogenesis. Pax3 and Pax7 are expressed by postnatal satellite cells or their progeny but are down regulated during myogenic differentiation. We now show that constitutive expression of Pax3 or Pax7 in either satellite cells or C2C12 myoblasts results in an increased proliferative rate and decreased cell size. Conversely, expression of dominant-negative constructs leads to slowing of cell division, a dramatic increase in cell size and altered morphology. Similarly to the effects of Pax7, retroviral expression of Pax3 increases levels of Myf5 mRNA and MyoD protein, but does not result in sustained inhibition of myogenic differentiation. However, expression of Pax3 or Pax7 dominant-negative constructs inhibits expression of Myf5, MyoD and myogenin, and prevents differentiation from proceeding. In fibroblasts, expression of Pax3 or Pax7, or dominant-negative inhibition of these factors, reproduce the effects on cell size, morphology and proliferation seen in myoblasts. Our results show that in muscle progenitor cells, Pax3 and Pax7 function to maintain expression of myogenic regulatory factors, and promote population expansion, but are also required for myogenic differentiation to proceed.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Tamanho Celular , Desenvolvimento Muscular/fisiologia , Mioblastos/fisiologia , Fator de Transcrição PAX7/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Animais , Divisão Celular/fisiologia , Linhagem Celular , Forma Celular , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos/citologia , Fator de Transcrição PAX3 , Fator de Transcrição PAX7/genética , Fatores de Transcrição Box Pareados/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...