Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38848717

RESUMEN

The histone H3 lysine 9 methyltransferase SETDB1 controls transcriptional repression to direct stem cell fate. Here, we show that Setdb1 expression by adult muscle stem cells (MuSCs) is required for skeletal muscle regeneration. We find that SETDB1 represses the expression of endogenous retroviruses (ERVs) in MuSCs. ERV de-repression in Setdb1-null MuSCs prevents their amplification following exit from quiescence and promotes cell death. Multi-omics profiling shows that chromatin decompaction at ERV loci activates the DNA-sensing cGAS-STING pathway, entailing cytokine expression by Setdb1-null MuSCs. This is followed by aberrant infiltration of inflammatory cells, including pathological macrophages. The ensuing histiocytosis is accompanied by myofiber necrosis, which, in addition to progressive MuSCs depletion, completely abolishes tissue repair. In contrast, loss of Setdb1 in fibro-adipogenic progenitors (FAPs) does not impact immune cells. In conclusion, genome maintenance by SETDB1 in an adult somatic stem cell is necessary for both its regenerative potential and adequate reparative inflammation.

3.
Stem Cell Res Ther ; 14(1): 246, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37697344

RESUMEN

BACKGROUND: Inflammatory memory or trained immunity is a recently described process in immune and non-immune tissue resident cells, whereby previous exposure to inflammation mediators leads to a faster and stronger responses upon secondary challenge. Whether previous muscle injury is associated with altered responses to subsequent injury by satellite cells (SCs), the muscle stem cells, is not known. METHODS: We used a mouse model of repeated muscle injury, in which intramuscular cardiotoxin (CTX) injections were administered 50 days apart in order to allow for full recovery of the injured muscle before the second injury. The effect of prior injury on the phenotype, proliferation and regenerative potential of satellite cells following a second injury was examined in vitro and in vivo by immunohistochemistry, RT-qPCR and histological analysis. RESULTS: We show that SCs isolated from muscle at 50 days post-injury (injury-experienced SCs (ieSCs)) enter the cell cycle faster and form bigger myotubes when cultured in vitro, compared to control SCs isolated from uninjured contralateral muscle. Injury-experienced SCs were characterized by the activation of the mTORC 1 signaling pathway, suggesting they are poised to activate sooner following a second injury. Consequently, upon second injury, SCs accumulate in greater numbers in muscle at 3 and 10 days after injury. These changes in SC phenotype and behavior were associated with accelerated muscle regeneration, as evidenced by an earlier appearance of bigger fibers and increased number of myonuclei per fiber at day 10 after the second injury. CONCLUSIONS: Overall, we show that skeletal muscle injury has a lasting effect on SC function priming them to respond faster to a subsequent injury. The ieSCs have long-term enhanced regenerative properties that contribute to accelerated regeneration following a secondary challenge.


Asunto(s)
Lesiones de Repetición , Animales , Ratones , Fibras Musculares Esqueléticas , Músculo Esquelético , Ciclo Celular , División Celular
5.
Cell Rep ; 41(12): 111861, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36543136

RESUMEN

Striated muscle is a highly organized structure composed of well-defined anatomical domains with integrated but distinct assignments. So far, the lack of a direct correlation between tissue architecture and gene expression has limited our understanding of how each unit responds to physio-pathologic contexts. Here, we show how the combined use of spatially resolved transcriptomics and immunofluorescence can bridge this gap by enabling the unbiased identification of such domains and the characterization of their response to external perturbations. Using a spatiotemporal analysis, we follow changes in the transcriptome of specific domains in muscle in a model of denervation. Furthermore, our approach enables us to identify the spatial distribution and nerve dependence of atrophic signaling pathway and polyamine metabolism to glycolytic fibers. Indeed, we demonstrate that perturbations of polyamine pathway can affect muscle function. Our dataset serves as a resource for future studies of the mechanisms underlying skeletal muscle homeostasis and innervation.


Asunto(s)
Atrofia Muscular , Transcriptoma , Humanos , Atrofia Muscular/metabolismo , Transcriptoma/genética , Músculo Esquelético/metabolismo , Perfilación de la Expresión Génica , Poliaminas/metabolismo
6.
J Cachexia Sarcopenia Muscle ; 13(3): 1771-1784, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35319169

RESUMEN

BACKGROUND: Fibrosis is defined as an excessive accumulation of extracellular matrix (ECM) components. Many organs are subjected to fibrosis including the lung, liver, heart, skin, kidney, and muscle. Muscle fibrosis occurs in response to trauma, aging, or dystrophies and impairs muscle function. Fibrosis represents a hurdle for the treatment of human muscular dystrophies. While data on the mechanisms of fibrosis have mostly been investigated in mice, dystrophic mouse models often do not recapitulate fibrosis as observed in human patients. Consequently, the cellular and molecular mechanisms that lead to fibrosis in human muscle still need to be identified. METHODS: Combining mass cytometry, transcriptome profiling, in vitro co-culture experiments, and in vivo transplantation in immunodeficient mice, we investigated the role and nature of nonmyogenic cells (fibroadipogenic progenitors, FAPs) from human fibrotic muscles of healthy individuals (FibMCT ) and individuals with oculopharyngeal muscular dystrophy (OPMD; FibMOP ), as compared with nonmyogenic cells from human nonfibrotic muscle (MCT ). RESULTS: We found that the proliferation rate of FAPs from fibrotic muscle is 3-4 times higher than those of FAPs from nonfibrotic muscle (population doubling per day: MCT 0.2 ± 0.1, FibMCT 0.7 ± 0.1, and FibMOP 0.8 ± 0.3). When cocultured with muscle cells, FAPs from fibrotic muscle impair the fusion index unlike MCT FAPs (myoblasts alone 57.3 ± 11.1%, coculture with MCT 43.1 ± 8.9%, with FibMCT 31.7 ± 8.2%, and with FibMOP 36.06 ± 10.29%). We also observed an increased proliferation of FAPs from fibrotic muscles in these co-cultures in differentiation conditions (FibMCT +17.4%, P < 0.01 and FibMOP +15.1%, P < 0.01). This effect is likely linked to the increased activation of the canonical TGFß-SMAD pathway in FAPs from fibrotic muscles evidenced by pSMAD3 immunostaining (P < 0.05). In addition to the profibrogenic TGFß pathway, we identified endothelin as a new actor implicated in the altered cross-talk between muscle cells and fibrotic FAPs, confirmed by an improvement of the fusion index in the presence of bosentan, an endothelin receptor antagonist (from 33.8 ± 10.9% to 52.9 ± 10.1%, P < 0.05). CONCLUSIONS: Our data demonstrate the key role of FAPs and their cross-talk with muscle cells through a paracrine signalling pathway in fibrosis of human skeletal muscle and identify endothelin as a new druggable target to counteract human muscle fibrosis.


Asunto(s)
Adipogénesis , Distrofia Muscular Oculofaríngea , Animales , Endotelinas/metabolismo , Retroalimentación , Fibrosis , Humanos , Ratones , Fibras Musculares Esqueléticas , Músculo Esquelético/patología , Distrofia Muscular Oculofaríngea/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
7.
Aging (Albany NY) ; 14(1): 28-53, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-35023852

RESUMEN

Aging-associated muscle wasting and impaired regeneration are caused by deficiencies in muscle stem cell (MuSC) number and function. We postulated that aged MuSCs are intrinsically impaired in their responsiveness to omnipresent mechanical cues through alterations in MuSC morphology, mechanical properties, and number of integrins, culminating in impaired proliferative capacity. Here we show that aged MuSCs exhibited significantly lower growth rate and reduced integrin-α7 expression as well as lower number of phospho-paxillin clusters than young MuSCs. Moreover, aged MuSCs were less firmly attached to matrigel-coated glass substrates compared to young MuSCs, as 43% of the cells detached in response to pulsating fluid shear stress (1 Pa). YAP nuclear localization was 59% higher than in young MuSCs, yet YAP target genes Cyr61 and Ctgf were substantially downregulated. When subjected to pulsating fluid shear stress, aged MuSCs exhibited reduced upregulation of proliferation-related genes. Together these results indicate that aged MuSCs exhibit impaired mechanosensitivity and growth potential, accompanied by altered morphology and mechanical properties as well as reduced integrin-α7 expression. Aging-associated impaired muscle regenerative capacity and muscle wasting is likely due to aging-induced intrinsic MuSC alterations and dysfunctional mechanosensitivity.


Asunto(s)
Proliferación Celular/fisiología , Senescencia Celular/fisiología , Mecanotransducción Celular/fisiología , Fibras Musculares Esqueléticas/fisiología , Células Madre/fisiología , Envejecimiento , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Adhesión Celular/fisiología , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/metabolismo , Ratones , Óxido Nítrico/metabolismo , Resistencia al Corte
8.
Biophys J ; 120(13): 2665-2678, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34087215

RESUMEN

Muscle stem cells (MuSCs) are requisite for skeletal muscle regeneration and homeostasis. Proper functioning of MuSCs, including activation, proliferation, and fate decision, is determined by an orchestrated series of events and communication between MuSCs and their niche. A multitude of biochemical stimuli are known to regulate MuSC fate and function. However, in addition to biochemical factors, it is conceivable that MuSCs are subjected to mechanical forces during muscle stretch-shortening cycles because of myofascial connections between MuSCs and myofibers. MuSCs respond to mechanical forces in vitro, but it remains to be proven whether physical forces are also exerted on MuSCs in their native niche and whether they contribute to the functioning and fate of MuSCs. MuSC deformation in their native niche resulting from mechanical loading of ex vivo myofiber bundles was visualized utilizing mT/mG double-fluorescent Cre-reporter mouse and multiphoton microscopy. MuSCs were subjected to 1 h pulsating fluid shear stress (PFSS) with a peak shear stress rate of 6.5 Pa/s. After PFSS treatment, nitric oxide, messenger RNA (mRNA) expression levels of genes involved in regulation of MuSC proliferation and differentiation, ERK 1/2, p38, and AKT activation were determined. Ex vivo stretching of extensor digitorum longus and soleus myofiber bundles caused compression as well as tensile and shear deformation of MuSCs in their niche. MuSCs responded to PFSS in vitro with increased nitric oxide production and an upward trend in iNOS mRNA levels. PFSS enhanced gene expression of c-Fos, Cdk4, and IL-6, whereas expression of Wnt1, MyoD, Myog, Wnt5a, COX2, Rspo1, Vangl2, Wnt10b, and MGF remained unchanged. ERK 1/2 and p38 MAPK signaling were also upregulated after PFSS treatment. We conclude that MuSCs in their native niche are subjected to force-induced deformations due to myofiber stretch-shortening. Moreover, MuSCs are mechanoresponsive, as evidenced by PFSS-mediated expression of factors by MuSCs known to promote proliferation.


Asunto(s)
Músculo Esquelético , Mioblastos , Animales , Diferenciación Celular , Expresión Génica , Ratones , Estrés Mecánico
9.
JCI Insight ; 6(7)2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33661767

RESUMEN

Here, we report on the identification of Itga7-expressing muscle-resident glial cells activated by loss of neuromuscular junction (NMJ) integrity. Gene expression analysis at the bulk and single-cell level revealed that these cells are distinct from Itga7-expressing muscle satellite cells. We show that a selective activation and expansion of Itga7+ glial cells occur in response to muscle nerve lesion. Upon activation, muscle glial-derived progenies expressed neurotrophic genes, including nerve growth factor receptor, which enables their isolation by FACS. We show that activated muscle glial cells also expressed genes potentially implicated in extracellular matrix remodeling at NMJs. We found that tenascin C, which was highly expressed by muscle glial cells, activated upon nerve injury and preferentially localized to NMJ. Interestingly, we observed that the activation of muscle glial cells by acute nerve injury was reversible upon NMJ repair. By contrast, in a mouse model of ALS, in which NMJ degeneration is progressive, muscle glial cells steadily increased over the course of the disease. However, they exhibited an impaired neurotrophic activity, suggesting that pathogenic activation of glial cells may be implicated in ALS progression.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Músculo Esquelético/citología , Neuroglía/fisiología , Traumatismos de la Médula Espinal/patología , Animales , Antígenos CD/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Cadenas alfa de Integrinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Neuroglía/citología , Unión Neuromuscular/citología , Receptor de Factor de Crecimiento Nervioso/genética , Receptores Colinérgicos/metabolismo , Nervio Ciático/lesiones , Análisis de la Célula Individual , Superóxido Dismutasa-1/genética
10.
Nat Commun ; 12(1): 750, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531466

RESUMEN

Muscle cell fusion is a multistep process involving cell migration, adhesion, membrane remodeling and actin-nucleation pathways to generate multinucleated myotubes. However, molecular brakes restraining cell-cell fusion events have remained elusive. Here we show that transforming growth factor beta (TGFß) pathway is active in adult muscle cells throughout fusion. We find TGFß signaling reduces cell fusion, regardless of the cells' ability to move and establish cell-cell contacts. In contrast, inhibition of TGFß signaling enhances cell fusion and promotes branching between myotubes in mouse and human. Exogenous addition of TGFß protein in vivo during muscle regeneration results in a loss of muscle function while inhibition of TGFßR2 induces the formation of giant myofibers. Transcriptome analyses and functional assays reveal that TGFß controls the expression of actin-related genes to reduce cell spreading. TGFß signaling is therefore requisite to limit mammalian myoblast fusion, determining myonuclei numbers and myofiber size.


Asunto(s)
Músculo Esquelético/citología , Factor de Crecimiento Transformador beta/metabolismo , Adolescente , Adulto , Animales , Western Blotting , Fusión Celular , Células Cultivadas , Biología Computacional , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Regeneración/genética , Regeneración/fisiología , Células Madre/citología , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/genética , Adulto Joven
11.
Mol Cell ; 74(3): 609-621.e6, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30922843

RESUMEN

Adult tissue repair and regeneration require stem-progenitor cells that can self-renew and generate differentiated progeny. Skeletal muscle regenerative capacity relies on muscle satellite cells (MuSCs) and their interplay with different cell types within the niche. However, our understanding of skeletal muscle tissue cellular composition is limited. Here, using a combined approach of single-cell RNA sequencing and mass cytometry, we precisely mapped 10 different mononuclear cell types in adult mouse muscle. We also characterized gene signatures and determined key discriminating markers of each cell type. We identified two previously understudied cell populations in the interstitial compartment. One expresses the transcription factor scleraxis and generated tenocytes in vitro. The second expresses markers of smooth muscle and mesenchymal cells (SMMCs) and, while distinct from MuSCs, exhibited myogenic potential and promoted MuSC engraftment following transplantation. The blueprint presented here yields crucial insights into muscle-resident cell-type identities and can be exploited to study muscle diseases.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Fibras Musculares Esqueléticas/citología , Células Satélite del Músculo Esquelético/citología , Animales , Ratones , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Análisis de la Célula Individual , Células Madre/citología , Células Madre/metabolismo
12.
Nat Commun ; 9(1): 3670, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30202063

RESUMEN

Fibro-adipogenic progenitors (FAPs) are currently defined by their anatomical position, expression of non-specific membrane-associated proteins, and ability to adopt multiple lineages in vitro. Gene expression analysis at single-cell level reveals that FAPs undergo dynamic transitions through a spectrum of cell states that can be identified by differential expression levels of Tie2 and Vcam1. Different patterns of Vcam1-negative Tie2high or Tie2low and Tie2low/Vcam1-expressing FAPs are detected during neonatal myogenesis, response to acute injury and Duchenne Muscular Dystrophy (DMD). RNA sequencing analysis identified cell state-specific transcriptional profiles that predict functional interactions with satellite and inflammatory cells. In particular, Vcam1-expressing FAPs, which exhibit a pro-fibrotic expression profile, are transiently activated by acute injury in concomitance with the inflammatory response. Aberrant persistence of Vcam1-expressing FAPs is detected in DMD muscles or upon macrophage depletion, and is associated with muscle fibrosis, thereby revealing how disruption of inflammation-regulated FAPs dynamics leads to a pathogenic outcome.


Asunto(s)
Adipogénesis/fisiología , Desarrollo de Músculos/fisiología , Distrofia Muscular de Duchenne/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular , Citometría de Flujo , Perfilación de la Expresión Génica , Inflamación , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Endogámicos mdx , Músculo Esquelético/fisiología , Receptor TIE-2/metabolismo , Regeneración , Análisis de Secuencia de ARN , Molécula 1 de Adhesión Celular Vascular/metabolismo
13.
Nat Cell Biol ; 20(8): 917-927, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30050118

RESUMEN

Fibro-adipogenic progenitors (FAPs) are typically activated in response to muscle injury, and establish functional interactions with inflammatory and muscle stem cells (MuSCs) to promote muscle repair. We found that denervation causes progressive accumulation of FAPs, without concomitant infiltration of macrophages and MuSC-mediated regeneration. Denervation-activated FAPs exhibited persistent STAT3 activation and secreted elevated levels of IL-6, which promoted muscle atrophy and fibrosis. FAPs with aberrant activation of STAT3-IL-6 signalling were also found in mouse models of spinal cord injury, spinal muscular atrophy, amyotrophic lateral sclerosis (ALS) and in muscles of ALS patients. Inactivation of STAT3-IL-6 signalling in FAPs effectively countered muscle atrophy and fibrosis in mouse models of acute denervation and ALS (SODG93A mice). Activation of pathogenic FAPs following loss of integrity of neuromuscular junctions further illustrates the functional versatility of FAPs in response to homeostatic perturbations and suggests their potential contribution to the pathogenesis of neuromuscular diseases.


Asunto(s)
Adipogénesis , Esclerosis Amiotrófica Lateral/metabolismo , Desnervación/métodos , Interleucina-6/metabolismo , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular/metabolismo , Mioblastos Esqueléticos/metabolismo , Músculo Cuádriceps/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Traumatismos de la Médula Espinal/metabolismo , Adipogénesis/efectos de los fármacos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/prevención & control , Animales , Cardiotoxinas , Línea Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Fibrosis , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Atrofia Muscular/genética , Atrofia Muscular/patología , Atrofia Muscular/prevención & control , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/prevención & control , Mutación , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/patología , Fármacos Neuromusculares/farmacología , Músculo Cuádriceps/efectos de los fármacos , Músculo Cuádriceps/inervación , Músculo Cuádriceps/patología , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Nervio Ciático/cirugía , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/prevención & control , Superóxido Dismutasa-1/genética
14.
Curr Top Dev Biol ; 126: 177-203, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29304998

RESUMEN

Adult skeletal muscle is endowed with regenerative potential through partially recapitulating the embryonic developmental program. Upon acute injury or in pathological conditions, quiescent muscle-resident stem cells, called satellite cells, become activated and give rise to myogenic progenitors that massively proliferate, differentiate, and fuse to form new myofibers and restore tissue functionality. In addition, a proportion of activated cells returns back to quiescence and replenish the pool of satellite cells in order to maintain the ability of skeletal muscle tissue to repair. Self-renewal is the process by which stem cells divide to make more stem cells to maintain the stem cell population throughout life. This process is controlled by cell-intrinsic transcription factors regulated by cell-extrinsic signals from the niche and the microenvironment. This chapter provides an overview about the general aspects of satellite cell biology and focuses on the cellular and molecular aspects of satellite cell self-renewal. To date, we are still far from understanding how a very small proportion of the satellite cell progeny maintain their stem cell identity when most of their siblings progress through the myogenic program to construct myofibers.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Autorrenovación de las Células/fisiología , Células Satélite del Músculo Esquelético/fisiología , Animales , Humanos , Desarrollo de Músculos/fisiología , Músculo Esquelético/fisiología , Músculo Esquelético/fisiopatología , Regeneración/fisiología , Cicatrización de Heridas/fisiología
15.
Cell Rep ; 18(10): 2320-2330, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28273449

RESUMEN

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.


Asunto(s)
Mioblastos/citología , Mioblastos/metabolismo , Trombospondinas/metabolismo , Vía de Señalización Wnt , Animales , Diferenciación Celular , Fusión Celular , Proliferación Celular , Células Cultivadas , Ratones Endogámicos C57BL , Desarrollo de Músculos , Factor de Transcripción PAX7/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , beta Catenina/metabolismo
16.
Cell Rep ; 15(6): 1277-90, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27134174

RESUMEN

Skeletal muscle regeneration relies on a pool of resident muscle stem cells called satellite cells (MuSCs). Following injury-induced destruction of the myofibers, quiescent MuSCs are activated and generate transient amplifying progenitors (myoblasts) that will fuse to form new myofibers. Here, we focus on the canonical Wnt signaling pathway and find that either conditional ß-catenin disruption or activation in adult MuSCs results in perturbation of muscle regeneration. Using both in vivo and in vitro approaches, we observed that myoblasts lacking ß-catenin show delayed differentiation, whereas myoblasts with constitutively active ß-catenin undergo precocious growth arrest and differentiation. Transcriptome analysis further demonstrated that Wnt/ß-catenin signaling interacts with multiple pathways and, more specifically, TGF-ß signaling. Indeed, exogenous TGF-ß2 stimulation restores the regenerative potential of muscles with targeted ß-catenin disruption in MuSCs. We conclude that a precise level of ß-catenin activity is essential for regulating the amplification and differentiation of MuSC descendants during adult myogenesis.


Asunto(s)
Músculos/citología , Células Madre/citología , Cicatrización de Heridas , beta Catenina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Eliminación de Gen , Marcación de Gen , Ratones Noqueados , Desarrollo de Músculos , Mioblastos/citología , Regeneración , Transducción de Señal , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
17.
J Cell Biol ; 210(5): 717-26, 2015 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-26304725

RESUMEN

The tumor suppressor adenomatous polyposis coli (APC) is a crucial regulator of many stem cell types. In constantly cycling stem cells of fast turnover tissues, APC loss results in the constitutive activation of a Wnt target gene program that massively increases proliferation and leads to malignant transformation. However, APC function in skeletal muscle, a tissue with a low turnover rate, has never been investigated. Here we show that conditional genetic disruption of APC in adult muscle stem cells results in the abrogation of adult muscle regenerative potential. We demonstrate that APC removal in adult muscle stem cells abolishes cell cycle entry and leads to cell death. By using double knockout strategies, we further prove that this phenotype is attributable to overactivation of ß-catenin signaling. Our results demonstrate that in muscle stem cells, APC dampens canonical Wnt signaling to allow cell cycle progression and radically diverge from previous observations concerning stem cells in actively self-renewing tissues.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Células Madre Adultas/fisiología , Apoptosis/genética , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/fisiología , Proteína de la Poliposis Adenomatosa del Colon/genética , Células Madre Adultas/citología , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Supervivencia Celular/genética , Femenino , Masculino , Ratones , Ratones Transgénicos , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Interferencia de ARN , ARN Interferente Pequeño/genética , Regeneración/genética , Células Satélite del Músculo Esquelético/citología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , Cicatrización de Heridas/genética , beta Catenina/metabolismo
18.
Genes Dev ; 28(8): 841-57, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24682306

RESUMEN

Fibro-adipogenic progenitors (FAPs) are important components of the skeletal muscle regenerative environment. Whether FAPs support muscle regeneration or promote fibro-adipogenic degeneration is emerging as a key determinant in the pathogenesis of muscular diseases, including Duchenne muscular dystrophy (DMD). However, the molecular mechanism that controls FAP lineage commitment and activity is currently unknown. We show here that an HDAC-myomiR-BAF60 variant network regulates the fate of FAPs in dystrophic muscles of mdx mice. Combinatorial analysis of gene expression microarray, genome-wide chromatin remodeling by nuclease accessibility (NA) combined with next-generation sequencing (NA-seq), small RNA sequencing (RNA-seq), and microRNA (miR) high-throughput screening (HTS) against SWI/SNF BAF60 variants revealed that HDAC inhibitors (HDACis) derepress a "latent" myogenic program in FAPs from dystrophic muscles at early stages of disease. Specifically, HDAC inhibition induces two core components of the myogenic transcriptional machinery, MYOD and BAF60C, and up-regulates the myogenic miRs (myomiRs) (miR-1.2, miR-133, and miR-206), which target the alternative BAF60 variants BAF60A and BAF60B, ultimately directing promyogenic differentiation while suppressing the fibro-adipogenic phenotype. In contrast, FAPs from late stage dystrophic muscles are resistant to HDACi-induced chromatin remodeling at myogenic loci and fail to activate the promyogenic phenotype. These results reveal a previously unappreciated disease stage-specific bipotency of mesenchimal cells within the regenerative environment of dystrophic muscles. Resolution of such bipotency by epigenetic intervention with HDACis provides a molecular rationale for the in situ reprogramming of target cells to promote therapeutic regeneration of dystrophic muscles.


Asunto(s)
Histona Desacetilasas/metabolismo , MicroARNs/metabolismo , Músculo Esquelético/fisiología , Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Células Madre/metabolismo , Animales , Reprogramación Celular/genética , Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Endogámicos mdx , Proteínas Musculares/genética , Proteínas Musculares/metabolismo
19.
Genes Dev ; 27(21): 2332-44, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24186978

RESUMEN

A critical but molecularly uncharacterized step in heart formation and regeneration is the process that commits progenitor cells to differentiate into cardiomyocytes. Here, we show that the endoderm-derived dual Nodal/bone morphogenetic protein (BMP) antagonist Cerberus-1 (Cer1) in embryonic stem cell cultures orchestrates two signaling pathways that direct the SWI/SNF chromatin remodeling complex to cardiomyogenic loci in multipotent (KDR/Flk1+) progenitors, activating lineage-specific transcription. Transient inhibition of Nodal by Cer1 induces Brahma-associated factor 60c (Baf60c), one of three Baf60 variants (a, b, and c) that are mutually exclusively assembled into SWI/SNF. Blocking Nodal and BMP also induces lineage-specific transcription factors Gata4 and Tbx5, which interact with Baf60c. siRNA to Cer1, Baf60c, or the catalytic SWI/SNF subunit Brg1 prevented the developmental opening of chromatin surrounding the Nkx2.5 early cardiac enhancer and cardiomyocyte differentiation. Overexpression of Baf60c fully rescued these deficits, positioning Baf60c and SWI/SNF function downstream from Cer1. Thus, antagonism of Nodal and BMP coordinates induction of the myogenic Baf60c variant and interacting transcription factors to program the developmental opening of cardiomyocyte-specific loci in chromatin. This is the first demonstration that cues from the progenitor cell environment direct the subunit variant composition of SWI/SNF to remodel the transcriptional landscape for lineage-specific differentiation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Miocitos Cardíacos/citología , Proteína Nodal/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Células Cultivadas , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona , Citocinas/genética , Citocinas/metabolismo , Endodermo/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Proteína Nodal/genética , ARN Interferente Pequeño/metabolismo , Células Madre/citología , Células Madre/metabolismo
20.
FEBS J ; 280(17): 4014-25, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23745685

RESUMEN

During embryonic development, pluripotent cells are genetically committed to specific lineages by the expression of cell-type-specific transcriptional activators that direct the formation of specialized tissues and organs in response to developmental cues. Chromatin-modifying proteins are emerging as essential components of the epigenetic machinery, which establishes the nuclear landscape that ultimately determines the final identity and functional specialization of adult cells. Recent evidence has revealed that discrete populations of adult cells can retain the ability to adopt alternative cell fates in response to environmental cues. These cells include conventional adult stem cells and a still poorly defined collection of cell types endowed with facultative phenotype and functional plasticity. Under physiological conditions or adaptive states, these cells cooperate to support tissue and organ homeostasis, and to promote growth or compensatory regeneration. However, during chronic diseases and aging these cells can adopt a pathological phenotype and mediate maladaptive responses, such as the formation of fibrotic scars and fat deposition that progressively replaces structural and functional units of tissues and organs. The molecular determinants of these phenotypic transitions are only emerging from recent studies that reveal how dynamic chromatin states can generate flexible epigenetic landscapes, which confer on cells the ability to retain partial pluripotency and adapt to environmental changes. This review summarizes our current knowledge on the role of the epigenetic machinery as a 'filter' between genetic commitment and environmental signals in cell types that can alternatively promote skeletal muscle regeneration or fibro-adipogenic degeneration.


Asunto(s)
Diferenciación Celular , Exposición a Riesgos Ambientales , Epigenómica , Músculo Esquelético/citología , Regeneración/fisiología , Animales , Humanos , Músculo Esquelético/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...