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












Base de datos
Intervalo de año de publicación
1.
Cell Physiol Biochem ; 54(3): 333-353, 2020 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-32275813

RESUMEN

BACKGROUND/AIMS: Cell migration and extracellular matrix remodeling underlie normal mammalian development and growth as well as pathologic tumor invasion. Skeletal muscle is no exception, where satellite cell migration replenishes nuclear content in damaged tissue and extracellular matrix reforms during regeneration. A key set of enzymes that regulate these processes are matrix metalloproteinases (MMP)s. The collagenase MMP-13 is transiently upregulated during muscle regeneration, but its contribution to damage resolution is unknown. The purpose of this work was to examine the importance of MMP-13 in muscle regeneration and growth in vivo and to delineate a satellite cell specific role for this collagenase. METHODS: Mice with total and satellite cell specific Mmp13 deletion were utilized to determine the importance of MMP-13 for postnatal growth, regeneration after acute injury, and in chronic injury from a genetic cross with dystrophic (mdx) mice. We also evaluated insulin-like growth factor 1 (IGF-1) mediated hypertrophy in the presence and absence of MMP-13. We employed live-cell imaging and 3D migration measurements on primary myoblasts obtained from these animals. Outcome measures included muscle morphology and function. RESULTS: Under basal conditions, Mmp13-/- mice did not exhibit histological or functional deficits in muscle. However, following acute injury, regeneration was impaired at 11 and 14 days post injury. Muscle hypertrophy caused by increased IGF-1 was blunted with minimal satellite cell incorporation in the absence of MMP-13. Mmp13-/- primary myoblasts displayed reduced migratory capacity in 2D and 3D, while maintaining normal proliferation and differentiation. Satellite cell specific deletion of MMP-13 recapitulated the effects of global MMP-13 ablation on muscle regeneration, growth and myoblast movement. CONCLUSION: These results show that satellite cells provide an essential autocrine source of MMP-13, which not only regulates their migration, but also supports postnatal growth and resolution of acute damage.


Asunto(s)
Movimiento Celular/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Músculo Esquelético/enzimología , Regeneración/genética , Células Satélite del Músculo Esquelético/enzimología , Animales , Movimiento Celular/fisiología , Matriz Extracelular/enzimología , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Regeneración/fisiología
2.
FASEB J ; 33(10): 10648-10667, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31268746

RESUMEN

Casein kinase 2 (CK2) is a tetrameric protein kinase composed of 2 catalytic (α and α') and 2 regulatory ß subunits. Our study provides the first molecular and cellular characterization of the different CK2 subunits, highlighting their individual roles in skeletal muscle specification and differentiation. Analysis of C2C12 cell knockout for each CK2 subunit reveals that: 1) CK2ß is mandatory for the expression of the muscle master regulator myogenic differentiation 1 in proliferating myoblasts, thus controlling both myogenic commitment and subsequent muscle-specific gene expression and myotube formation; 2) CK2α is involved in the activation of the muscle-specific gene program; and 3) CK2α' activity regulates myoblast fusion by mediating plasma membrane translocation of fusogenic proteins essential for membrane coalescence, like myomixer. Accordingly, CK2α' overexpression in C2C12 cells and in mouse regenerating muscle is sufficient to increase myofiber size and myonuclei content via enhanced satellite cell fusion. Consistent with these results, pharmacological inhibition of CK2 activity substantially blocks the expression of myogenic markers and muscle cell fusion both in vitro in C2C12 and primary myoblasts and in vivo in mouse regenerating muscle and zebrafish development. Overall, our work describes the specific and coordinated functions of CK2 subunits in orchestrating muscle differentiation and fusogenic activity, highlighting CK2 relevance in the physiopathology of skeletal muscle tissue.-Salizzato, V., Zanin, S., Borgo, C., Lidron, E., Salvi, M., Rizzuto, R., Pallafacchina, G., Donella-Deana, A. Protein kinase CK2 subunits exert specific and coordinated functions in skeletal muscle differentiation and fusogenic activity.


Asunto(s)
Quinasa de la Caseína II/fisiología , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Animales , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Fusión Celular , Línea Celular , Técnicas de Inactivación de Genes , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/enzimología , Subunidades de Proteína , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/enzimología , Pez Cebra , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
3.
FASEB J ; 33(7): 8094-8109, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30939245

RESUMEN

Skeletal muscle satellite cells (SMSCs), the major stem cells responsible for the regeneration of skeletal muscle, are normally cell cycle arrested but differentiate to generate myocytes upon muscle damage, forming new myofibers along with self-renewing stem cells in preparation for subsequent injury. In this study, we investigated which factors stimulate the proliferation and differentiation of SMSCs and found that pyruvate, the end product of glycolysis, stimulates their differentiation. Pyruvate antagonizes the effects of hypoxia on preferential self-renewal of SMSCs through dephosphorylation or activation of pyruvate dehydrogenase (PDH), which mediates opening of the gateway from glycolysis to the tricarboxylic acid (TCA) cycle by producing acetyl coenzyme A from pyruvate. PDH kinase 1, highly expressed under hypoxia, is down-regulated under normoxic conditions, leading to an increase in dephosphorylated PDH. Conditional deletion of PDH in SMSCs affects cell divisions generating myocytes and subsequent myotube formation, inefficient skeletal muscle regeneration upon injury, and aggravated pathogenesis of a dystrophin-deficient mouse model of Duchenne muscular dystrophy. Thus, the flow from glycolysis to the TCA cycle mediated by PDH plays a pivotal role in the differentiation of SMSCs, which is critical for the progression of skeletal muscle regeneration.-Hori, S., Hiramuki, Y., Nishimura, D., Sato, F., Sehara-Fujisawa, A. PDH-mediated metabolic flow is critical for skeletal muscle stem cell differentiation and myotube formation during regeneration in mice.


Asunto(s)
Diferenciación Celular , Cetona Oxidorreductasas/metabolismo , Fibras Musculares Esqueléticas/fisiología , Regeneración , Células Satélite del Músculo Esquelético/enzimología , Animales , Línea Celular , Ciclo del Ácido Cítrico , Eliminación de Gen , Glucólisis , Cetona Oxidorreductasas/genética , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/citología , Células Satélite del Músculo Esquelético/citología
4.
J Cell Physiol ; 234(4): 3192-3196, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30471096

RESUMEN

The phosphatase and tensin homolog (PTEN), originally identified as a tumor suppressor, is an important regulator of the PI3K-Akt pathway. PTEN plays crucial roles in various cellular processes, including cell survival, cell growth, cell proliferation, cell differentiation, and cell metabolism. In metabolic tissues, PTEN expression affects insulin sensitivity and glucose homeostasis. In skeletal muscle, the deletion of PTEN regulates muscle development and protects the mutant mice from insulin resistance and diabetes. Notably, the regulatory role of PTEN in skeletal muscle stem cells has been recently reported. In this review, we mainly discuss the role of PTEN in regulating the development, glucose metabolism, stem cell fate decision, and regeneration of skeletal muscle.


Asunto(s)
Músculo Esquelético/enzimología , Fosfohidrolasa PTEN/metabolismo , Células Satélite del Músculo Esquelético/enzimología , Animales , Glucosa/metabolismo , Homeostasis , Humanos , Desarrollo de Músculos , Regeneración , Transducción de Señal
5.
Antioxid Redox Signal ; 29(2): 128-148, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29669436

RESUMEN

AIMS: Muscle damage in Duchenne muscular dystrophy (DMD) caused by the lack of dystrophin is strongly linked to inflammation. Heme oxygenase-1 (HO-1; Hmox1) is an anti-inflammatory and cytoprotective enzyme affecting myoblast differentiation by inhibiting myomiRs. The role of HO-1 has not been so far well addressed in DMD. RESULTS: In dystrophin-deficient mdx mice, expression of Hmox1 in limb skeletal muscles and diaphragm is higher than in wild-type animals, being consistently elevated from 8 up to 52 weeks, both in myofibers and inflammatory leukocytes. Accordingly, HO-1 expression is induced in muscles of DMD patients. Pharmacological inhibition of HO-1 activity or genetic ablation of Hmox1 aggravates muscle damage and inflammation in mdx mice. Double knockout animals (Hmox1-/-mdx) demonstrate impaired exercise capacity in comparison with mdx mice. Interestingly, in contrast to the effect observed in muscle fibers, in dystrophin-deficient muscle satellite cells (SCs) expression of Hmox1 is decreased, while MyoD, myogenin, and miR-206 are upregulated compared with wild-type counterparts. Mdx SCs demonstrate disturbed and enhanced differentiation, which is further intensified by Hmox1 deficiency. RNA sequencing revealed downregulation of Atf3, MafK, Foxo1, and Klf2 transcription factors, known to activate Hmox1 expression, as well as attenuation of nitric oxide-mediated cGMP-dependent signaling in mdx SCs. Accordingly, treatment with NO-donor induces Hmox1 expression and inhibits differentiation. Finally, differentiation of mdx SCs was normalized by CO, a product of HO-1 activity. Innovation and Conclusions: HO-1 is induced in DMD, and HO-1 inhibition aggravates DMD pathology. Therefore, HO-1 can be considered a therapeutic target to alleviate this disease. Antioxid. Redox Signal. 00, 000-000.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Distrofia Muscular de Duchenne/enzimología , Células Satélite del Músculo Esquelético/enzimología , Animales , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Progresión de la Enfermedad , Distrofina/genética , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Humanos , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones Endogámicos mdx , Ratones Noqueados , MicroARNs/metabolismo , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Fenotipo , Células Satélite del Músculo Esquelético/citología
6.
Am J Physiol Regul Integr Comp Physiol ; 314(5): R741-R751, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29443546

RESUMEN

Skeletal muscle mass is determined by the net dynamic balance between protein synthesis and degradation. Although the Akt/mechanistic target of rapamycin (mTOR)-dependent pathway plays an important role in promoting protein synthesis and subsequent skeletal muscle hypertrophy, the precise molecular regulation of mTOR activity by the upstream protein kinase Akt is largely unknown. In addition, the activation of satellite cells has been indicated as a key regulator of muscle mass. However, the requirement of satellite cells for load-induced skeletal muscle hypertrophy is still under intense debate. In this study, female germline Akt1 knockout (KO) mice were used to examine whether Akt1 deficiency attenuates load-induced skeletal muscle hypertrophy through suppressing mTOR-dependent signaling and satellite cell proliferation. Akt1 KO mice showed a blunted hypertrophic response of skeletal muscle, with a diminished rate of satellite cell proliferation following mechanical overload. In contrast, Akt1 deficiency did not affect the load-induced activation of mTOR signaling and the subsequent enhanced rate of protein synthesis in skeletal muscle. These observations suggest that the load-induced activation of mTOR signaling occurs independently of Akt1 regulation and that Akt1 plays a critical role in regulating satellite cell proliferation during load-induced muscle hypertrophy.


Asunto(s)
Proliferación Celular , Músculo Esquelético/enzimología , Proteínas Proto-Oncogénicas c-akt/deficiencia , Células Satélite del Músculo Esquelético/enzimología , Animales , Femenino , Hipertrofia , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Noqueados , Músculo Esquelético/patología , Biosíntesis de Proteínas , Proteolisis , Proteínas Proto-Oncogénicas c-akt/genética , Células Satélite del Músculo Esquelético/patología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
7.
Med Sci Monit ; 23: 3562-3570, 2017 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-28731988

RESUMEN

BACKGROUND Preterm skeletal muscle genesis is a paradigm for myogenesis. The role of mitogen-activating protein kinase kinase kinase kinase-3 (MAP4K3) in preterm skeletal muscle satellite cells myogenesis or its relationship to mammalian target of rapamycin complex 1 (mTORC1) activity have not been previously elaborated. MATERIAL AND METHODS Small interfering RNA (siRNA) interference technology was used to inhibit MAP4K3 expression. Leucine stimulation experiments were performed following MAP4K3-siRNA interference. The differentiation of primary preterm skeletal muscle satellite cells was observed after siRNA-MAP4K3 interference. Western blot analysis was used to determine the expression of MAP4K3, MyHC, MyoD, myogenin, p-mTOR, and p-S6K1. The immunofluorescence fusion index of MyHC and myogenin were detected. MAP4K3 effects on preterm rat satellite cells differentiation and its relationship to mTORC1 activity are reported. RESULTS MAP4K3 siRNA knockdown inhibited myotube formation and both MyoD and myogenin expression in primary preterm rat skeletal muscle satellite cells, but MAP4K3 siRNA had no effect on the activity of mTORC1. In primary preterm rat skeletal muscle satellite cells, MAP4K3 knockdown resulted in significantly weaker, but not entirely blunted, leucine-induced mTORC1 signaling. CONCLUSIONS MAP4K3 positively regulates preterm skeletal muscle satellite cell myogenesis, but may not regulate mTORC1 activity. MAP4K3 may play a role in mTORC1 full activation in response to leucine.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Leucina/farmacología , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/enzimología , Miogenina/metabolismo , Fosforilación , ARN Interferente Pequeño/metabolismo , Ratas , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/enzimología , Transducción de Señal/efectos de los fármacos
8.
Nat Commun ; 8: 14328, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28094257

RESUMEN

Satellite cells (SCs) are myogenic stem cells required for regeneration of adult skeletal muscles. A proper balance among quiescence, activation and differentiation is essential for long-term maintenance of SCs and their regenerative function. Here we show a function of Pten (phosphatase and tensin homologue) in quiescent SCs. Deletion of Pten in quiescent SCs leads to their spontaneous activation and premature differentiation without proliferation, resulting in depletion of SC pool and regenerative failure. However, prior to depletion, Pten-null activated SCs can transiently proliferate upon injury and regenerate injured muscles, but continually decline during regeneration, suggesting an inability to return to quiescence. Mechanistically, Pten deletion increases Akt phosphorylation, which induces cytoplasmic translocation of FoxO1 and suppression of Notch signalling. Accordingly, constitutive activation of Notch1 prevents SC depletion despite Pten deletion. Our findings delineate a critical function of Pten in maintaining SC quiescence and reveal an interaction between Pten and Notch signalling.


Asunto(s)
Células Madre Adultas/enzimología , Senescencia Celular , Fosfohidrolasa PTEN/metabolismo , Células Satélite del Músculo Esquelético/enzimología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Femenino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Masculino , Ratones , Ratones Noqueados , Desarrollo de Músculos , Fosfohidrolasa PTEN/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo
9.
Skelet Muscle ; 6: 22, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27239264

RESUMEN

BACKGROUND: Euchromatic histone-lysine N-methyltransferase 2 (G9a/Ehmt2) is the main enzyme responsible for the apposition of H3K9 di-methylation on histones. Due to its dual role as an epigenetic regulator and in the regulation of non-histone proteins through direct methylation, G9a has been implicated in a number of biological processes relevant to cell fate control. Recent reports employing in vitro cell lines indicate that Ehmt2 methylates MyoD to repress its transcriptional activity and therefore its ability to induce differentiation of activated myogenic cells. METHODS: To further investigate the importance of G9a in modulating myogenic regeneration in vivo, we crossed Ehmt2 (floxed) mice to animals expressing Cre recombinase from the Myod locus, resulting in efficient knockout in the entire skeletal muscle lineage (Ehmt2 (ΔmyoD) ). RESULTS: Surprisingly, despite a dramatic drop in the global levels of H3K9me2, knockout animals did not show any developmental phenotype in muscle size and appearance. Consistent with this finding, purified Ehmt2 (ΔmyoD) satellite cells had rates of activation and proliferation similar to wild-type controls. When induced to differentiate in vitro, Ehmt2 knockout cells differentiated with kinetics similar to those of control cells and demonstrated normal capacity to form myotubes. After acute muscle injury, knockout mice regenerated as efficiently as wildtype. To exclude possible compensatory mechanisms elicited by the loss of G9a during development, we restricted the knockout within adult satellite cells by crossing Ehmt2 (floxed) mice to Pax7 (CreERT2) and also found normal muscle regeneration capacity. CONCLUSIONS: Thus, Ehmt2 and H3K9me2 do not play significant roles in skeletal muscle development and regeneration in vivo.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/deficiencia , Desarrollo de Músculos , Músculo Esquelético/enzimología , Enfermedades Musculares/enzimología , Regeneración , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Venenos Elapídicos , Regulación del Desarrollo de la Expresión Génica , Genotipo , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Cinética , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Fenotipo , Células Satélite del Músculo Esquelético/enzimología , Células Satélite del Músculo Esquelético/patología , Transducción de Señal
10.
J Clin Invest ; 126(4): 1555-65, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26999603

RESUMEN

The X chromosome-encoded histone demethylase UTX (also known as KDM6A) mediates removal of repressive trimethylation of histone H3 lysine 27 (H3K27me3) to establish transcriptionally permissive chromatin. Loss of UTX in female mice is embryonic lethal. Unexpectedly, male UTX-null mice escape embryonic lethality due to expression of UTY, a paralog that lacks H3K27 demethylase activity, suggesting an enzyme-independent role for UTX in development and thereby challenging the need for active H3K27 demethylation in vivo. However, the requirement for active H3K27 demethylation in stem cell-mediated tissue regeneration remains untested. Here, we employed an inducible mouse KO that specifically ablates Utx in satellite cells (SCs) and demonstrated that active H3K27 demethylation is necessary for muscle regeneration. Loss of UTX in SCs blocked myofiber regeneration in both male and female mice. Furthermore, we demonstrated that UTX mediates muscle regeneration through its H3K27 demethylase activity, as loss of demethylase activity either by chemical inhibition or knock-in of demethylase-dead UTX resulted in defective muscle repair. Mechanistically, dissection of the muscle regenerative process revealed that the demethylase activity of UTX is required for expression of the transcription factor myogenin, which in turn drives differentiation of muscle progenitors. Thus, we have identified a critical role for the enzymatic activity of UTX in activating muscle-specific gene expression during myofiber regeneration and have revealed a physiological role for active H3K27 demethylation in vivo.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Histona Demetilasas/biosíntesis , Miofibrillas/fisiología , Miogenina/metabolismo , Regeneración/fisiología , Células Satélite del Músculo Esquelético/enzimología , Animales , Femenino , Técnicas de Sustitución del Gen , Histona Demetilasas/genética , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Ratones Noqueados , Miogenina/genética , Células Satélite del Músculo Esquelético/citología
11.
J Clin Invest ; 126(4): 1233-5, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26999609

RESUMEN

Precise epigenetic modifications in stem cells control developmental programs and cell fate decisions. In particular, the addition or removal of trimethylation of histone 3 lysine 27 (H3K27me3) at lineage-specific genes has been linked to the repression of gene expression, and a precise balance of methyltransferases and demethylases within cells determines H3K27me3 levels. The demethylase UTX is essential for development and tissue homeostasis; however, a role for UTX in stem cell-mediated tissue regeneration is unknown. In this issue of the JCI, Dilworth and colleagues reveal that UTX and its demethylase activity are required in the muscle stem cell lineage for muscle regeneration in response to injury. Specifically, UTX mediates the removal of H3K27me3 in the promoter of the transcription factor myogenin, which regulates myogenic differentiation. The results of this study provide important insight into the contribution of epigenetic regulation in stem cell-mediated regeneration of adult tissues.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Histona Demetilasas/biosíntesis , Miofibrillas/fisiología , Miogenina/metabolismo , Regeneración/fisiología , Células Satélite del Músculo Esquelético/enzimología , Animales , Femenino , Masculino
12.
Skelet Muscle ; 6: 9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26981231

RESUMEN

BACKGROUND: Extracellular stimuli induce gene expression responses through intracellular signaling mediators. The p38 signaling pathway is a paradigm of the mitogen-activated protein kinase (MAPK) family that, although originally identified as stress-response mediator, contributes to establishing stem cell differentiation fates. p38α is central for induction of the differentiation fate of the skeletal muscle stem cells (satellite cells) through not fully characterized mechanisms. METHODS: To investigate the global gene transcription program regulated by p38α during satellite cell differentiation (myogenesis), and to specifically address whether this regulation occurs through direct action of p38α on gene promoters, we performed a combination of microarray gene expression and genome-wide binding analyses. For experimental robustness, two myogenic cellular systems with genetic and chemical loss of p38α function were used: (1) satellite cells derived from mice with muscle-specific deletion of p38α, and (2) the C2C12 murine myoblast cell line cultured in the absence or presence of the p38α/ß inhibitor SB203580. Analyses were performed at cell proliferation and early differentiation stages. RESULTS: We show that p38α binds to a large set of active promoters during the transition of myoblasts from proliferation to differentiation stages. p38α-bound promoters are enriched with binding motifs for several transcription factors, with Sp1, Tcf3/E47, Lef1, FoxO4, MyoD, and NFATc standing out in all experimental conditions. p38α association with chromatin correlates very well with high levels of transcription, in agreement with its classical function as an activator of myogenic differentiation. Interestingly, p38α also associates with genes repressed at the onset of differentiation, thus highlighting the relevance of p38-dependent chromatin regulation for transcriptional activation and repression during myogenesis. CONCLUSIONS: These results uncover p38α association and function on chromatin at novel classes of target genes during skeletal muscle cell differentiation. This is consistent with this MAPK isoform being a transcriptional regulator.


Asunto(s)
Diferenciación Celular , Inmunoprecipitación de Cromatina , Cromatina/metabolismo , Perfilación de la Expresión Génica , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Desarrollo de Músculos , Células Satélite del Músculo Esquelético/enzimología , Animales , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Genotipo , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Desarrollo de Músculos/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Células Satélite del Músculo Esquelético/efectos de los fármacos , Transducción de Señal , Transcripción Genética
13.
Nat Commun ; 6: 10123, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26648529

RESUMEN

Satellite cells are resident adult stem cells that are required for regeneration of skeletal muscle. However, signalling mechanisms that regulate satellite cell function are less understood. Here we demonstrate that transforming growth factor-ß-activated kinase 1 (TAK1) is important in satellite stem cell homeostasis and function. Inactivation of TAK1 in satellite cells inhibits muscle regeneration in adult mice. TAK1 is essential for satellite cell proliferation and its inactivation causes precocious differentiation. Moreover, TAK1-deficient satellite cells exhibit increased oxidative stress and undergo spontaneous cell death, primarily through necroptosis. TAK1 is required for the activation of NF-κB and JNK in satellite cells. Forced activation of NF-κB improves survival and proliferation of TAK1-deficient satellite cells. Furthermore, TAK1-mediated activation of JNK is essential to prevent oxidative stress and precocious differentiation of satellite cells. Collectively, our study suggests that TAK1 is required for maintaining the pool of satellite stem cells and for regenerative myogenesis.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Músculo Esquelético/fisiología , Regeneración , Células Satélite del Músculo Esquelético/enzimología , Células Madre/enzimología , Animales , Muerte Celular , Diferenciación Celular , Femenino , Quinasas Quinasa Quinasa PAM/genética , Masculino , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Células Satélite del Músculo Esquelético/citología , Células Madre/citología
14.
J Biol Chem ; 290(44): 26445-56, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26370082

RESUMEN

Satellite cells are the major myogenic stem cells residing inside skeletal muscle and are indispensable for muscle regeneration. Satellite cells remain largely quiescent but are rapidly activated in response to muscle injury, and the derived myogenic cells then fuse to repair damaged muscle fibers or form new muscle fibers. However, mechanisms eliciting metabolic activation, an inseparable step for satellite cell activation following muscle injury, have not been defined. We found that a noncanonical Sonic Hedgehog (Shh) pathway is rapidly activated in response to muscle injury, which activates AMPK and induces a Warburg-like glycolysis in satellite cells. AMPKα1 is the dominant AMPKα isoform expressed in satellite cells, and AMPKα1 deficiency in satellite cells impairs their activation and myogenic differentiation during muscle regeneration. Drugs activating noncanonical Shh promote proliferation of satellite cells, which is abolished because of satellite cell-specific AMPKα1 knock-out. Taken together, AMPKα1 is a critical mediator linking noncanonical Shh pathway to Warburg-like glycolysis in satellite cells, which is required for satellite activation and muscle regeneration.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucólisis/fisiología , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/enzimología , Proteínas Quinasas Activadas por AMP/genética , Animales , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/citología
15.
Cell Biol Int ; 39(11): 1264-73, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26041412

RESUMEN

Mammalian target of rapamycin (mTOR) signaling pathway plays a key role in muscle development and is involved in multiple intracellular signaling pathways. Myocyte enhancer factor-2 (MEF2) regulates muscle cell proliferation and differentiation. However, how the mTOR signaling pathway regulates MEF2 activity remains unclear. We isolated goat skeletal muscle satellite cells (gSSCs) as model cells to explore mTOR signaling pathway regulation of MEF2C. We inhibited mTOR activity in gSSCs with PP242 and found that MEF2C phosphorylation was decreased and that muscle creatine kinase (MCK) expression was suppressed. Subsequently, we detected integrin-linked kinase (ILK) using MEF2C coimmunoprecipitation; ILK and MEF2C were colocalized in the gSSCs. We found that inhibiting mTOR activity increased ILK phosphorylation levels and that inhibiting ILK activity with Cpd 22 and knocking down ILK with small interfering RNA increased MEF2C phosphorylation and MCK expression. In the presence of Cpd 22, mTOR activity inhibition did not affect MEF2C phosphorylation. Moreover, ILK dephosphorylated MEF2C in vitro. These results suggest that the mTOR signaling pathway regulates MEF2C positively and regulates ILK negatively and that ILK regulates MEF2C negatively. It appears that the mTOR signaling pathway regulates MEF2C through ILK, further regulating the expression of muscle-related genes in gSSCs.


Asunto(s)
Factores de Transcripción MEF2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Cabras , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Fosforilación , Células Satélite del Músculo Esquelético/enzimología , Transducción de Señal
16.
Am J Physiol Heart Circ Physiol ; 309(2): H325-34, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25980021

RESUMEN

Chronic failure in maintenance and regeneration of skeletal muscles leads to lower muscle mass (sarcopenia), muscle weakness, and poor response to injury. Evidence suggests that aberrant p38 MAPK signaling undermines the repair process after injury in aged mice. Previous studies have shown that hyperhomocysteinemia (HHcy) has been associated with muscle weakness and lower than normal body weights. However, whether or not HHcy condition also compromises skeletal muscle regenerative capabilities is not clear. In the current study, we show that CBS-/+ mice, a model for HHcy condition, exhibited compromised regenerative function and cell proliferation upon injury. However, there was no significant difference in Pax7 expression levels in the satellite cells from CBS-/+ mouse skeletal muscles. Interestingly, the satellite cells from CBS-/+ mice not only exhibited diminished in vitro proliferative capabilities, but also there was heightened oxidative stress. In addition, there was enhanced p38 MAPK activation as well as p16 and p21 expression in the CBS-/+ mouse satellite cells. Moreover, the C2C12 myoblasts also exhibited higher p38 MAPK activation and p16 expression upon treatment with homocysteine in addition to enhanced ROS presence. Tissue engraftment potential and regeneration after injury were restored to some extent upon treatment with the p38-MAPK inhibitor, SB203580, in the CBS-/+ mice. These results together suggest that HHcy-induced diminished satellite cell proliferation involves excessive oxidative stress and p38 MAPK signaling. Our study further proposes that HHcy is a potential risk factor for elderly frailty, and need to be considered as a therapeutic target while designing the alleviation interventions/postinjury rehabilitation measures for adults with HHcy.


Asunto(s)
Proliferación Celular , Hiperhomocisteinemia/enzimología , Sistema de Señalización de MAP Quinasas , Músculo Esquelético/enzimología , Regeneración , Células Satélite del Músculo Esquelético/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Cistationina betasintasa/deficiencia , Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Activación Enzimática , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/patología , Hiperhomocisteinemia/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Estrés Oxidativo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Sarcopenia/enzimología , Sarcopenia/patología , Sarcopenia/fisiopatología , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/patología , Células Satélite del Músculo Esquelético/trasplante , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
17.
J Am Heart Assoc ; 2(6): e000376, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24308935

RESUMEN

BACKGROUND: Surgical treatment of peripheral artery disease, even if successful, does not prevent reoccurrence. Under these conditions, increased oxidative stress is a crucial determinant of tissue damage. Given its reported antioxidant effects, we investigated the potential of unacylated-ghrelin (UnAG) to reduce ischemia-induced tissue damage in a mouse model of peripheral artery disease. METHODS AND RESULTS: We show that UnAG but not acylated ghrelin (AG) induces skeletal muscle regeneration in response to ischemia via canonical p38/mitogen-actived protein kinase signaling UnAG protected against reactive oxygen species-induced cell injuries by inducing the expression of superoxide dismutase-2 (SOD-2) in satellite cells. This led to a reduced number of infiltrating CD68(+) cells and was followed by induction of the myogenic process and a reduction in functional impairment. Moreover, we found that miR-221/222, previously linked to muscle regeneration processes, was up-regulated and negatively correlated with p57(Kip2) expression in UnAG-treated mice. UnAG, unlike AG, promoted cell-cycle entry in satellite cells of mice lacking the genes for ghrelin and its receptor (GHSR1a). UnAG-induced p38/mitogen-actived protein kinase phosphorylation, leading to activation of the myogenic process, was prevented in SOD-2-depleted SCs. By siRNA technology, we also demonstrated that SOD-2 is the antioxidant enzyme involved in the control of miR-221/222-driven posttranscriptional p57(Kip2) regulation. Loss-of-function experiments targeting miR-221/222 and local pre-miR-221/222 injection in vivo confirmed a role for miR-221/222 in driving skeletal muscle regeneration after ischemia. CONCLUSIONS: These results indicate that UnAG-induced skeletal muscle regeneration after ischemia depends on SOD-2-induced miR-221/222 expression and highlight its clinical potential for the treatment of reactive oxygen species-mediated skeletal muscle damage.


Asunto(s)
Antioxidantes/farmacología , Ghrelina/farmacología , Isquemia/tratamiento farmacológico , MicroARNs/metabolismo , Músculo Esquelético/efectos de los fármacos , Regeneración/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Ghrelina/análogos & derivados , Ghrelina/deficiencia , Ghrelina/genética , Miembro Posterior , Isquemia/enzimología , Isquemia/genética , Isquemia/patología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Proteína MioD/metabolismo , Estrés Oxidativo/efectos de los fármacos , Factor de Transcripción PAX7/metabolismo , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Receptores de Ghrelina/deficiencia , Receptores de Ghrelina/genética , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/enzimología , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/genética , Factores de Tiempo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Cell Death Dis ; 4: e955, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24336075

RESUMEN

Satellite cell (SC) proliferation and differentiation have critical roles in skeletal muscle recovery after injury and adaptation in response to hypertrophic stimuli. Normal ageing hinders SC proliferation and differentiation, and is associated with increased expression of a number of pro-apoptotic factors in skeletal muscle. In light of previous studies that have demonstrated age-related altered expression of genes involved in SC antioxidant and repair activity, this investigation was aimed at evaluating the incidence of apoptotic features in human SCs. Primary cells were obtained from vastus lateralis of nine young (27.3±2.0 years old) and nine old (71.1±1.8 years old) subjects, and cultured in complete medium for analyses at 4, 24, 48, and 72 h. Apoptosis was assessed using AnnexinV/propidium iodide staining, the terminal deoxynucleotidyl transferase dUTP nick-end labelling technique, RT-PCR, DNA microarrays, flow cytometry, and immunofluorescence analysis. There was an increased rate of apoptotic cells in aged subjects at all of the experimental time points, with no direct correlation between AnnexinV-positive cells and caspase-8 activity. On the other hand, CASP2, CASP6, CASP7, and CASP9 and a number of cell death genes were upregulated in the aged SCs. Altogether, our data show age-related enhanced susceptibility of human SCs to apoptosis, which might be responsible for their reduced response to muscle damage.


Asunto(s)
Envejecimiento/fisiología , Caspasas/metabolismo , Células Satélite del Músculo Esquelético/enzimología , Adulto , Anciano , Apoptosis/fisiología , Caspasa 2/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Cisteína Endopeptidasas/metabolismo , Femenino , Citometría de Flujo , Humanos , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
Stem Cells ; 31(8): 1597-610, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23592450

RESUMEN

Stem cell function is essential for the maintenance of adult tissue homeostasis. Controlling the balance between self-renewal and differentiation is crucial to maintain a receptive satellite cell pool capable of responding to growth and regeneration cues. The mitogen-activated protein kinase p38α has been implicated in the regulation of these processes but its influence in adult muscle remains unknown. Using conditional satellite cell p38α knockout mice we have demonstrated that p38α restricts excess proliferation in the postnatal growth phase while promoting timely myoblast differentiation. Differentiation was still able to occur in the p38α-null satellite cells, however, but was delayed. An absence of p38α resulted in a postnatal growth defect along with the persistence of an increased reservoir of satellite cells into adulthood. This population was still capable of responding to cardiotoxin-induced injury, resulting in complete, albeit delayed, regeneration, with further enhancement of the satellite cell population. Increased p38γ phosphorylation accompanied the absence of p38α, and inhibition of p38γ ex vivo substantially decreased the myogenic defect. We have used genome-wide transcriptome analysis to characterize the changes in expression that occur between resting and regenerating muscle, and the influence p38α has on these expression profiles. This study provides novel evidence for the fundamental role of p38α in adult muscle homeostasis in vivo.


Asunto(s)
Células Madre Adultas/química , Células Madre Adultas/enzimología , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/enzimología , Animales , Animales Recién Nacidos , Procesos de Crecimiento Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Músculos/lesiones , Músculos/fisiología , Fosforilación , Regeneración/fisiología
20.
Biochim Biophys Acta ; 1831(1): 167-75, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22750505

RESUMEN

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid whose actions are essential for many physiological processes including angiogenesis, lymphocyte trafficking and development. In addition, S1P serves as a muscle trophic factor that enables efficient muscle regeneration. This is due in part to S1P's ability to activate quiescent muscle stem cells called satellite cells (SCs) that are needed for muscle repair. However, the molecular mechanism by which S1P activates SCs has not been well understood. Further, strategies for harnessing S1P signaling to recruit SCs for therapeutic benefit have been lacking. S1P is irreversibly catabolized by S1P lyase (SPL), a highly conserved enzyme that catalyzes the cleavage of S1P at carbon bond C(2-3), resulting in formation of hexadecenal and ethanolamine-phosphate. SPL enhances apoptosis through substrate- and product-dependent events, thereby regulating cellular responses to chemotherapy, radiation and ischemia. SPL is undetectable in resting murine skeletal muscle. However, we recently found that SPL is dynamically upregulated in skeletal muscle after injury. SPL upregulation occurred in the context of a tightly orchestrated genetic program that resulted in a transient S1P signal in response to muscle injury. S1P activated quiescent SCs via a sphingosine-1-phosphate receptor 2 (S1P2)/signal transducer and activator of transcription 3 (STAT3)-dependent pathway, thereby facilitating skeletal muscle regeneration. Mdx mice, which serve as a model for muscular dystrophy (MD), exhibited skeletal muscle SPL upregulation and S1P deficiency. Pharmacological SPL inhibition raised skeletal muscle S1P levels, enhanced SC recruitment and improved mdx skeletal muscle regeneration. These findings reveal how S1P can activate SCs and indicate that SPL suppression may provide a therapeutic strategy for myopathies. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.


Asunto(s)
Aldehído-Liasas/metabolismo , Músculo Esquelético/enzimología , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/enzimología , Aldehído-Liasas/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Humanos , Lisofosfolípidos/metabolismo , Células Satélite del Músculo Esquelético/patología , Esfingosina/análogos & derivados , Esfingosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...