Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Development ; 147(9)2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32198156

RESUMO

Murine muscle stem cells (MuSCs) experience a transition from quiescence to activation that is required for regeneration, but it remains unknown if the trajectory and dynamics of activation change with age. Here, we use time-lapse imaging and single cell RNA-seq to measure activation trajectories and rates in young and aged MuSCs. We find that the activation trajectory is conserved in aged cells, and we develop effective machine-learning classifiers for cell age. Using cell-behavior analysis and RNA velocity, we find that activation kinetics are delayed in aged MuSCs, suggesting that changes in stem cell dynamics may contribute to impaired stem cell function with age. Intriguingly, we also find that stem cell activation appears to be a random walk-like process, with frequent reversals, rather than a continuous linear progression. These results support a view of the aged stem cell phenotype as a combination of differences in the location of stable cell states and differences in transition rates between them.


Assuntos
Senescência Celular/fisiologia , Músculo Esquelético/metabolismo , Células-Tronco/metabolismo , Animais , Células Cultivadas , Imuno-Histoquímica , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/citologia , RNA-Seq , Células-Tronco/citologia , Imagem com Lapso de Tempo
2.
Genes Dev ; 28(14): 1578-91, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25030697

RESUMO

Lineage or cell of origin of cancers is often unknown and thus is not a consideration in therapeutic approaches. Alveolar rhabdomyosarcoma (aRMS) is an aggressive childhood cancer for which the cell of origin remains debated. We used conditional genetic mouse models of aRMS to activate the pathognomonic Pax3:Foxo1 fusion oncogene and inactivate p53 in several stages of prenatal and postnatal muscle development. We reveal that lineage of origin significantly influences tumor histomorphology and sensitivity to targeted therapeutics. Furthermore, we uncovered differential transcriptional regulation of the Pax3:Foxo1 locus by tumor lineage of origin, which led us to identify the histone deacetylase inhibitor entinostat as a pharmacological agent for the potential conversion of Pax3:Foxo1-positive aRMS to a state akin to fusion-negative RMS through direct transcriptional suppression of Pax3:Foxo1.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Piridinas/farmacologia , Rabdomiossarcoma Alveolar/patologia , Animais , Linhagem Celular Tumoral , Linhagem da Célula , Modelos Animais de Doenças , Epigênese Genética/efeitos dos fármacos , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/metabolismo , Proteína Supressora de Tumor p53/metabolismo
3.
PLoS Comput Biol ; 14(1): e1005927, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29338005

RESUMO

Cell populations display heterogeneous and dynamic phenotypic states at multiple scales. Similar to molecular features commonly used to explore cell heterogeneity, cell behavior is a rich phenotypic space that may allow for identification of relevant cell states. Inference of cell state from cell behavior across a time course may enable the investigation of dynamics of transitions between heterogeneous cell states, a task difficult to perform with destructive molecular observations. Cell motility is one such easily observed cell behavior with known biomedical relevance. To investigate heterogenous cell states and their dynamics through the lens of cell behavior, we developed Heteromotility, a software tool to extract quantitative motility features from timelapse cell images. In mouse embryonic fibroblasts (MEFs), myoblasts, and muscle stem cells (MuSCs), Heteromotility analysis identifies multiple motility phenotypes within the population. In all three systems, the motility state identity of individual cells is dynamic. Quantification of state transitions reveals that MuSCs undergoing activation transition through progressive motility states toward the myoblast phenotype. Transition rates during MuSC activation suggest non-linear kinetics. By probability flux analysis, we find that this MuSC motility state system breaks detailed balance, while the MEF and myoblast systems do not. Balanced behavior state transitions can be captured by equilibrium formalisms, while unbalanced switching between states violates equilibrium conditions and would require an external driving force. Our data indicate that the system regulating cell behavior can be decomposed into a set of attractor states which depend on the identity of the cell, together with a set of transitions between states. These results support a conceptual view of cell populations as dynamical systems, responding to inputs from signaling pathways and generating outputs in the form of state transitions and observable motile behaviors.


Assuntos
Movimento Celular , Fibroblastos/citologia , Dinâmica não Linear , Algoritmos , Animais , Análise por Conglomerados , Biologia Computacional , Feminino , Fibroblastos/metabolismo , Cinética , Leucócitos Mononucleares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculos/citologia , Fenótipo , Probabilidade , Transdução de Sinais , Células-Tronco/citologia
4.
Nature ; 490(7420): 355-60, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23023126

RESUMO

The niche is a conserved regulator of stem cell quiescence and function. During ageing, stem cell function declines. To what extent and by what means age-related changes within the niche contribute to this phenomenon are unknown. Here we demonstrate that the aged muscle stem cell niche, the muscle fibre, expresses Fgf2 under homeostatic conditions, driving a subset of satellite cells to break quiescence and lose their self-renewing capacity. We show in mice that relatively dormant aged satellite cells robustly express sprouty 1 (Spry1), an inhibitor of fibroblast growth factor (FGF) signalling. Increasing FGF signalling in aged satellite cells under homeostatic conditions by removing Spry1 results in the loss of quiescence, satellite cell depletion and diminished regenerative capacity. Conversely, reducing niche-derived FGF activity through inhibition of Fgfr1 signalling or overexpression of Spry1 in satellite cells prevents their depletion. These experiments identify an age-dependent change in the stem cell niche that directly influences stem cell quiescence and function.


Assuntos
Envelhecimento/fisiologia , Ciclo Celular , Células Musculares/citologia , Células Satélites de Músculo Esquelético/citologia , Nicho de Células-Tronco/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Contagem de Células , Diferenciação Celular , Senescência Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Citometria de Fluxo , Homeostase , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/citologia , Fator de Transcrição PAX7/metabolismo , Fosfoproteínas/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/transplante , Transdução de Sinais , Fatores de Tempo
5.
EMBO J ; 32(16): 2189-90, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23860129

RESUMO

Age-associated changes in tissue maintenance and repair have severe consequences to human physiology. The signals and mechanisms that cause age-related tissue demise are unclear. A recently published study in Cell (Loffredo et al, 2013) proposes that blood-borne factors in the adult systemic environment are lost during ageing, which leads to cardiac hypertrophy. One such factor is GDF11. Exposure of aged mice to youthful systemic factors or GDF11 decreases cardiac hypertrophy of the heart.


Assuntos
Envelhecimento , Proteínas Morfogenéticas Ósseas/metabolismo , Cardiomegalia/metabolismo , Fatores de Diferenciação de Crescimento/metabolismo , Miócitos Cardíacos/metabolismo , Parabiose , Animais , Feminino , Humanos , Masculino
6.
Development ; 141(8): 1649-59, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24715455

RESUMO

Across different niches, subsets of highly functional stem cells are maintained in a relatively dormant rather than proliferative state. Our understanding of proliferative dynamics in tissue-specific stem cells during conditions of increased tissue turnover remains limited. Using a TetO-H2B-GFP reporter of proliferative history, we identify skeletal muscle stem cell, or satellite cells, that retain (LRC) or lose (nonLRC) the H2B-GFP label. We show in mice that LRCs and nonLRCs are formed at birth and persist during postnatal growth and adult muscle repair. Functionally, LRCs and nonLRCs are born equivalent and transition during postnatal maturation into distinct and hierarchically organized subsets. Adult LRCs give rise to LRCs and nonLRCs; the former are able to self-renew, whereas the latter are restricted to differentiation. Expression analysis revealed the CIP/KIP family members p21(cip1) (Cdkn1a) and p27(kip1) (Cdkn1b) to be expressed at higher levels in LRCs. In accordance with a crucial role in LRC fate, loss of p27(kip1) promoted proliferation and differentiation of LRCs in vitro and impaired satellite cell self-renewal after muscle injury. By contrast, loss of p21(cip1) only affected nonLRCs, in which myogenic commitment was inhibited. Our results provide evidence that restriction of self-renewal potential to LRCs is established early in life and is maintained during increased tissue turnover through the cell cycle inhibitor p27(kip1). They also reveal the differential role of CIP/KIP family members at discrete steps within the stem cell hierarchy.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Músculo Esquelético/citologia , Coloração e Rotulagem , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Progressão da Doença , Proteínas de Fluorescência Verde/metabolismo , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Distrofia Muscular Animal/patologia , Fenótipo
7.
8.
Elife ; 112022 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-36583937

RESUMO

The quiescent muscle stem cell (QSC) pool is heterogeneous and generally characterized by the presence and levels of intrinsic myogenic transcription factors. Whether extrinsic factors maintain the diversity of states across the QSC pool remains unknown. The muscle fiber is a multinucleated syncytium that serves as a niche to QSCs, raising the possibility that the muscle fiber regulates the diversity of states across the QSC pool. Here, we show that the muscle fiber maintains a continuum of quiescent states, through a gradient of Notch ligand, Dll4, produced by the fiber and captured by QSCs. The abundance of Dll4 captured by the QSC correlates with the protein levels of the stem cell (SC) identity marker, Pax7. Niche-specific loss of Dll4 decreases QSC diversity and shifts the continuum to cell states that are biased toward more proliferative and committed fates. We reveal that fiber-derived Mindbomb1 (Mib1), an E3 ubiquitin ligase activates Dll4 and controls the heterogeneous levels of Dll4. In response to injury, with a Dll4-replenished niche, the normal continuum and diversity of the SC pool is restored, demonstrating bidirectionality within the SC continuum. Our data show that a post-translational mechanism controls heterogeneity of Notch ligands in a multinucleated niche cell to maintain a continuum of metastable states within the SC pool during tissue homeostasis.


Assuntos
Células Satélites de Músculo Esquelético , Transdução de Sinais , Fibras Musculares Esqueléticas/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Divisão Celular , Células-Tronco/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Nicho de Células-Tronco
9.
Artigo em Inglês | MEDLINE | ID: mdl-31251191

RESUMO

Cells in culture display diverse motility behaviors that may reflect differences in cell state and function, providing motivation to discriminate between different motility behaviors. Current methods to do so rely upon manual feature engineering. However, the types of features necessary to distinguish between motility behaviors can vary greatly depending on the biological context, and it is not always clear which features may be most predictive in each setting for distinguishing particular cell types or disease states. Convolutional neural networks (CNNs) are machine learning models allowing for relevant features to be learned directly from spatial data. Similarly, recurrent neural networks (RNNs) are a class of models capable of learning long term temporal dependencies. Given that cell motility is inherently spacio-temporal data, we present an approach utilizing both convolutional and long- short-term memory (LSTM) recurrent neural network units to analyze cell motility data. These RNN models provide accurate classification of simulated motility and experimentally measured motility from multiple cell types, comparable to results achieved with hand-engineered features. The variety of cell motility differences we can detect suggests that the algorithm is generally applicable to additional cell types not analyzed here. RNN autoencoders based on the same architecture are capable of learning motility features in an unsupervised manner and capturing variation between myogenic cells in the latent space. Adapting these RNN models to motility prediction, RNNs are capable of predicting muscle stem cell motility from past tracking data with performance superior to standard motion prediction models. This advance in cell motility prediction may be of practical utility in cell tracking applications.


Assuntos
Movimento Celular/fisiologia , Biologia Computacional/métodos , Aprendizado Profundo , Animais , Células Cultivadas , Camundongos , Redes Neurais de Computação , Imagem com Lapso de Tempo
10.
Dev Biol ; 335(1): 93-105, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19699733

RESUMO

Muscle stem cells and their progeny play a fundamental role in the regeneration of adult skeletal muscle. We have previously shown that activation of the canonical Wnt/beta-catenin signaling pathway in adult myogenic progenitors is required for their transition from rapidly dividing transient amplifying cells to more differentiated progenitors. Whereas Wnt signaling in Drosophila is dependent on the presence of the co-regulator Legless, previous studies of the mammalian ortholog of Legless, BCL9 (and its homolog, BCL9-2), have not revealed an essential role of these proteins in Wnt signaling in specific tissues during development. Using Cre-lox technology to delete BCL9 and BCL9-2 in the myogenic lineage in vivo and RNAi technology to knockdown the protein levels in vitro, we show that BCL9 is required for activation of the Wnt/beta-catenin cascade in adult mammalian myogenic progenitors. We observed that the nuclear localization of beta-catenin and downstream TCF/LEF-mediated transcription, which are normally observed in myogenic progenitors upon addition of exogenous Wnt and during muscle regeneration, were abrogated when BCL9/9-2 levels were reduced. Furthermore, reductions of BCL9/9-2 inhibited the promotion of myogenic differentiation by Wnt and the normal regenerative response of skeletal muscle. These results suggest a critical role of BCL9/9-2 in the Wnt-mediated regulation of adult, as opposed to embryonic, myogenic progenitors.


Assuntos
Diferenciação Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/fisiologia , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Proteínas Wnt/metabolismo , Animais , Linhagem da Célula , Células Cultivadas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Músculo Esquelético/citologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Interferência de RNA , Células-Tronco/citologia , Fatores de Transcrição , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
11.
Elife ; 92020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32234209

RESUMO

Although heterogeneity is recognized within the murine satellite cell pool, a comprehensive understanding of distinct subpopulations and their functional relevance in human satellite cells is lacking. We used a combination of single cell RNA sequencing and flow cytometry to identify, distinguish, and physically separate novel subpopulations of human PAX7+ satellite cells (Hu-MuSCs) from normal muscles. We found that, although relatively homogeneous compared to activated satellite cells and committed progenitors, the Hu-MuSC pool contains clusters of transcriptionally distinct cells with consistency across human individuals. New surface marker combinations were enriched in transcriptional subclusters, including a subpopulation of Hu-MuSCs marked by CXCR4/CD29/CD56/CAV1 (CAV1+). In vitro, CAV1+ Hu-MuSCs are morphologically distinct, and characterized by resistance to activation compared to CAV1- Hu-MuSCs. In vivo, CAV1+ Hu-MuSCs demonstrated increased engraftment after transplantation. Our findings provide a comprehensive transcriptional view of normal Hu-MuSCs and describe new heterogeneity, enabling separation of functionally distinct human satellite cell subpopulations.


Assuntos
Células Satélites de Músculo Esquelético/fisiologia , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Caveolina 1/análise , Linhagem da Célula , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Fator de Transcrição PAX7/análise , Células Satélites de Músculo Esquelético/química , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/transplante , Adulto Jovem
12.
Cell Stem Cell ; 25(5): 654-665.e4, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31495781

RESUMO

Satellite cells (SCs) reside in a dormant state during tissue homeostasis. The specific paracrine agents and niche cells that maintain SC quiescence remain unknown. We find that Wnt4 produced by the muscle fiber maintains SC quiescence through RhoA. Using cell-specific inducible genetics, we find that a Wnt4-Rho signaling axis constrains SC numbers and activation during tissue homeostasis in adult mice. Wnt4 activates Rho in quiescent SCs to maintain mechanical strain, restrict movement in the niche, and repress YAP. The induction of YAP upon disruption of RhoA is essential for SC activation under homeostasis. In the context of injury, the loss of Wnt4 from the niche accelerates SC activation and muscle repair, whereas overexpression of Wnt4 transitions SCs into a deeper state of quiescence and delays muscle repair. In conclusion, the SC pool undergoes dynamic transitions during early activation with changes in mechano-properties and cytoskeleton signaling preceding cell-cycle entry.


Assuntos
Proliferação de Células/genética , Fibras Musculares Esqueléticas/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Proteína Wnt4/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/fisiologia , Citoesqueleto/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Força Atômica , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Regeneração/genética , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais/genética , Nicho de Células-Tronco/genética , Proteína Wnt4/genética , Proteínas de Sinalização YAP
13.
Cell Stem Cell ; 24(6): 944-957.e5, 2019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-31006621

RESUMO

Stem cell heterogeneity is recognized as functionally relevant for tissue homeostasis and repair. The identity, context dependence, and regulation of skeletal muscle satellite cell (SC) subsets remains poorly understood. We identify a minor subset of Pax7+ SCs that is indelibly marked by an inducible Mx1-Cre transgene in vivo, is enriched for Pax3 expression, and has reduced ROS (reactive oxygen species) levels. Mx1+ SCs possess potent stem cell activity upon transplantation but minimally contribute to endogenous muscle repair, due to their relative low abundance. In contrast, a dramatic clonal expansion of Mx1+ SCs allows extensive contribution to muscle repair and niche repopulation upon selective pressure of radiation stress, consistent with reserve stem cell (RSC) properties. Loss of Pax3 in RSCs increased ROS content and diminished survival and stress tolerance. These observations demonstrate that the Pax7+ SC pool contains a discrete population of radiotolerant RSCs that undergo clonal expansion under severe stress.


Assuntos
Células-Tronco Adultas/fisiologia , Dano ao DNA/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Sobrevivência Celular , Células Clonais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Resistência a Myxovirus/metabolismo , Fator de Transcrição PAX3/metabolismo , Fator de Transcrição PAX7/metabolismo , Radiação Ionizante , Espécies Reativas de Oxigênio/metabolismo , Regeneração , Regulação para Cima
14.
BMC Dev Biol ; 8: 5, 2008 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18215268

RESUMO

BACKGROUND: MyoD is a transcription factor implicated in the regulation of adult muscle gene expression. Distinguishing the expression of MyoD in satellite myoblasts and muscle fibres has proved difficult in vivo leading to controversy over the significance of MyoD expression within adult innervated muscle fibres. Here we employ the MD6.0-lacZ transgenic mouse, in which the 6 kb proximal enhancer/promoter (DRR/PRR) of MyoD drives lacZ, to show that MyoD is present and transcriptionally active in many adult muscle fibres. RESULTS: In culture, MD6.0-lacZ expresses in myotubes but not myogenic cells, unlike endogenous MyoD. Reporter expression in vivo is in muscle fibre nuclei and is reduced in MyoD null mice. The MD6.0-lacZ reporter is down-regulated both in adult muscle fibres by denervation or muscle disuse and in cultured myotubes by inhibition of activity. Activity induces and represses MyoD through the DRR and PRR, respectively. During the postnatal period, accumulation of beta-galactosidase correlates with maturation of innervation. Strikingly, endogenous MyoD expression is up-regulated in fibres by complete denervation, arguing for a separate activity-dependent suppression of MyoD requiring regulatory elements outside the DRR/PRR. CONCLUSION: The data show that MyoD regulation is more complex than previously supposed. Two factors, MyoD protein itself and fibre activity are required for essentially all expression of the 6 kb proximal enhancer/promoter (DRR/PRR) of MyoD in adult fibres. We propose that modulation of MyoD positive feedback by electrical activity determines the set point of MyoD expression in innervated fibres through the DRR/PRR element.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento , Fibras Musculares Esqueléticas/metabolismo , Proteína MyoD/genética , Regiões Promotoras Genéticas/genética , Animais , Células Cultivadas , Regulação para Baixo , Estimulação Elétrica , Óperon Lac , Camundongos , Camundongos Transgênicos , Denervação Muscular
15.
Curr Top Dev Biol ; 126: 299-322, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29305003

RESUMO

Satellite cells (SCs) are a population of muscle-resident stem cells that are essential for efficient tissue repair. SCs reside in a relatively quiescent state during normal tissue turnover, but are activated in response to injury through the microenvironment and cell-intrinsic signals. During aging, SC dysfunction is a major contributor to the decline in regenerative potential of muscle tissue. Recent studies have demonstrated that both cell-intrinsic and cell-extrinsic factors are deregulated during aging. Interventions that reverse age-associated changes in SCs or the niche have shown to partially rejuvenate the regenerative capacity of aged muscle SCs. In this review, we discuss recent advances in SC biology as it pertains to the deleterious effects of aging. A better understanding of how age-dependent changes in the SC and its environment niche impact muscle regeneration could lead to interventions to ameliorate the effects of aging in humans.


Assuntos
Envelhecimento/fisiologia , Músculo Esquelético/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Autofagia/fisiologia , Proliferação de Células/fisiologia , Humanos , Modelos Biológicos , Músculo Esquelético/citologia , Regeneração/fisiologia
16.
Stem Cell Rev ; 3(3): 226-37, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17917136

RESUMO

The myogenic stem cell (satellite cell) is almost solely responsible for the remarkable regeneration of adult skeletal muscle fibers after injury. The availability and the functionality of satellite cells are the determinants of efficient muscle regeneration. During aging, the efficiency of muscle regeneration declines, suggesting that the functionality of satellite cells and their progeny may be altered. Satellite cells do not sit in isolation but rather are surrounded by, and influenced by, many extrinsic factors within the muscle tissue that can alter their functionality. These factors likely change during aging and impart both reversible and irreversible changes to the satellite cells and on their proliferating progeny. In this review, we discuss the possible mechanisms of impaired muscle regeneration with respect to the biology of satellite cells. Future studies that enhance our understanding of the interactions between stem cells and the environment in which they reside will offer promise for therapeutic applications in age-related diseases.


Assuntos
Senescência Celular , Músculo Esquelético/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Humanos , Modelos Biológicos
17.
Cell Stem Cell ; 18(1): 5-7, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26748748

RESUMO

Quiescence is highly regulated to preserve stem cell function. In this issue of Cell Stem Cell, Zismanov et al. (2016) show that P-eIF2α, a translational initiation factor, reinforces the quiescent state of muscle stem cells by safeguarding against cell-cycle entry and lineage progression.


Assuntos
Diferenciação Celular , Células Satélites de Músculo Esquelético , Ciclo Celular , Humanos , Células-Tronco/citologia
18.
Skelet Muscle ; 6: 1, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26783424

RESUMO

Skeletal muscle has a remarkable capacity to regenerate by virtue of its resident stem cells (satellite cells). This capacity declines with aging, although whether this is due to extrinsic changes in the environment and/or to cell-intrinsic mechanisms associated to aging has been a matter of intense debate. Furthermore, while some groups support that satellite cell aging is reversible by a youthful environment, others support cell-autonomous irreversible changes, even in the presence of youthful factors. Indeed, whereas the parabiosis paradigm has unveiled the environment as responsible for the satellite cell functional decline, satellite cell transplantation studies support cell-intrinsic deficits with aging. In this review, we try to shed light on the potential causes underlying these discrepancies. We propose that the experimental paradigm used to interrogate intrinsic and extrinsic regulation of stem cell function may be a part of the problem. The assays deployed are not equivalent and may overburden specific cellular regulatory processes and thus probe different aspects of satellite cell properties. Finally, distinct subsets of satellite cells may be under different modes of molecular control and mobilized preferentially in one paradigm than in the other. A better understanding of how satellite cells molecularly adapt during aging and their context-dependent deployment during injury and transplantation will lead to the development of efficacious compensating strategies that maintain stem cell fitness and tissue homeostasis throughout life.


Assuntos
Senescência Celular , Músculo Esquelético/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Humanos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Fenótipo , Células Satélites de Músculo Esquelético/metabolismo , Transdução de Sinais , Nicho de Células-Tronco
19.
Stem Cell Reports ; 4(6): 1061-74, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-25960061

RESUMO

To ensure accurate genomic segregation, cells evolved the spindle assembly checkpoint (SAC), whose role in adult stem cells remains unknown. Inducible perturbation of a SAC kinase, Mps1, and its downstream effector, Mad2, in skeletal muscle stem cells shows the SAC to be critical for normal muscle growth, repair, and self-renewal of the stem cell pool. SAC-deficient muscle stem cells arrest in G1 phase of the cell cycle with elevated aneuploidy, resisting differentiation even under inductive conditions. p21(CIP1) is responsible for these SAC-deficient phenotypes. Despite aneuploidy's correlation with aging, we find that aged proliferating muscle stem cells display robust SAC activity without elevated aneuploidy. Thus, muscle stem cells have a two-step mechanism to safeguard their genomic integrity. The SAC prevents chromosome missegregation and, if it fails, p21(CIP1)-dependent G1 arrest limits cellular propagation and tissue integration. These mechanisms ensure that muscle stem cells with compromised genomes do not contribute to tissue homeostasis.


Assuntos
Células Satélites de Músculo Esquelético/metabolismo , Fuso Acromático/metabolismo , Aneuploidia , Animais , Diferenciação Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular , Hibridização in Situ Fluorescente , Cinetocoros/química , Cinetocoros/metabolismo , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Mitose/efeitos dos fármacos , Músculo Esquelético/fisiologia , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Nocodazol/farmacologia , Fenótipo , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Regeneração , Células Satélites de Músculo Esquelético/citologia
20.
Cell Metab ; 22(1): 164-74, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26001423

RESUMO

Age-related frailty may be due to decreased skeletal muscle regeneration. The role of TGF-ß molecules myostatin and GDF11 in regeneration is unclear. Recent studies showed an age-related decrease in GDF11 and that GDF11 treatment improves muscle regeneration, which were contrary to prior studies. We now show that these recent claims are not reproducible and the reagents previously used to detect GDF11 are not GDF11 specific. We develop a GDF11-specific immunoassay and show a trend toward increased GDF11 levels in sera of aged rats and humans. GDF11 mRNA increases in rat muscle with age. Mechanistically, GDF11 and myostatin both induce SMAD2/3 phosphorylation, inhibit myoblast differentiation, and regulate identical downstream signaling. GDF11 significantly inhibited muscle regeneration and decreased satellite cell expansion in mice. Given early data in humans showing a trend for an age-related increase, GDF11 could be a target for pharmacologic blockade to treat age-related sarcopenia.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Diferenciação de Crescimento/metabolismo , Músculo Esquelético/fisiologia , Regeneração , Envelhecimento , Animais , Proteínas Morfogenéticas Ósseas/sangue , Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular , Linhagem Celular , Fatores de Diferenciação de Crescimento/sangue , Fatores de Diferenciação de Crescimento/genética , Humanos , Camundongos , Mioblastos/citologia , Mioblastos/metabolismo , Miostatina/metabolismo , Ratos , Transdução de Sinais , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA