Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
EMBO J ; 41(14): e110655, 2022 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-35703167

RESUMO

Fate decisions in the embryo are controlled by a plethora of microenvironmental interactions in a three-dimensional niche. To investigate whether aspects of this microenvironmental complexity can be engineered to direct myogenic human-induced pluripotent stem cell (hiPSC) differentiation, we here screened murine cell types present in the developmental or adult stem cell niche in heterotypic suspension embryoids. We identified embryonic endothelial cells and fibroblasts as highly permissive for myogenic specification of hiPSCs. After two weeks of sequential Wnt and FGF pathway induction, these three-component embryoids are enriched in Pax7-positive embryonic-like myogenic progenitors that can be isolated by flow cytometry. Myogenic differentiation of hiPSCs in heterotypic embryoids relies on a specialized structural microenvironment and depends on MAPK, PI3K/AKT, and Notch signaling. After transplantation in a mouse model of Duchenne muscular dystrophy, embryonic-like myogenic progenitors repopulate the stem cell niche, reactivate after repeated injury, and, compared to adult human myoblasts, display enhanced fusion and lead to increased muscle function. Altogether, we provide a two-week protocol for efficient and scalable suspension-based 3D derivation of Pax7-positive myogenic progenitors from hiPSCs.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular , Células Endoteliais , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Desenvolvimento Muscular , Fosfatidilinositol 3-Quinases/metabolismo , Nicho de Células-Tronco
2.
Biochem J ; 466(1): 123-35, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25431931

RESUMO

The protein mammalian target of rapamycin (mTOR) is a serine/threonine kinase regulating a number of biochemical pathways controlling cell growth. mTOR exists in two complexes termed mTORC1 and mTORC2. Regulatory associated protein of mTOR (raptor) is associated with mTORC1 and is essential for its function. Ablation of raptor in skeletal muscle results in several phenotypic changes including decreased life expectancy, increased glycogen deposits and alterations of the twitch kinetics of slow fibres. In the present paper, we show that in muscle-specific raptor knockout (RamKO), the bulk of glycogen phosphorylase (GP) is mainly associated in its cAMP-non-stimulated form with sarcoplasmic reticulum (SR) membranes. In addition, 3[H]-ryanodine and 3[H]-PN200-110 equilibrium binding show a ryanodine to dihydropyridine receptors (DHPRs) ratio of 0.79 and 1.35 for wild-type (WT) and raptor KO skeletal muscle membranes respectively. Peak amplitude and time to peak of the global calcium transients evoked by supramaximal field stimulation were not different between WT and raptor KO. However, the increase in the voltage sensor-uncoupled RyRs leads to an increase of both frequency and mass of elementary calcium release events (ECRE) induced by hyper-osmotic shock in flexor digitorum brevis (FDB) fibres from raptor KO. The present study shows that the protein composition and function of the molecular machinery involved in skeletal muscle excitation-contraction (E-C) coupling is affected by mTORC1 signalling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Canais de Cálcio Tipo L/genética , Complexos Multiproteicos/genética , Músculo Esquelético/metabolismo , Retículo Sarcoplasmático/metabolismo , Serina-Treonina Quinases TOR/genética , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Potenciais Evocados/fisiologia , Acoplamento Excitação-Contração/fisiologia , Regulação da Expressão Gênica , Glicogênio Fosforilase/genética , Glicogênio Fosforilase/metabolismo , Contração Isométrica , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Proteína Regulatória Associada a mTOR , Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
3.
EMBO Rep ; 14(12): 1062-72, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24232182

RESUMO

Satellite cells, the quintessential skeletal muscle stem cells, reside in a specialized local environment whose anatomy changes dynamically during tissue regeneration. The plasticity of this niche is attributable to regulation by the stem cells themselves and to a multitude of functionally diverse cell types. In particular, immune cells, fibrogenic cells, vessel-associated cells and committed and differentiated cells of the myogenic lineage have emerged as important constituents of the satellite cell niche. Here, we discuss the cellular dynamics during muscle regeneration and how disease can lead to perturbation of these mechanisms. To define the role of cellular components in the muscle stem cell niche is imperative for the development of cell-based therapies, as well as to better understand the pathobiology of degenerative conditions of the skeletal musculature.


Assuntos
Desenvolvimento Muscular , Músculo Esquelético/fisiologia , Regeneração , Células Satélites de Músculo Esquelético/metabolismo , Nicho de Células-Tronco , Animais , Humanos , Músculo Esquelético/citologia , Músculo Esquelético/crescimento & desenvolvimento , Células Satélites de Músculo Esquelético/fisiologia
4.
Bioessays ; 35(3): 231-41, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22886714

RESUMO

Cell-based therapies for degenerative diseases of the musculature remain on the verge of feasibility. Myogenic cells are relatively abundant, accessible, and typically harbor significant proliferative potential ex vivo. However, their use for therapeutic intervention is limited due to several critical aspects of their complex biology. Recent insights based on mouse models have advanced our understanding of the molecular mechanisms controlling the function of myogenic progenitors significantly. Moreover, the discovery of atypical myogenic cell types with the ability to cross the blood-muscle barrier has opened exciting new therapeutic avenues. In this paper, we outline the major problems that are currently associated with the manipulation of myogenic cells and discuss promising strategies to overcome these obstacles.


Assuntos
Músculo Esquelético/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Comunicação Celular , Humanos , Células Satélites de Músculo Esquelético/citologia , Nicho de Células-Tronco
5.
Proc Natl Acad Sci U S A ; 108(51): 20808-13, 2011 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-22143799

RESUMO

Mammalian target of rapamycin complex 1 (mTORC1) is central to the control of cell, organ, and body size. Skeletal muscle-specific inactivation of mTORC1 in mice results in smaller muscle fibers, fewer mitochondria, increased glycogen stores, and a progressive myopathy that causes premature death. In mTORC1-deficient muscles, peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), which regulates mitochondrial biogenesis and glucose homeostasis, is strongly down-regulated. Here we tested whether induction of mitochondrial biogenesis pharmacologically or by the overexpression of PGC-1α is sufficient to reverse the phenotype of mice deficient for mTORC1. We show that both approaches normalize mitochondrial function, such as oxidative capacity and expression of mitochondrial genes. However, they do not prevent or delay the progressive myopathy. In addition, we find that mTORC1 has a much stronger effect than PGC-1α on the glycogen content in muscle. This effect is based on the strong activation of PKB/Akt in mTORC1-deficient mice. We also show that activation of PKB/Akt not only affects glycogen synthesis but also diminishes glycogen degradation. Thus, our work provides strong functional evidence that mitochondrial dysfunction in mice with inactivated mTORC1 signaling is caused by the down-regulation of PGC-1α. However, our data also show that the impairment of mitochondria does not lead directly to the lethal myopathy.


Assuntos
Bezafibrato/farmacologia , Regulação da Expressão Gênica , Doenças Musculares/metabolismo , Proteínas/metabolismo , Animais , Glicogênio/química , Glicogênio/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mitocôndrias Musculares/metabolismo , Modelos Genéticos , Complexos Multiproteicos , Músculo Esquelético/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Serina-Treonina Quinases TOR , Transativadores/metabolismo , Fatores de Transcrição
6.
Stem Cell Reports ; 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38876109

RESUMO

Increasing evidence suggests that the muscle stem cell (MuSC) pool is heterogeneous. In particular, a rare subset of PAX7-positive MuSCs that has never expressed the myogenic regulatory factor MYF5 displays unique self-renewal and engraftment characteristics. However, the scarcity and limited availability of protein markers make the characterization of these cells challenging. Here, we describe the generation of StemRep reporter mice enabling the monitoring of PAX7 and MYF5 proteins based on equimolar levels of dual nuclear fluorescence. High levels of PAX7 protein and low levels of MYF5 delineate a deeply quiescent MuSC subpopulation with an increased capacity for asymmetric division and distinct dynamics of activation, proliferation, and commitment. Aging primarily reduces the MYF5Low MuSCs and skews the stem cell pool toward MYF5High cells with lower quiescence and self-renewal potential. Altogether, we establish the StemRep model as a versatile tool to study MuSC heterogeneity and broaden our understanding of mechanisms regulating MuSC quiescence and self-renewal in homeostatic, regenerating, and aged muscles.

7.
Sci Transl Med ; 16(739): eabn8529, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38507466

RESUMO

Impaired skeletal muscle stem cell (MuSC) function has long been suspected to contribute to the pathogenesis of muscular dystrophy (MD). Here, we showed that defects in the endothelial cell (EC) compartment of the vascular stem cell niche in mouse models of Duchenne MD, laminin α2-related MD, and collagen VI-related myopathy were associated with inefficient mobilization of MuSCs after tissue damage. Using chemoinformatic analysis, we identified the 13-amino acid form of the peptide hormone apelin (AP-13) as a candidate for systemic stimulation of skeletal muscle ECs. Systemic administration of AP-13 using osmotic pumps generated a pro-proliferative EC-rich niche that supported MuSC function through angiocrine factors and markedly improved tissue regeneration and muscle strength in all three dystrophic mouse models. Moreover, EC-specific knockout of the apelin receptor led to regenerative defects that phenocopied key pathological features of MD, including vascular defects, fibrosis, muscle fiber necrosis, impaired MuSC function, and reduced force generation. Together, these studies provide in vivo proof of concept that enhancing endogenous skeletal muscle repair by targeting the vascular niche is a viable therapeutic avenue for MD and characterized AP-13 as a candidate for further study for the systemic treatment of MuSC dysfunction.


Assuntos
Distrofia Muscular de Duchenne , Nicho de Células-Tronco , Camundongos , Animais , Apelina/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Transdução de Sinais
8.
Front Cell Dev Biol ; 10: 1056523, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36523505

RESUMO

The extracellular matrix (ECM) is an interconnected macromolecular scaffold occupying the space between cells. Amongst other functions, the ECM provides structural support to tissues and serves as a microenvironmental niche that conveys regulatory signals to cells. Cell-matrix adhesions, which link the ECM to the cytoskeleton, are dynamic multi-protein complexes containing surface receptors and intracellular effectors that control various downstream pathways. In skeletal muscle, the most abundant tissue of the body, each individual muscle fiber and its associated muscle stem cells (MuSCs) are surrounded by a layer of ECM referred to as the basal lamina. The core scaffold of the basal lamina consists of self-assembling polymeric laminins and a network of collagens that tether proteoglycans, which provide lateral crosslinking, establish collateral associations with cell surface receptors, and serve as a sink and reservoir for growth factors. Skeletal muscle also contains the fibrillar collagenous interstitial ECM that plays an important role in determining tissue elasticity, connects the basal laminae to each other, and contains matrix secreting mesenchymal fibroblast-like cell types and blood vessels. During skeletal muscle regeneration fibroblast-like cell populations expand and contribute to the transitional fibronectin-rich regenerative matrix that instructs angiogenesis and MuSC function. Here, we provide a comprehensive overview of the role of the skeletal muscle ECM in health and disease and outline its role in orchestrating tissue regeneration and MuSC function.

9.
Elife ; 112022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35245177

RESUMO

Sustained exposure to a young systemic environment rejuvenates aged organisms and promotes cellular function. However, due to the intrinsic complexity of tissues it remains challenging to pinpoint niche-independent effects of circulating factors on specific cell populations. Here, we describe a method for the encapsulation of human and mouse skeletal muscle progenitors in diffusible polyethersulfone hollow fiber capsules that can be used to profile systemic aging in vivo independent of heterogeneous short-range tissue interactions. We observed that circulating long-range signaling factors in the old systemic environment lead to an activation of Myc and E2F transcription factors, induce senescence, and suppress myogenic differentiation. Importantly, in vitro profiling using young and old serum in 2D culture does not capture all pathways deregulated in encapsulated cells in aged mice. Thus, in vivo transcriptomic profiling using cell encapsulation allows for the characterization of effector pathways of systemic aging with unparalleled accuracy.


Assuntos
Células Satélites de Músculo Esquelético , Células-Tronco , Envelhecimento , Animais , Diferenciação Celular , Encapsulamento de Células , Camundongos , Músculo Esquelético/metabolismo , Células-Tronco/metabolismo , Transcriptoma
10.
Curr Protoc ; 1(10): e263, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34612611

RESUMO

Skeletal muscle stem cells (MuSCs) reside in a complex niche composed of the muscle fiber plasma membrane and the laminin-rich basal lamina surrounded by the microvasculature, as well as different supportive cell types such as fibro-adipogenic progenitors residing in the interstitial extracellular matrix. Within the first few hours after tissue damage, MuSCs undergo cytoskeletal rearrangements and transcriptional changes that prime the cells for activation. Due to their time-consuming nature, enzymatic methods for liberation of single muscle fibers with fully quiescent MuSCs are challenging. Moreover, during enzymatic digestion, important niche components including the microvasculature and the collagenous interstitial matrix are destroyed. Here, we provide a method for the visualization of MuSCs on muscle fibers in their intact niche. Our method relies on mechanical teasing of fiber bundles from fixed skeletal muscles. We demonstrate that teased muscle fiber bundles allow the investigator to capture a representative snapshot of the MuSC niche in skeletal muscle, and outline how stem cell morphology and different microenvironmental components can be visualized. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Isolation of fiber bundles Basic Protocol 2: Immunofluorescence staining of MuSCs on fiber bundles Support Protocol: Preparation of Sylgard dishes.


Assuntos
Músculo Esquelético , Nicho de Células-Tronco , Adipogenia , Mioblastos , Células-Tronco
11.
Cell Stem Cell ; 24(3): 433-446.e7, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30686765

RESUMO

Research on age-related regenerative failure of skeletal muscle has extensively focused on the phenotypes of muscle stem cells (MuSCs). In contrast, the impact of aging on regulatory cells in the MuSC niche remains largely unexplored. Here, we demonstrate that aging impairs the function of mouse fibro-adipogenic progenitors (FAPs) and thereby indirectly affects the myogenic potential of MuSCs. Using transcriptomic profiling, we identify WNT1 Inducible Signaling Pathway Protein 1 (WISP1) as a FAP-derived matricellular signal that is lost during aging. WISP1 is required for efficient muscle regeneration and controls the expansion and asymmetric commitment of MuSCs through Akt signaling. Transplantation of young FAPs or systemic treatment with WISP1 restores the myogenic capacity of MuSCs in aged mice and rescues skeletal muscle regeneration. Our work establishes that loss of WISP1 from FAPs contributes to MuSC dysfunction in aged skeletal muscles and demonstrates that this mechanism can be targeted to rejuvenate myogenesis.


Assuntos
Adipócitos/metabolismo , Envelhecimento/metabolismo , Proteínas de Sinalização Intercelular CCN/metabolismo , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Células-Tronco/metabolismo , Adipócitos/citologia , Adipogenia , Animais , Proteínas de Sinalização Intercelular CCN/deficiência , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/citologia , Proteínas Proto-Oncogênicas/deficiência , Células-Tronco/citologia
12.
Curr Top Dev Biol ; 126: 23-65, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29305000

RESUMO

The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.


Assuntos
Músculo Esquelético/fisiologia , Doenças Musculares/fisiopatologia , Células Satélites de Músculo Esquelético/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Humanos , Músculo Esquelético/citologia , Doenças Musculares/patologia , Regeneração , Células Satélites de Músculo Esquelético/citologia , Células-Tronco/citologia
13.
Cell Rep ; 18(10): 2320-2330, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28273449

RESUMO

Wnt-mediated signals are involved in many important steps in mammalian regeneration. In multiple cell types, the R-spondin (Rspo) family of secreted proteins potently activates the canonical Wnt/ß-catenin pathway. Here, we identify Rspo1 as a mediator of skeletal muscle tissue repair. First, we show that deletion of Rspo1 results in global alteration of muscle regeneration kinetics following acute injury. We find that muscle progenitor cells lacking Rspo1 show delayed differentiation due to reduced activation of Wnt/ß-catenin target genes. Furthermore, muscle cells lacking Rspo1 have a fusion phenotype leading to larger myotubes containing supernumerary nuclei both in vitro and in vivo. The increase in muscle fusion was dependent on downregulation of Wnt/ß-catenin and upregulation of non-canonical Wnt7a/Fzd7/Rac1 signaling. We conclude that reciprocal control of antagonistic Wnt signaling pathways by Rspo1 in muscle stem cell progeny is a key step ensuring normal tissue architecture restoration following acute damage.


Assuntos
Mioblastos/citologia , Mioblastos/metabolismo , Trombospondinas/metabolismo , Via de Sinalização Wnt , Animais , Diferenciação Celular , Fusão Celular , Proliferação de Células , Células Cultivadas , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular , Fator de Transcrição PAX7/metabolismo , Regeneração , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , beta Catenina/metabolismo
14.
FASEB J ; 19(8): 934-42, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15923403

RESUMO

Mutations in the gene encoding the alpha2 subunit of laminins cause the severe "merosin-deficient congenital muscular dystrophy" (MDC1A). We have recently shown that overexpression of a miniaturized form of the molecule agrin (mini-agrin) counteracts the disease in dy(W)/dy(W) mice, a model for MDC1A. However, these mice express some residual truncated laminin-alpha2, suggesting that the observed amelioration might be due to mini-agrin's presenting the residual laminin-alpha2 to its receptors. Here we show that the mini-agrin counteracts the disease in dy(3K)/dy(3K) mice, which are null for laminin-alpha2. As in dy(W)/dy(W) mice, mini-agrin improves both the function and structure of muscle. We show that muscle regeneration after injury is severely impaired in dy(3K)/dy(3K) mice but is restored in the mini-agrin-expressing littermates. In summary, our results 1) show that the direct linkage of muscle basal lamina with the sarcolemma is the basis of mini-agrin-mediated amelioration and 2) provide unprecedented evidence that this linkage is important for proper regeneration of muscle fibers after injury. Our findings thus suggest that treatment with mini-agrin might be beneficial over the entire spectrum of the MDC1A disease, whose severity inversely correlates with expression levels and the size of the truncation in laminin-alpha2.


Assuntos
Agrina/genética , Agrina/fisiologia , Laminina/deficiência , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal , Regeneração/fisiologia , Animais , Membrana Basal/fisiopatologia , Membrana Basal/ultraestrutura , Distroglicanas/análise , Regulação da Expressão Gênica , Imuno-Histoquímica , Laminina/análise , Laminina/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora , Músculo Esquelético/química , Músculo Esquelético/ultraestrutura , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular Animal/prevenção & controle , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcolema/fisiologia , Sarcolema/ultraestrutura
15.
Nat Med ; 22(8): 897-905, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27376579

RESUMO

Age-related changes in the niche have long been postulated to impair the function of somatic stem cells. Here we demonstrate that the aged stem cell niche in skeletal muscle contains substantially reduced levels of fibronectin (FN), leading to detrimental consequences for the function and maintenance of muscle stem cells (MuSCs). Deletion of the gene encoding FN from young regenerating muscles replicates the aging phenotype and leads to a loss of MuSC numbers. By using an extracellular matrix (ECM) library screen and pathway profiling, we characterize FN as a preferred adhesion substrate for MuSCs and demonstrate that integrin-mediated signaling through focal adhesion kinase and the p38 mitogen-activated protein kinase pathway is strongly de-regulated in MuSCs from aged mice because of insufficient attachment to the niche. Reconstitution of FN levels in the aged niche remobilizes stem cells and restores youth-like muscle regeneration. Taken together, we identify the loss of stem cell adhesion to FN in the niche ECM as a previously unknown aging mechanism.


Assuntos
Envelhecimento/metabolismo , Fibronectinas/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Músculo Esquelético/metabolismo , Regeneração/genética , Nicho de Células-Tronco , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Western Blotting , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Citometria de Fluxo , Integrinas/metabolismo , Camundongos , Músculo Esquelético/citologia , Reação em Cadeia da Polimerase
16.
Compr Physiol ; 5(3): 1027-59, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26140708

RESUMO

Skeletal muscles are essential for vital functions such as movement, postural support, breathing, and thermogenesis. Muscle tissue is largely composed of long, postmitotic multinucleated fibers. The life-long maintenance of muscle tissue is mediated by satellite cells, lying in close proximity to the muscle fibers. Muscle satellite cells are a heterogeneous population with a small subset of muscle stem cells, termed satellite stem cells. Under homeostatic conditions all satellite cells are poised for activation by stimuli such as physical trauma or growth signals. After activation, satellite stem cells undergo symmetric divisions to expand their number or asymmetric divisions to give rise to cohorts of committed satellite cells and thus progenitors. Myogenic progenitors proliferate, and eventually differentiate through fusion with each other or to damaged fibers to reconstitute fiber integrity and function. In the recent years, research has begun to unravel the intrinsic and extrinsic mechanisms controlling satellite cell behavior. Nonetheless, an understanding of the complex cellular and molecular interactions of satellite cells with their dynamic microenvironment remains a major challenge, especially in pathological conditions. The goal of this review is to comprehensively summarize the current knowledge on satellite cell characteristics, functions, and behavior in muscle regeneration and in pathological conditions.


Assuntos
Diferenciação Celular , Músculo Esquelético/citologia , Regeneração , Células Satélites de Músculo Esquelético/citologia , Animais , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Células Satélites de Músculo Esquelético/metabolismo
17.
Skelet Muscle ; 5: 39, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26635949

RESUMO

BACKGROUND: The mammalian esophageal musculature is unique in that it makes a transition from smooth to skeletal muscle, with most of this process occurring after birth. In order to better understand the mechanisms that control esophageal musculature development, we investigated the roles in this process of the paired box transcription factor, PAX7, a principal regulator of skeletal myogenic progenitor cells. Previous studies showed that Pax7 is important for determining the esophageal muscle composition. RESULTS: We characterized the postnatal development of the esophageal musculature in Pax7 (-/-) mice by analyzing morphology, muscle composition, and the expression of markers of myogenesis, cell proliferation, and apoptosis. Pax7 (-/-) mice displayed megaesophagus with a severe defect in the postnatal developmental process whereby esophageal smooth muscle is replaced by skeletal muscle. Pax7 (-/-) esophagi have substantially reduced skeletal muscle, most likely due to diminished proliferation and premature differentiation of skeletal muscle precursor cells. This impaired the proximal-to-distal progression of skeletal myogenesis and indirectly affected the patterning of the smooth muscle-containing portion of the esophageal musculature. CONCLUSIONS: Postnatal patterning of the esophageal musculature appears to require robust, PAX7-dependent cell proliferation to drive the proximal-to-distal progression of skeletal myogenesis. This process in turn influences distal smooth muscle morphogenesis and development of the mature pattern of the esophageal musculature.

18.
Nat Med ; 21(12): 1455-63, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26569381

RESUMO

Dystrophin is expressed in differentiated myofibers, in which it is required for sarcolemmal integrity, and loss-of-function mutations in the gene that encodes it result in Duchenne muscular dystrophy (DMD), a disease characterized by progressive and severe skeletal muscle degeneration. Here we found that dystrophin is also highly expressed in activated muscle stem cells (also known as satellite cells), in which it associates with the serine-threonine kinase Mark2 (also known as Par1b), an important regulator of cell polarity. In the absence of dystrophin, expression of Mark2 protein is downregulated, resulting in the inability to localize the cell polarity regulator Pard3 to the opposite side of the cell. Consequently, the number of asymmetric divisions is strikingly reduced in dystrophin-deficient satellite cells, which also display a loss of polarity, abnormal division patterns (including centrosome amplification), impaired mitotic spindle orientation and prolonged cell divisions. Altogether, these intrinsic defects strongly reduce the generation of myogenic progenitors that are needed for proper muscle regeneration. Therefore, we conclude that dystrophin has an essential role in the regulation of satellite cell polarity and asymmetric division. Our findings indicate that muscle wasting in DMD not only is caused by myofiber fragility, but also is exacerbated by impaired regeneration owing to intrinsic satellite cell dysfunction.


Assuntos
Divisão Celular Assimétrica , Polaridade Celular , Distrofina/metabolismo , Músculo Esquelético/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Separação Celular , Distrofina/deficiência , Citometria de Fluxo , Camundongos Endogâmicos mdx , Análise de Sequência com Séries de Oligonucleotídeos , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Regeneração , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Fuso Acromático/metabolismo
20.
Nat Med ; 20(10): 1174-81, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25194569

RESUMO

Diminished regenerative capacity of skeletal muscle occurs during adulthood. We identified a reduction in the intrinsic capacity of mouse adult satellite cells to contribute to muscle regeneration and repopulation of the niche. Gene expression analysis identified higher expression of JAK-STAT signaling targets in 3-week [corrected] 18-month-old mice [corrected]. Knockdown of Jak2 or Stat3 significantly stimulated symmetric satellite stem cell divisions on cultured myofibers. Genetic knockdown of Jak2 or Stat3 expression in prospectively isolated satellite cells markedly enhanced their ability to repopulate the satellite cell niche after transplantation into regenerating tibialis anterior muscle. Pharmacological inhibition of Jak2 and Stat3 activity similarly stimulated symmetric expansion of satellite cells in vitro and their engraftment in vivo. Intramuscular injection of these drugs resulted in a marked enhancement of muscle repair and force generation after cardiotoxin injury. Together these results reveal age-related intrinsic properties that functionally distinguish satellite cells and suggest a promising therapeutic avenue for the treatment of muscle-wasting diseases.


Assuntos
Janus Quinases/antagonistas & inibidores , Fatores de Transcrição STAT/antagonistas & inibidores , Células Satélites de Músculo Esquelético/fisiologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Feminino , Técnicas de Silenciamento de Genes , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Transgênicos , RNA Interferente Pequeno/genética , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Células Satélites de Músculo Esquelético/transplante , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA