Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
1.
Mol Cell ; 81(2): 340-354.e5, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33450210

RESUMEN

In addition to its role as an electron transporter, mitochondrial nicotinamide adenine dinucleotide (NAD+) is an important co-factor for enzymatic reactions, including ADP-ribosylation. Although mitochondria harbor the most intra-cellular NAD+, mitochondrial ADP-ribosylation remains poorly understood. Here we provide evidence for mitochondrial ADP-ribosylation, which was identified using various methodologies including immunofluorescence, western blot, and mass spectrometry. We show that mitochondrial ADP-ribosylation reversibly increases in response to respiratory chain inhibition. Conversely, H2O2-induced oxidative stress reciprocally induces nuclear and reduces mitochondrial ADP-ribosylation. Elevated mitochondrial ADP-ribosylation, in turn, dampens H2O2-triggered nuclear ADP-ribosylation and increases MMS-induced ARTD1 chromatin retention. Interestingly, co-treatment of cells with the mitochondrial uncoupler FCCP decreases PARP inhibitor efficacy. Together, our results suggest that mitochondrial ADP-ribosylation is a dynamic cellular process that impacts nuclear ADP-ribosylation and provide evidence for a NAD+-mediated mitochondrial-nuclear crosstalk.


Asunto(s)
ADP-Ribosilación , Núcleo Celular/enzimología , Mitocondrias/enzimología , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , ADP-Ribosilación/efectos de los fármacos , Animales , Antimicina A/análogos & derivados , Antimicina A/farmacología , Línea Celular , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Cromatina/química , Cromatina/metabolismo , Transporte de Electrón/efectos de los fármacos , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Metacrilatos/farmacología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mioblastos/citología , Mioblastos/efectos de los fármacos , Mioblastos/enzimología , Oligomicinas/farmacología , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/enzimología , Poli(ADP-Ribosa) Polimerasa-1/genética , Rotenona/farmacología , Tiazoles/farmacología
2.
Nucleic Acids Res ; 48(20): 11452-11467, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33080014

RESUMEN

Msh homeobox (Msx) is a subclass of homeobox transcriptional regulators that control cell lineage development, including the early stage of vertebrate limb development, although the underlying mechanisms are not clear. Here, we demonstrate that Msx1 promotes the proliferation of myoblasts and mesenchymal stem cells (MSCs) by enhancing mitogen-activated protein kinase (MAPK) signaling. Msx1 directly binds to and upregulates the expression of fibroblast growth factor 9 (Fgf9) and Fgf18. Accordingly, knockdown or antibody neutralization of Fgf9/18 inhibits Msx1-activated extracellular signal-regulated kinase 1/2 (Erk1/2) phosphorylation. Mechanistically, we determined that the phosphorylation of Msx1 at Ser136 is critical for enhancing Fgf9 and Fgf18 expression and cell proliferation, and cyclin-dependent kinase 1 (CDK1) is apparently responsible for Ser136 phosphorylation. Furthermore, mesenchymal deletion of Msx1/2 results in decreased Fgf9 and Fgf18 expression and Erk1/2 phosphorylation, which leads to serious defects in limb development in mice. Collectively, our findings established an important function of the Msx1-Fgf-MAPK signaling axis in promoting cell proliferation, thus providing a new mechanistic insight into limb development.


Asunto(s)
Proliferación Celular , Extremidades/embriología , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Sistema de Señalización de MAP Quinasas , Factor de Transcripción MSX1/metabolismo , Animales , Proteína Quinasa CDC2/metabolismo , Línea Celular , Factor 9 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/genética , Factor de Transcripción MSX1/química , Factor de Transcripción MSX1/genética , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Mioblastos/citología , Mioblastos/enzimología , Mioblastos/metabolismo , Fosforilación , Serina/metabolismo
3.
J Cell Physiol ; 236(7): 5293-5305, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33378552

RESUMEN

The ubiquitin-proteasome system is a major protein degradation pathway in the cell. Proteasomes produce several peptides that are rapidly degraded to free amino acids by intracellular aminopeptidases. Our previous studies reported that proteolysis via proteasomes and aminopeptidases is required for myoblast proliferation and differentiation. However, the role of intracellular aminopeptidases in myoblast proliferation and differentiation had not been clarified. In this study, we investigated the effects of puromycin-sensitive aminopeptidase (PSA) on C2C12 myoblast proliferation and differentiation by knocking down PSA. Aminopeptidase enzymatic activity was reduced in PSA-knockdown myoblasts. Knockdown of PSA induced impaired cell cycle progression in C2C12 myoblasts and accumulation of cells at the G2/M phase. Additionally, after the induction of myogenic differentiation in PSA-knockdown myoblasts, multinucleated circular-shaped myotubes with impaired cell polarity were frequently identified. Cell division cycle 42 (CDC42) knockdown in myoblasts resulted in a loss of cell polarity and the formation of multinucleated circular-shaped myotubes, which were similar to PSA-knockdown myoblasts. These data suggest that PSA is required for the proliferation of myoblasts in the growth phase and for the determination of cell polarity and elongation of myotubes in the differentiation phase.


Asunto(s)
Aminopeptidasas/metabolismo , Desarrollo de Músculos/fisiología , Mioblastos/enzimología , Animales , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular/fisiología , Ratones
4.
Exp Cell Res ; 397(1): 112337, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33091420

RESUMEN

A large number of intracellular proteins are degraded by the ubiquitin-proteasome system, one of the major protein degradation pathways. It produces peptides of several different sizes through protein degradation, and these peptides are rapidly degraded into free amino acids by various intracellular aminopeptidases. Previously, we reported that the activity of proteasomes and aminopeptidases in the proteolysis pathway are necessary for myoblast proliferation and differentiation. However, the detailed function of intracellular aminopeptidases in myoblast proliferation and differentiation has not yet been elucidated. In this study, we focused on alanine aminopeptidase (APN) and investigated the function of APN in C2C12 myoblast proliferation and differentiation. In myoblasts and myotubes, APN was mainly localized in the cell membrane as well as expressed at low levels in the cytoplasm and nucleus. The reduction of the APN enzymatic activity impaired the cell cycle progression in C2C12 myoblasts. In addition, apoptosis was induced after APN-knockdown. Finally, myogenic differentiation was also delayed in the APN-suppressed myoblasts. These findings indicate that APN is required for myoblast proliferation and differentiation.


Asunto(s)
Antígenos CD13/antagonistas & inhibidores , Diferenciación Celular , Proliferación Celular , Mioblastos/patología , ARN Interferente Pequeño/genética , Animales , Apoptosis , Antígenos CD13/genética , Antígenos CD13/metabolismo , Ratones , Mioblastos/enzimología
5.
Biochem Biophys Res Commun ; 524(3): 608-613, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32029277

RESUMEN

The ubiquitin-proteasome pathway is essential for skeletal muscle growth and development. Proteasomes generate oligopeptides in the cytoplasm, and these peptides are considered to be rapidly degraded to amino acids by several intracellular aminopeptidases. However, the role of intracellular aminopeptidases in muscle growth remains unknown. In this study, therefore, we investigated the role of intracellular aminopeptidases in C2C12 myoblast proliferation and differentiation. Inhibition of intracellular aminopeptidases by Bestatin methyl ester (Bes-ME) decreased leucine and alanine aminopeptidase activity, and impaired proliferation and differentiation of C2C12 myoblasts. Furthermore, we observed that the inhibition of intracellular aminopeptidases reduced intracellular levels of amino acid and ATP level, and suppressed the phosphorylation of the mTOR pathway. These results suggested that intracellular aminopeptidases affect C2C12 myoblast proliferation and differentiation via mTOR pathway; however, further studies are required to clarify the role of aminopeptidase in skeletal muscle.


Asunto(s)
Aminopeptidasas/metabolismo , Diferenciación Celular , Espacio Intracelular/enzimología , Mioblastos/citología , Mioblastos/enzimología , Adenosina Trifosfato/metabolismo , Aminoácidos/metabolismo , Aminopeptidasas/antagonistas & inhibidores , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ratones , Mioblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
6.
Am J Physiol Cell Physiol ; 317(5): C964-C968, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31461343

RESUMEN

Whether the histone deacetylase (HDAC) and sirtuin families of protein deacetylases regulate insulin-stimulated glucose uptake, independent of their transcriptional effects, has not been studied. Our objective was to determine the nontranscriptional role of HDACs and sirtuins in regulation of skeletal muscle insulin action. Basal and insulin-stimulated glucose uptake and signaling and acetylation were assessed in L6 myotubes and skeletal muscle from C57BL/6J mice that were treated acutely (1 h) with HDAC (trichostatin A, panobinostat, TMP195) and sirtuin inhibitors (nicotinamide). Treatment of L6 myotubes with HDAC inhibitors or skeletal muscle with a combination of HDAC and sirtuin inhibitors increased tubulin and pan-protein acetylation, demonstrating effective impairment of HDAC and sirtuin deacetylase activities. Despite this, neither basal nor insulin-stimulated glucose uptake or insulin signaling was impacted. Acute reduction of the deacetylase activity of HDACs and/or sirtuins does not impact insulin action in skeletal muscle.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Insulina/metabolismo , Músculo Esquelético/enzimología , Mioblastos/enzimología , Animales , Células Cultivadas , Femenino , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Mioblastos/efectos de los fármacos
7.
Exp Cell Res ; 364(2): 184-190, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29425714

RESUMEN

Mammalian target of rapamycin (mTOR) signaling controls skeletal muscle cell differentiation, growth, and metabolism by sensing the intracellular energy status and nutrients. Recently, leucyl-tRNA synthetase (Lars) was identified as an intracellular sensor of leucine involved in the activation of mTOR signaling. However, there is still no evidence for the activation of mTOR signaling by Lars and its physiological roles in skeletal muscle cells. In this study, we determined the potential roles of Lars for the activation of mTOR signaling, skeletal muscle cell differentiation, hypertrophy, and metabolism using small interfering (si)-RNA knockdown. siRNA-mediated knockdown of Lars decreased phosphorylated p70 S6 kinase and inhibited the differentiation of C2C12 mouse myoblasts into myotubes, as evidenced by a decreased fusion index and decreased mRNA and protein expression levels of myogenic markers. Importantly, si-Lars decreased the level of Insulin-like growth factor 2 (Igf2) mRNA expression from the early stages of differentiation, indicating the possibility of an association between the mTOR-IGF2 axis and Lars. However, Lars knockdown did not decrease phosphorylated mTOR in differentiated myotubes, nor did it affect the hypertrophy of myotubes as evidenced by measuring their diameters and detecting the mRNA and protein expression of hypertrophy markers. Similarly, an extracellular flux analyzer showed that Lars knockdown did not affect the metabolism (glycolysis and mitochondrial respiration) of myotubes. These results demonstrate that Lars is required for skeletal muscle differentiation through the activation of mTOR signaling, but not for hypertrophy or metabolic alteration of myotubes.


Asunto(s)
Leucina-ARNt Ligasa/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citología , Mioblastos/enzimología , Animales , Células Cultivadas , Ratones , Mioblastos/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
8.
Am J Physiol Cell Physiol ; 314(4): C415-C427, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351412

RESUMEN

Fibrosis is a common feature of several chronic diseases and is characterized by exacerbated accumulation of ECM. An understanding of the cellular and molecular mechanisms involved in the development of this condition is crucial for designing efficient treatments for those pathologies. Connective tissue growth factor (CTGF/CCN2) is a pleiotropic protein with strong profibrotic activity. In this report, we present experimental evidence showing that ECM stimulates the synthesis of CTGF in response to lysophosphatidic acid (LPA).The integrin/focal adhesion kinase (FAK) signaling pathway mediates this effect, since CTGF expression is abolished by the use of the Arg-Gly-Asp-Ser peptide and also by an inhibitor of FAK autophosphorylation at tyrosine 397. Cilengitide, a specific inhibitor of αv integrins, inhibits the expression of CTGF mediated by LPA or transforming growth factor ß1. We show that ECM obtained from decellularized myofibroblast cultures or derived from activated fibroblasts from muscles of the Duchenne muscular dystrophy mouse model ( mdx) induces the expression of CTGF. This effect is dependent on FAK phosphorylation in response to its activation by integrin. We also found that the fibrotic ECM inhibits skeletal muscle differentiation. This novel regulatory mechanism of CTGF expression could be acting as a positive profibrotic feedback between the ECM and CTGF, revealing a novel concept in the control of fibrosis under chronic damage.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Matriz Extracelular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Integrina alfaV/metabolismo , Lisofosfolípidos/farmacología , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/enzimología , Mioblastos/efectos de los fármacos , Animales , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Modelos Animales de Enfermedad , Matriz Extracelular/enzimología , Matriz Extracelular/patología , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Integrina alfaV/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Mioblastos/enzimología , Mioblastos/patología , Fosforilación , Transducción de Señal/efectos de los fármacos
9.
J Cell Physiol ; 233(12): 9473-9487, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29943814

RESUMEN

Mechanical overloading can lead to skeletal muscle damage instead of remodeling. This is attributed to the excessive apoptosis of myoblasts, mechanism of which remains to be elucidated. The present study aimed to investigate the involvement of endoplasmic reticulum stress (ERS) and caspase-12 in mediating the stretch-induced apoptosis of myoblasts. Myoblast apoptosis was evaluated by Hoechst staining, DNA fragmentation assay, Annexin V binding, and propidium iodide staining, as well as caspase-3 and poly-ADP-ribose polymerase 1 cleavage. First, our results showed that apoptosis was elevated in a time-dependent manner when myoblasts were subjected to cyclic mechanical stretch (CMS) for 12, 24, and 36 hr. Concomitantly, CMS triggered the ERS and caspase-12 cleavage; ERS inhibitor GSK 2606414 suppressed the CMS-induced cleavage of caspase-12 and myoblast apoptosis. Silencing caspase-12 attenuated the apoptosis of myoblasts under CMS. Furthermore, CMS-induced myoblast apoptosis was partially recovered by overexpressing wild-type caspase-12 in caspase-12-silenced myoblasts. In contrast, overexpressing mutant caspase-12 (D94N), which cannot be cleaved into the active caspase-12 fragments, failed to accomplish the same effect. Finally, C2C12 overexpressing truncated caspase-12 segment (TC-casp12-D94), which starts from Asp94 and ends at Asn419, underwent apoptosis under both static and stretched conditions. Interestingly, C2C12 myoblasts seemed to be resistant to stretch-induced apoptosis upon low-serum-induced differentiation. In conclusion, our study provided evidence that caspase-12 cleavage at Asp94, induced by ERS under mechanical stimuli, is the key molecule in initiating the stretch-triggered apoptosis of myoblasts.


Asunto(s)
Apoptosis , Ácido Aspártico/metabolismo , Caspasa 12/metabolismo , Estrés del Retículo Endoplásmico , Mioblastos/enzimología , Mioblastos/patología , Estrés Mecánico , Animales , Línea Celular , Ratones , Fenotipo , Factores de Tiempo
10.
Int J Mol Sci ; 19(6)2018 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-29865254

RESUMEN

Myoblast proliferation is crucial to skeletal muscle hypertrophy and regeneration. Our previous study indicated that mechanical stretch altered the proliferation of C2C12 myoblasts, associated with insulin growth factor 1 (IGF-1)-mediated phosphoinositide 3-kinase (PI3K)/Akt (also known as protein kinase B) and mitogen-activated protein kinase (MAPK) pathways through IGF-1 receptor (IGF-1R). The purpose of this study was to explore the same stretches on the proliferation of L6 myoblasts and its association with IGF-1-regulated PI3K/Akt and MAPK activations. L6 myoblasts were divided into three groups: control, 15% stretch, and 20% stretch. Stretches were achieved using FlexCell Strain Unit. Cell proliferation and IGF-1 concentration were detected by CCK8 and ELISA, respectively. IGF-1R expression, and expressions and activities of PI3K, Akt, and MAPKs (including extracellular signal-regulated kinases 1 and 2 (ERK1/2) and p38) were determined by Western blot. We found that 15% stretch promoted, while 20% stretch inhibited L6 myoblast proliferation. A 15% stretch increased IGF-1R level, although had no effect on IGF-1 secretion of L6 myoblasts, and PI3K/Akt and ERK1/2 (not p38) inhibitors attenuated 15% stretch-induced pro-proliferation. Exogenous IGF-1 reversed 20% stretch-induced anti-proliferation, accompanied with increases in IGF-1R level as well as PI3K/Akt and MAPK (ERK1/2 and p38) activations. In conclusion, stretch regulated L6 myoblasts proliferation, which may be mediated by the changes in PI3K/Akt and MAPK activations regulated by IGF-1R, despite no detectable IGF-1 from stretched L6 myoblasts.


Asunto(s)
Proliferación Celular , Mecanotransducción Celular , Mioblastos/fisiología , Receptor IGF Tipo 1/metabolismo , Animales , Línea Celular , Sistema de Señalización de MAP Quinasas , Mioblastos/enzimología , Mioblastos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
11.
Am J Physiol Cell Physiol ; 312(6): C689-C696, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28298333

RESUMEN

Mutations in the gene that encodes the principal l-carnitine transporter, OCTN2, can lead to a reduced intracellular l-carnitine pool and the disease Primary Carnitine Deficiency. l-Carnitine supplementation is used therapeutically to increase intracellular l-carnitine. As AMPK and insulin regulate fat metabolism and substrate uptake, we hypothesized that AMPK-activating compounds and insulin would increase l-carnitine uptake in C2C12 myotubes. The cells express all three OCTN transporters at the mRNA level, and immunohistochemistry confirmed expression at the protein level. Contrary to our hypothesis, despite significant activation of PKB and 2DG uptake, insulin did not increase l-carnitine uptake at 100 nM. However, l-carnitine uptake was modestly increased at a dose of 150 nM insulin. A range of AMPK activators that increase intracellular calcium content [caffeine (10 mM, 5 mM, 1 mM, 0.5 mM), A23187 (10 µM)], inhibit mitochondrial function [sodium azide (75 µM), rotenone (1 µM), berberine (100 µM), DNP (500 µM)], or directly activate AMPK [AICAR (250 µM)] were assessed for their ability to regulate l-carnitine uptake. All compounds tested significantly inhibited l-carnitine uptake. Inhibition by caffeine was not dantrolene (10 µM) sensitive despite dantrolene inhibiting caffeine-mediated calcium release. Saturation curve analysis suggested that caffeine did not competitively inhibit l-carnitine transport. To assess the potential role of AMPK in this process, we assessed the ability of the AMPK inhibitor Compound C (10 µM) to rescue the effect of caffeine. Compound C offered a partial rescue of l-carnitine uptake with 0.5 mM caffeine, suggesting that AMPK may play a role in the inhibitory effects of caffeine. However, caffeine likely inhibits l-carnitine uptake by alternative mechanisms independently of calcium release. PKA activation or direct interference with transporter function may play a role.


Asunto(s)
Carnitina/antagonistas & inhibidores , Activadores de Enzimas/farmacología , Mioblastos/efectos de los fármacos , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Berberina/farmacología , Transporte Biológico/efectos de los fármacos , Cafeína/farmacología , Calcimicina/farmacología , Calcio/metabolismo , Carnitina/metabolismo , Línea Celular , Dantroleno/farmacología , Activación Enzimática/efectos de los fármacos , Expresión Génica , Insulina/farmacología , Ratones , Mioblastos/citología , Mioblastos/enzimología , Proteínas de Transporte de Catión Orgánico/genética , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ribonucleótidos/farmacología , Rotenona/farmacología , Azida Sódica/farmacología , Miembro 5 de la Familia 22 de Transportadores de Solutos
12.
Am J Physiol Endocrinol Metab ; 312(6): E471-E481, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28174179

RESUMEN

S100 calcium-binding protein B (S100B), a multifunctional macromolecule mainly expressed in nerve tissues and adipocytes, has been suggested to contribute to the pathogenesis of obesity. To clarify the role of S100B in insulin action and glucose metabolism in peripheral tissues, we investigated the effect of S100B on glycolysis in myoblast and myotube cells. Rat myoblast L6 cells were treated with recombinant mouse S100B to examine glucose consumption, lactate production, glycogen accumulation, glycolytic metabolites and enzyme activity, insulin signaling, and poly(ADP-ribosyl)ation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Glycolytic metabolites were investigated by enzyme assays or metabolome analysis, and insulin signaling was assessed by Western blot analysis. Enzyme activity and poly(ADP-ribosyl)ation of GAPDH was evaluated by an enzyme assay and immunoprecipitation followed by dot blot with an anti-poly(ADP-ribose) antibody, respectively. S100B significantly decreased glucose consumption, glucose analog uptake, and lactate production in L6 cells, in either the presence or absence of insulin. In contrast, S100B had no effect on glycogen accumulation and insulin signaling. Metabolome analysis revealed that S100B increased the concentration of glycolytic intermediates upstream of GAPDH. S100B impaired GAPDH activity and increased poly(ADP-ribosyl)ated GAPDH proteins. The effects of S100B on glucose metabolism were mostly canceled by a poly(ADP-ribose) polymerase inhibitor. Similar results were obtained in C2C12 myotube cells. We conclude that S100B as a humoral factor may impair glycolysis in muscle cells independent of insulin action, and the effect may be attributed to the inhibition of GAPDH activity from enhanced poly(ADP-ribosyl)ation of the enzyme.


Asunto(s)
Gliceraldehído-3-Fosfato Deshidrogenasas/antagonistas & inhibidores , Glucólisis , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Procesamiento Proteico-Postraduccional , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Animales , Línea Celular , Células Cultivadas , Inducción Enzimática/efectos de los fármacos , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Glucólisis/efectos de los fármacos , Hexoquinasa/química , Hexoquinasa/genética , Hexoquinasa/metabolismo , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/enzimología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Mioblastos/efectos de los fármacos , Mioblastos/enzimología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Proteínas Recombinantes/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/genética
13.
Hum Mol Genet ; 24(10): 2873-83, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25701873

RESUMEN

Limb girdle muscular dystrophy 2H is caused by mutations in the gene encoding the E3 ubiquitin ligase, TRIM32. Previously, we generated and characterized a Trim32 knockout mouse (T32KO) that displays both neurogenic and myopathic features. The myopathy in these mice is attributable to impaired muscle growth, associated with satellite cell senescence and premature sarcopenia. This satellite cell senescence is due to accumulation of the SUMO ligase PIASy, a substrate of TRIM32. The goal of this investigation was to identify additional substrates of TRIM32 using 2D fluorescence difference gel electrophoresis (2D-DIGE) in order to further explore its role in skeletal muscle. Because TRIM32 is an E3 ubiquitin ligase, we reasoned that TRIM32's substrates would accumulate in its absence. 2D-DIGE identified 19 proteins that accumulate in muscles from the T32KO mouse. We focused on two of these proteins, NDRG2 and TRIM72, due to their putative roles in myoblast proliferation and myogenesis. Follow-up analysis confirmed that both proteins were ubiquitinated by TRIM32 in vitro; however, only NDRG2 accumulated in skeletal muscle and myoblasts in the absence of TRIM32. NDRG2 overexpression in myoblasts led to reduced cell proliferation and delayed cell cycle withdrawal during differentiation. Thus, we identified NDRG2 as a novel target for TRIM32; these findings further corroborate the hypothesis that TRIM32 is involved in control of myogenic cells proliferation and differentiation.


Asunto(s)
Proliferación Celular , Distrofia Muscular de Cinturas/genética , Mioblastos/enzimología , Proteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Ciclo Celular , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Ratones , Ratones Noqueados , Distrofia Muscular de Cinturas/enzimología , Mioblastos/fisiología , Proteínas/genética , Electroforesis Bidimensional Diferencial en Gel , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba
14.
Apoptosis ; 22(4): 531-543, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28181111

RESUMEN

Reduced Na+-K+-ATPase activity has close relationship with cardiomyocyte death. Reactive oxygen species (ROS) also plays an important role in cardiac cell damage. It has been proved that Na+-K+-ATPase and ROS form a feed-forward amplifier. The aim of this study was to explore whether DRm217, a proved Na+/K+-ATPase's DR-region specific monoclonal antibody and direct activator, could disrupt Na+-K+-ATPase/ROS amplifier and protect cardiac cells from ROS-induced injury. We found that DRm217 protected myocardial cells against hydrogen peroxide (H2O2)-induced cardiac cell injury and mitochondrial dysfunction. DRm217 also alleviated the effect of H2O2 on inhibition of Na+-K+-ATPase activity, Na+-K+-ATPase cell surface expression, and Src phosphorylation. H2O2-treatment increased intracellular ROS, mitochondrial ROS and induced intracellular Ca2+, mitochondrial Ca2+ overload. DRm217 closed Na+-K+-ATPase/ROS amplifier, alleviated Ca2+ accumulation and finally inhibited ROS and mitochondrial ROS generation. These novel results may help us to understand the important role of the Na+-K+-ATPase in oxidative stress and oxidative stress-related disease.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Mioblastos/enzimología , Miocitos Cardíacos/enzimología , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Señalización del Calcio , Línea Celular , Activación Enzimática/inmunología , Humanos , Peróxido de Hidrógeno/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Mioblastos/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Cultivo de Órganos , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/inmunología , Familia-src Quinasas/fisiología
15.
J Cell Sci ; 128(9): 1707-17, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25770104

RESUMEN

Muscle differentiation requires a complex signaling cascade that leads to the production of multinucleated myofibers. Genes regulating the intrinsic mitochondrial apoptotic pathway also function in controlling cell differentiation. How such signaling pathways are regulated during differentiation is not fully understood. Bit-1 (also known as PTRH2) mutations in humans cause infantile-onset multisystem disease with muscle weakness. We demonstrate here that Bit-1 controls skeletal myogenesis through a caspase-mediated signaling pathway. Bit-1-null mice exhibit a myopathy with hypotrophic myofibers. Bit-1-null myoblasts prematurely express muscle-specific proteins. Similarly, knockdown of Bit-1 expression in C2C12 myoblasts promotes early differentiation, whereas overexpression delays differentiation. In wild-type mice, Bit-1 levels increase during differentiation. Bit-1-null myoblasts exhibited increased levels of caspase 9 and caspase 3 without increased apoptosis. Bit-1 re-expression partially rescued differentiation. In Bit-1-null muscle, Bcl-2 levels are reduced, suggesting that Bcl-2-mediated inhibition of caspase 9 and caspase 3 is decreased. Bcl-2 re-expression rescued Bit-1-mediated early differentiation in Bit-1-null myoblasts and C2C12 cells with knockdown of Bit-1 expression. These results support an unanticipated yet essential role for Bit-1 in controlling myogenesis through regulation of Bcl-2.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Diferenciación Celular , Desarrollo de Músculos , Animales , Apoptosis , Hidrolasas de Éster Carboxílico/deficiencia , Caspasa 3/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Fibras Musculares Esqueléticas/patología , Mioblastos/enzimología , Mioblastos/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/metabolismo , Transfección
16.
Cell Tissue Res ; 369(3): 591-602, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28623422

RESUMEN

In Duchenne muscular dystrophy (DMD), lack of dystrophin leads to progressive muscle degeneration, with DMD patients suffering from cardiorespiratory failure. Cell therapy is an alternative to life-long corticoid therapy. Satellite cells, the stem cells of skeletal muscles, do not completely compensate for the muscle damage in dystrophic muscles. Elevated levels of proinflammatory and profibrotic factors, such as metalloproteinase 9 (MMP-9), impair muscle regeneration, leading to extensive fibrosis and poor results with myoblast transplantation therapies. Omega-3 is an anti-inflammatory drug that protects against muscle degeneration in the mdx mouse model of DMD. In the present study, we test our hypothesis that omega-3 affects MMP-9 and thereby benefits muscle regeneration and myoblast transplantation in the mdx mouse. We observe that omega-3 reduces MMP-9 gene expression and improves myoblast engraftment, satellite cell activation, and muscle regeneration by mechanisms involving, at least in part, the regulation of macrophages, as shown here with the fluorescence-activated cell sorting technique. The present study demonstrates the benefits of omega-3 on satellite cell survival and muscle regeneration, further supporting its use in clinical trials and cell therapies in DMD.


Asunto(s)
Distrofina/deficiencia , Ácidos Grasos Omega-3/farmacología , Metaloproteinasa 9 de la Matriz/metabolismo , Fibras Musculares Esqueléticas/patología , Mioblastos/enzimología , Mioblastos/trasplante , Células Satélite del Músculo Esquelético/patología , Animales , Biomarcadores/metabolismo , Distrofina/metabolismo , Femenino , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/genética , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/efectos de los fármacos , Atrofia Muscular/patología , Mioblastos/efectos de los fármacos , Necrosis , Receptores Notch/metabolismo , Regeneración/efectos de los fármacos , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
17.
J Biol Chem ; 290(34): 20774-20781, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26139603

RESUMEN

The vast majority of newly synthesized acetylcholinesterase (AChE) molecules do not assemble into catalytically active oligomeric forms and are rapidly degraded intracellularly by the endoplasmic reticulum-associated protein degradation pathway. We have previously shown that AChE in skeletal muscle is regulated in part post-translationally by the availability of the noncatalytic subunit collagen Q, and others have shown that expression of a 17-amino acid N-terminal proline-rich attachment domain of collagen Q is sufficient to promote AChE tetramerization in cells producing AChE. In this study we show that muscle cells, or cell lines expressing AChE catalytic subunits, incubated with synthetic proline-rich attachment domain peptides containing the endoplasmic reticulum retrieval sequence KDEL take up and retrogradely transport them to the endoplasmic reticulum network where they induce assembly of AChE tetramers. The peptides act to enhance AChE folding thereby rescuing them from reticulum degradation. This enhanced folding efficiency occurs in the presence of inhibitors of protein synthesis and in turn increases total cell-associated AChE activity and active tetramer secretion. Pulse-chase studies of isotopically labeled AChE molecules show that the enzyme is rescued from intracellular degradation. These studies provide a mechanistic explanation for the large scale intracellular degradation of AChE previously observed and indicate that simple peptides alone can increase the production and secretion of this critical synaptic enzyme in muscle tissue.


Asunto(s)
Acetilcolinesterasa/metabolismo , Proteínas Aviares/metabolismo , Dominio Catalítico/genética , Mioblastos/efectos de los fármacos , Péptidos/farmacología , Acetilcolinesterasa/genética , Secuencia de Aminoácidos , Animales , Proteínas Aviares/genética , Células COS , Chlorocebus aethiops , Embrión no Mamífero , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Expresión Génica , Células HEK293 , Humanos , Ratones , Datos de Secuencia Molecular , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Mioblastos/citología , Mioblastos/enzimología , Péptidos/síntesis química , Cultivo Primario de Células , Multimerización de Proteína , Estabilidad Proteica/efectos de los fármacos , Codorniz , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia
18.
Biochem Biophys Res Commun ; 469(4): 1117-22, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26742424

RESUMEN

AMP-activated protein kinase (AMPK) is a serine/threonine kinase that functions as a sensor of intracellular energy. Activation of AMPK is associated with increased phosphorylation of the α-subunit at threonine 172 (T172) and decreased phosphorylation at serine 485 in AMPKα1 and serine 491 in AMPKα2 (S485/491). One potential mediator of AMPK phosphorylation is phosphatidylinositol 3-kinase (PI3K); however, the mechanism and the identities of the specific PI3K isoforms that regulate AMPK activation are not known. To determine whether PI3K p110α regulated AMPK activation in muscle cells, C2C12 myoblasts were subjected to pharmacological inhibition of p110α, siRNA directed against p110α, or overexpression of constitutively-active or dominant negative p110α. Chemical inhibition, siRNA, and expression of dominant-negative p110α were all associated with increased AMPK T172 phosphorylation, whereas expression of constitutively-active p110α reduced T172 phosphorylation. Conversely, pharmacological inhibition of p110α reduced AMPK S485/491 phosphorylation, while constitutively-active p110α increased AMPK S485/491 phosphorylation. This p110α-mediated increase in AMPK S485/491 phosphorylation was eliminated in the presence of the Akt inhibitor MK2206, suggesting that p110α-mediated phosphorylation of AMPKα at S485/491 is Akt-dependent. In response to oligomycin or serum-starvation, AMPK T172 phosphorylation was elevated in p110α-deficient myoblasts compared to control myoblasts. Overall, our findings identify PI3K p110α as a mediator of AMPK phosphorylation in myoblasts.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Mioblastos/enzimología , Estrés Oxidativo/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Línea Celular , Fosfatidilinositol 3-Quinasa Clase I , Ratones , Fosforilación
19.
Biochem Biophys Res Commun ; 469(4): 1049-54, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26740179

RESUMEN

Chronic inflammation augments the deleterious effects of several diseases, particularly diabetes, cancer, and sepsis. It is also involved in the process of metabolic shift from glucose oxidation to lactate production. Although several studies suggest that the change in activity of the pyruvate dehydrogenase complex (PDC) is a major factor causing this metabolic change, the exact mechanism of the inflammatory state remains unclear. In this study, we investigated the effect of lipopolysaccharide (LPS) on the expression of pyruvate dehydrogenase kinase 4 (PDK4), which is strongly associated with inactivation of the PDC in C2C12 myoblasts. In C2C12 myoblasts, LPS exposure led to increased PDK4 mRNA and protein expression levels as well as lactate production in culture medium. However, the expression levels of other PDK isoenzymes (PDK1 - 3) remained unchanged. Additionally, we observed that LPS treatment induced phosphorylation of Jun N-Terminal Kinases (JNK). To confirm the role of JNK, we inhibited the JNK pathway and observed that PDK4 expression and lactate production were decreased, but p38 and ERK were not significantly changed. Taken together, our results suggest that LPS induces PDK4 expression and alters glucose metabolism via the JNK pathway.


Asunto(s)
MAP Quinasa Quinasa 4/metabolismo , Mioblastos/enzimología , Miositis/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Lipopolisacáridos , Ratones , Mioblastos/efectos de los fármacos , Miositis/inducido químicamente , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
20.
Am J Physiol Cell Physiol ; 308(11): C919-31, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810257

RESUMEN

Myoblast fusion is critical for proper muscle growth and regeneration. During myoblast fusion, the localization of some molecules is spatially restricted; however, the exact reason for such localization is unknown. Creatine kinase B (CKB), which replenishes local ATP pools, localizes near the ends of cultured primary mouse myotubes. To gain insights into the function of CKB, we performed a yeast two-hybrid screen to identify CKB-interacting proteins. We identified molecules with a broad diversity of roles, including actin polymerization, intracellular protein trafficking, and alternative splicing, as well as sarcomeric components. In-depth studies of α-skeletal actin and α-cardiac actin, two predominant muscle actin isoforms, demonstrated their biochemical interaction and partial colocalization with CKB near the ends of myotubes in vitro. In contrast to other cell types, specific knockdown of CKB did not grossly affect actin polymerization in myotubes, suggesting other muscle-specific roles for CKB. Interestingly, knockdown of CKB resulted in significantly increased myoblast fusion and myotube size in vitro, whereas knockdown of creatine kinase M had no effect on these myogenic parameters. Our results suggest that localized CKB plays a key role in myotube formation by limiting myoblast fusion during myogenesis.


Asunto(s)
Forma BB de la Creatina-Quinasa/genética , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/enzimología , Mioblastos/enzimología , Actinas/genética , Actinas/metabolismo , Empalme Alternativo , Animales , Fusión Celular , Forma BB de la Creatina-Quinasa/antagonistas & inhibidores , Forma BB de la Creatina-Quinasa/metabolismo , Forma MM de la Creatina-Quinasa/genética , Forma MM de la Creatina-Quinasa/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/citología , Mioblastos/citología , Polimerizacion , Cultivo Primario de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA