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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Appl Physiol ; 124(3): 681-751, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38206444

RESUMEN

This historical review traces key discoveries regarding K+ and Na+ ions in skeletal muscle at rest and with exercise, including contents and concentrations, Na+,K+-ATPase (NKA) and exercise effects on plasma [K+] in humans. Following initial measures in 1896 of muscle contents in various species, including humans, electrical stimulation of animal muscle showed K+ loss and gains in Na+, Cl- and H20, then subsequently bidirectional muscle K+ and Na+ fluxes. After NKA discovery in 1957, methods were developed to quantify muscle NKA activity via rates of ATP hydrolysis, Na+/K+ radioisotope fluxes, [3H]-ouabain binding and phosphatase activity. Since then, it became clear that NKA plays a central role in Na+/K+ homeostasis and that NKA content and activity are regulated by muscle contractions and numerous hormones. During intense exercise in humans, muscle intracellular [K+] falls by 21 mM (range - 13 to - 39 mM), interstitial [K+] increases to 12-13 mM, and plasma [K+] rises to 6-8 mM, whilst post-exercise plasma [K+] falls rapidly, reflecting increased muscle NKA activity. Contractions were shown to increase NKA activity in proportion to activation frequency in animal intact muscle preparations. In human muscle, [3H]-ouabain-binding content fully quantifies NKA content, whilst the method mainly detects α2 isoforms in rats. Acute or chronic exercise affects human muscle K+, NKA content, activity, isoforms and phospholemman (FXYD1). Numerous hormones, pharmacological and dietary interventions, altered acid-base or redox states, exercise training and physical inactivity modulate plasma [K+] during exercise. Finally, historical research approaches largely excluded female participants and typically used very small sample sizes.


Asunto(s)
Ouabaína , ATPasa Intercambiadora de Sodio-Potasio , Humanos , Ratas , Animales , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Ouabaína/metabolismo , Músculo Esquelético/metabolismo , Contracción Muscular , Hormonas/metabolismo , Isoformas de Proteínas/metabolismo , Iones/metabolismo
2.
J Physiol ; 601(24): 5669-5687, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37934587

RESUMEN

A reduced muscle glycogen content and potassium (K+ ) disturbances across muscle membranes occur concomitantly during repeated intense exercise and together may contribute to skeletal muscle fatigue. Therefore, we examined whether raised extracellular K+ concentration ([K+ ]o ) (4 to 11 mM) interacts with lowered glycogen to reduce force production. Isometric contractions were evoked in isolated mouse soleus muscles (37°C) using direct supramaximal field stimulation. (1) Glycogen declined markedly in non-fatigued muscle with >2 h exposure in glucose-free physiological saline compared with control solutions (11 mM glucose), i.e. to <45% control. (2) Severe glycogen depletion was associated with increased 5'-AMP-activated protein kinase activity, indicative of metabolic stress. (3) The decline of peak tetanic force at 11 mM [K+ ]o was exacerbated from 67% initial at normal glycogen to 22% initial at lowered glycogen. This was due to a higher percentage of inexcitable fibres (71% vs. 43%), yet without greater sarcolemmal depolarisation or smaller amplitude action potentials. (4) Returning glucose while at 11 mM [K+ ]o increased both glycogen and force. (5) Exposure to 4 mM [K+ ]o glucose-free solutions (15 min) did not increase fatiguability during repeated tetani; however, after recovery there was a greater force decline at 11 mM [K+ ]o at lower than normal glycogen. (6) An important exponential relationship was established between relative peak tetanic force at 11 mM [K+ ]o and muscle glycogen content. These findings provide direct evidence of a synergistic interaction between raised [K+ ]o and lowered muscle glycogen as the latter shifts the peak tetanic force-resting EM relationship towards more negative resting EM due to lowered sarcolemmal excitability, which hence may contribute to muscle fatigue. KEY POINTS: Diminished muscle glycogen levels and raised extracellular potassium concentrations ([K+ ]o ) occur simultaneously during intense exercise and together may contribute to muscle fatigue. Prolonged exposure of isolated non-fatigued soleus muscles of mice to glucose-free physiological saline solutions markedly lowered muscle glycogen levels, as does fatigue then recovery in glucose-free solutions. For both approaches, the subsequent decline of maximal force at 11 mM [K+ ]o , which mimics interstitial [K+ ] levels during intense exercise, was exacerbated at lowered compared with normal glycogen. This was mainly due to many more muscle fibres becoming inexcitable. We established an important relationship that provides evidence of a synergistic interaction between raised [K+ ]o and lowered glycogen content to reduce force production. This paper indicates that partially lowered muscle glycogen (and/or metabolic stress) together with elevated interstitial [K+ ] interactively lowers muscle force, and hence may diminish performance especially during repeated high-intensity exercise.


Asunto(s)
Glucógeno , Contracción Muscular , Ratones , Animales , Contracción Muscular/fisiología , Potasio/metabolismo , Músculo Esquelético/fisiología , Fatiga Muscular/fisiología , Glucosa/farmacología
3.
Eur J Appl Physiol ; 123(11): 2345-2378, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37584745

RESUMEN

Perturbations in K+ have long been considered a key factor in skeletal muscle fatigue. However, the exercise-induced changes in K+ intra-to-extracellular gradient is by itself insufficiently large to be a major cause for the force decrease during fatigue unless combined to other ion gradient changes such as for Na+. Whilst several studies described K+-induced force depression at high extracellular [K+] ([K+]e), others reported that small increases in [K+]e induced potentiation during submaximal activation frequencies, a finding that has mostly been ignored. There is evidence for decreased Cl- ClC-1 channel activity at muscle activity onset, which may limit K+-induced force depression, and large increases in ClC-1 channel activity during metabolic stress that may enhance K+ induced force depression. The ATP-sensitive K+ channel (KATP channel) is also activated during metabolic stress to lower sarcolemmal excitability. Taking into account all these findings, we propose a revised concept in which K+ has two physiological roles: (1) K+-induced potentiation and (2) K+-induced force depression. During low-moderate intensity muscle contractions, the K+-induced force depression associated with increased [K+]e is prevented by concomitant decreased ClC-1 channel activity, allowing K+-induced potentiation of sub-maximal tetanic contractions to dominate, thereby optimizing muscle performance. When ATP demand exceeds supply, creating metabolic stress, both KATP and ClC-1 channels are activated. KATP channels contribute to force reductions by lowering sarcolemmal generation of action potentials, whilst ClC-1 channel enhances the force-depressing effects of K+, thereby triggering fatigue. The ultimate function of these changes is to preserve the remaining ATP to prevent damaging ATP depletion.


Asunto(s)
Fatiga Muscular , Músculo Esquelético , Humanos , Músculo Esquelético/fisiología , Fatiga Muscular/fisiología , Contracción Muscular/fisiología , Potenciales de Acción/fisiología , Iones/metabolismo , Adenosina Trifosfato/metabolismo
4.
Am J Physiol Cell Physiol ; 322(6): C1151-C1165, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35385328

RESUMEN

We endeavored to understand the factors determining the peak force-resting membrane potential (EM) relationships of isolated slow-twitch soleus and fast-twitch extensor digitorum longus (EDL) muscles from mice (25°C), especially in relation to fatigue. Interrelationships between intracellular K+ activity ([Formula: see text]), extracellular K+ concentration ([K+]o), resting EM, action potentials, and force were studied. The large resting EM variation was mainly due to the variability of [Formula: see text]. Action potential overshoot-resting EM relationships determined at 4 and 8-10 mM [K+]o after short (<5 min) and prolonged (>50 min) depolarization periods revealed a constant overshoot from -90 to -70 mV providing a safety margin. Overshoot decline with depolarization beyond -70 mV was less after short than prolonged depolarization. Inexcitable fibers occurred only with prolonged depolarization. The overshoot decline during action potential trains (2 s) exceeded that during short depolarizations. Concomitant lower extracellular [Na+] and raised [K+]o depressed the overshoot in an additive manner and peak force in a synergistic manner. Raised [K+]o-induced force loss was exacerbated with transverse wire versus parallel plate stimulation in soleus, implicating action potential propagation failure in the surface membrane. Increasing stimulus pulse parameters restored tetanic force at 9-10 mM [K+]o in soleus but not EDL, indicative of action potential failure within trains. The peak tetanic force-resting EM relationships (determined with resting EM from deeper rather than surface fibers) were dynamic and showed pronounced force depression over -69 to -60 mV in both muscle types, implicating that such depolarization contributes to fatigue. The K+-Na+ interaction shifted this relationship toward less depolarized potentials, suggesting that the combined ionic effect is physiologically important during fatigue.


Asunto(s)
Contracción Muscular , Potasio , Animales , Fatiga , Potenciales de la Membrana/fisiología , Ratones , Contracción Muscular/fisiología , Fatiga Muscular/fisiología , Fibras Musculares de Contracción Lenta/fisiología , Músculo Esquelético/fisiología , Sodio
5.
Am J Physiol Cell Physiol ; 323(6): C1681-C1696, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36280388

RESUMEN

Recent studies reported that in skeletal muscle angiotensin 1-7 (Ang 1-7), via its receptor Mas (MasR), prevents the atrophy induced by angiotensin II and by cast immobilization; it also improves muscle integrity and function in the mdx mouse, a muscular dystrophy model. The objectives of this study were to document 1) the extent of the Ang 1-7's hypertrophic effect in terms of muscle mass and muscle fiber cross-sectional area (CSA), 2) how Ang 1-7 affects muscle contractile function in terms of twitch and tetanic force, force-frequency relationship, and 3) whether the effect involves MasR. Wild-type and MasR-deficient [Mas receptor knockout mouse model (MasR-/-)] mice were treated with Ang 1-7 (100 ng/kg body wt·min using an osmotic pump) for 4 or 16 wk. Ang 1-7 significantly increased skeletal muscle/body weight ratio of soleus, tibialis, and gastrocnemius, but not of extensor digitorum longus (EDL). It significantly increased fiber cross-sectional area in the order of type I > IIA > IIB. In EDL and soleus muscles, it significantly increased twitch and tetanic force while causing a shift in the force-frequency relationship toward lower stimulation frequencies. It had no effect on fiber type composition. None of the Ang 1-7 effects observed in wild-type mice were observed in MasR-/- muscles. It caused a transient increase in phosphorylated protein kinase B (Akt) and 4EBP proteins while having no effect on S6 phosphorylation, MuRF-1, and atrogin-1 and a decrease in PAX7 expression in satellite cells. This is the first study demonstrating the hypertrophic effects of Ang 1-7 in normal muscle acting via its MasR.


Asunto(s)
Angiotensina I , Fragmentos de Péptidos , Ratones , Animales , Ratones Endogámicos mdx , Angiotensina I/farmacología , Angiotensina I/metabolismo , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/metabolismo , Músculo Esquelético/metabolismo
6.
Hum Mol Genet ; 26(10): 1821-1838, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28369467

RESUMEN

Converging lines of evidence have now highlighted the key role for post-transcriptional regulation in the neuromuscular system. In particular, several RNA-binding proteins are known to be misregulated in neuromuscular disorders including myotonic dystrophy type 1, spinal muscular atrophy and amyotrophic lateral sclerosis. In this study, we focused on the RNA-binding protein Staufen1, which assumes multiple functions in both skeletal muscle and neurons. Given our previous work that showed a marked increase in Staufen1 expression in various physiological and pathological conditions including denervated muscle, in embryonic and undifferentiated skeletal muscle, in rhabdomyosarcomas as well as in myotonic dystrophy type 1 muscle samples from both mouse models and humans, we investigated the impact of sustained Staufen1 expression in postnatal skeletal muscle. To this end, we generated a skeletal muscle-specific transgenic mouse model using the muscle creatine kinase promoter to drive tissue-specific expression of Staufen1. We report that sustained Staufen1 expression in postnatal skeletal muscle causes a myopathy characterized by significant morphological and functional deficits. These deficits are accompanied by a marked increase in the expression of several atrophy-associated genes and by the negative regulation of PI3K/AKT signaling. We also uncovered that Staufen1 mediates PTEN expression through indirect transcriptional and direct post-transcriptional events thereby providing the first evidence for Staufen1-regulated PTEN expression. Collectively, our data demonstrate that Staufen1 is a novel atrophy-associated gene, and highlight its potential as a biomarker and therapeutic target for neuromuscular disorders and conditions.


Asunto(s)
Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Expresión Génica , Ratones , Ratones Noqueados , Desnervación Muscular , Músculo Esquelético/metabolismo , Músculos/metabolismo , Atrofia Muscular/metabolismo , Atrofia Muscular Espinal/metabolismo , Distrofia Miotónica/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética , ARN/metabolismo , Procesamiento Postranscripcional del ARN , Transducción de Señal , Tensinas
7.
Hum Mol Genet ; 25(1): 24-43, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26494902

RESUMEN

Upregulation of utrophin A is an attractive therapeutic strategy for treating Duchenne muscular dystrophy (DMD). Over the years, several studies revealed that utrophin A is regulated by multiple transcriptional and post-transcriptional mechanisms, and that pharmacological modulation of these pathways stimulates utrophin A expression in dystrophic muscle. In particular, we recently showed that activation of p38 signaling causes an increase in the levels of utrophin A mRNAs and protein by decreasing the functional availability of the destabilizing RNA-binding protein called K-homology splicing regulatory protein, thereby resulting in increases in the stability of existing mRNAs. Here, we treated 6-week-old mdx mice for 4 weeks with the clinically used anticoagulant drug heparin known to activate p38 mitogen-activated protein kinase, and determined the impact of this pharmacological intervention on the dystrophic phenotype. Our results show that heparin treatment of mdx mice caused a significant ∼1.5- to 3-fold increase in utrophin A expression in diaphragm, extensor digitorum longus and tibialis anterior (TA) muscles. In agreement with these findings, heparin-treated diaphragm and TA muscle fibers showed an accumulation of utrophin A and ß-dystroglycan along their sarcolemma and displayed improved morphology and structural integrity. Moreover, combinatorial drug treatment using both heparin and 5-amino-4-imidazolecarboxamide riboside (AICAR), the latter targeting 5' adenosine monophosphate-activated protein kinase and the transcriptional activation of utrophin A, caused an additive effect on utrophin A expression in dystrophic muscle. These findings establish that heparin is a relevant therapeutic agent for treating DMD, and illustrate that combinatorial treatment of heparin with AICAR may serve as an effective strategy to further increase utrophin A expression in dystrophic muscle via activation of distinct signaling pathways.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Heparina/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Ribonucleótidos/uso terapéutico , Utrofina/biosíntesis , Aminoimidazol Carboxamida/uso terapéutico , Animales , Línea Celular , Quimioterapia Combinada , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Utrofina/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Hum Mol Genet ; 24(5): 1243-55, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25324540

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by the absence of dystrophin along muscle fibers. An attractive therapeutic avenue for DMD consists in the upregulation of utrophin A, a protein with high sequence identity and functional redundancy with dystrophin. Recent work has shown that pharmacological interventions that induce a muscle fiber shift toward a slower, more oxidative phenotype with increased expression of utrophin A confer morphological and functional improvements in mdx mice. Whether such improvements result from the increased expression of utrophin A per se or are linked to other beneficial adaptations associated with the slow, oxidative phenotype remain to be established. To address this central issue, we capitalized on the use of double knockout (dKO) mice, which are mdx mice also deficient in utrophin. We first compared expression of signaling molecules and markers of the slow, oxidative phenotype in muscles of mdx versus dKO mice and found that both strains exhibit similar phenotypes. Chronic activation of 5' adenosine monophosphate-activated protein kinase with 5-amino-4-imidazolecarboxamide riboside (AICAR) resulted in expression of a slower, more oxidative phenotype in both mdx and dKO mice. In mdx mice, this fiber type shift was accompanied by clear functional improvements that included reductions in central nucleation, IgM sarcoplasmic penetration and sarcolemmal damage resulting from eccentric contractions, as well as in increased grip strength. These important morphological and functional adaptations were not seen in AICAR-treated dKO mice. Our findings show the central role of utrophin A in mediating the functional benefits associated with expression of a slower, more oxidative phenotype in dystrophic animals.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Miofibrillas/fisiología , Utrofina/genética , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Animales , Modelos Animales de Enfermedad , Distroglicanos/genética , Distroglicanos/metabolismo , Femenino , Técnicas de Genotipaje , Fuerza de la Mano/fisiología , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular de Duchenne/genética , Fenotipo , Ribonucleósidos/metabolismo , Utrofina/metabolismo
9.
Am J Physiol Cell Physiol ; 311(4): C559-C571, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27488667

RESUMEN

The skeletal muscle ATP-sensitive K+ (KATP) channel is crucial in preventing fiber damage and contractile dysfunction, possibly by preventing damaging ATP depletion. The objective of this study was to investigate changes in energy metabolism during fatigue in wild-type and inwardly rectifying K+ channel (Kir6.2)-deficient (Kir6.2-/-) flexor digitorum brevis (FDB), a muscle that lacks functional KATP channels. Fatigue was elicited with one tetanic contraction every second. Decreases in ATP and total adenylate levels were significantly greater in wild-type than Kir6.2-/- FDB during the last 2 min of the fatigue period. Glycogen depletion was greater in Kir6.2-/- FDB for the first 60 s, but not by the end of the fatigue period, while there was no difference in glucose uptake. The total amount of glucosyl units entering glycolysis was the same in wild-type and Kir6.2-/- FDB. During the first 60 s, Kir6.2-/- FDB generated less lactate and more CO2; in the last 120 s, Kir6.2-/- FDB stopped generating CO2 and produced more lactate. The ATP generated during fatigue from phosphocreatine, glycolysis (lactate), and oxidative phosphorylation (CO2) was 3.3-fold greater in Kir6.2-/- than wild-type FDB. Because ATP and total adenylate were significantly less in Kir6.2-/- FDB, it is suggested that Kir6.2-/- FDB has a greater energy deficit, despite a greater ATP production, which is further supported by greater glucose uptake and lactate and CO2 production in Kir6.2-/- FDB during the recovery period. It is thus concluded that a lack of functional KATP channels results in an impairment of energy metabolism.


Asunto(s)
Metabolismo Energético/fisiología , Canales KATP/deficiencia , Canales KATP/metabolismo , Fatiga Muscular/fisiología , Músculo Esquelético/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Dióxido de Carbono/metabolismo , Glucólisis/fisiología , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/fisiología , Fosforilación Oxidativa , Canales de Potasio de Rectificación Interna/metabolismo
10.
Mol Ther ; 23(6): 1003-1021, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25762009

RESUMEN

The development of therapeutic strategies for skeletal muscle diseases, such as physical injuries and myopathies, depends on the knowledge of regulatory signals that control the myogenic process. The obestatin/GPR39 system operates as an autocrine signal in the regulation of skeletal myogenesis. Using a mouse model of skeletal muscle regeneration after injury and several cellular strategies, we explored the potential use of obestatin as a therapeutic agent for the treatment of trauma-induced muscle injuries. Our results evidenced that the overexpression of the preproghrelin, and thus obestatin, and GPR39 in skeletal muscle increased regeneration after muscle injury. More importantly, the intramuscular injection of obestatin significantly enhanced muscle regeneration by simulating satellite stem cell expansion as well as myofiber hypertrophy through a kinase hierarchy. Added to the myogenic action, the obestatin administration resulted in an increased expression of vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor 2 (VEGFR2) and the consequent microvascularization, with no effect on collagen deposition in skeletal muscle. Furthermore, the potential inhibition of myostatin during obestatin treatment might contribute to its myogenic action improving muscle growth and regeneration. Overall, our data demonstrate successful improvement of muscle regeneration, indicating obestatin is a potential therapeutic agent for skeletal muscle injury and would benefit other myopathies related to muscle regeneration.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Ghrelina/farmacología , Fibras Musculares Esqueléticas/efectos de los fármacos , Enfermedades Musculares/tratamiento farmacológico , Regeneración/efectos de los fármacos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Inyecciones Intramusculares , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/genética , Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Hum Mol Genet ; 22(5): 867-78, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23184147

RESUMEN

The cellular inhibitor of apoptosis 1 (cIAP1) protein is an essential regulator of canonical and noncanonical nuclear factor κB (NF-κB) signaling pathways. NF-κB signaling is known to play important roles in myogenesis and degenerative muscle disorders such as Duchenne muscular dystrophy (DMD), but the involvement of cIAP1 in muscle disease has not been studied directly. Here, we asked whether the loss of cIAP1 would influence the pathology of skeletal muscle in the mdx mouse model of DMD. Double-mutant cIAP1(-/-);mdx mice exhibited reduced muscle damage and decreased fiber centronucleation in the soleus, compared with single-mutant cIAP1(+/+);mdx mice. This improvement in pathology was associated with a reduction in muscle infiltration by macrophages and diminished expression of inflammatory cytokines such as IL-6 and tumor necrosis factor-α. Furthermore, the cIAP1(-/-);mdx mice exhibited reduced serum creatine kinase, and improved exercise endurance associated with improved exercise resilience by the diaphragm. Mechanistically, the loss of cIAP1 was sufficient to drive constitutive activation of the noncanonical NF-κB pathway, which led to increased myoblast fusion in vitro and in vivo. Collectively, these results show that the loss of cIAP1 protects skeletal muscle from the degenerative pathology resulting from systemic loss of dystrophin.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , FN-kappa B/metabolismo , Animales , Creatina Quinasa/sangre , Diafragma/metabolismo , Diafragma/fisiopatología , Distrofina/genética , Distrofina/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos mdx , Desarrollo de Músculos/genética , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , FN-kappa B/genética , Resistencia Física/genética , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/metabolismo
13.
Am J Physiol Regul Integr Comp Physiol ; 308(6): R467-79, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25568074

RESUMEN

The objective of this study was to optimize the approach to obtain viable single flexor digitorum brevis (FDB) fibers following a collagenase digestion. A first aim was to determine the culture medium conditions for the collagenase digestion. The MEM yielded better fibers in terms of morphology and contractility than the DMEM. The addition of FBS to culture media was crucial to prevent fiber supercontraction. The addition of FBS to the physiological solution used during an experiment was also beneficial, especially during fatigue. Optimum FBS concentration in MEM was 10% (vol/vol), and for the physiological solution, it ranged between 0.2 and 1.0%. A second aim was to document the stability of single FDB fibers. If tested the day of the preparation, most fibers (∼80%) had stable contractions for up to 3 h, normal stimulus duration strength to elicit contractions, and normal and stable resting membrane potential during prolonged microelectrode penetration. A third aim was to document their fatigue kinetics. Major differences in fatigue resistance were observed between fibers as expected from the FDB fiber-type composition. All sarcoplasmic [Ca(2+)] and sarcomere length parameters returned to their prefatigue levels after a short recovery. The pCa-sarcomere shortening relationship of unfatigued fibers is very similar to the pCa-force curve reported in other studies. The pCa-sarcomere shortening from fatigue data is complicated by large decreases in sarcomere length between contractions. It is concluded that isolation of single fibers by a collagenase digestion is a viable preparation to study contractility and fatigue kinetics.


Asunto(s)
Calcio/metabolismo , Separación Celular/métodos , Colagenasas/metabolismo , Contracción Muscular , Fatiga Muscular , Fibras Musculares Esqueléticas/fisiología , Sarcómeros/fisiología , Animales , Forma de la Célula , Medios de Cultivo/metabolismo , Estimulación Eléctrica , Cinética , Potenciales de la Membrana , Ratones , Fibras Musculares Esqueléticas/metabolismo , Sarcómeros/metabolismo , Retículo Sarcoplasmático/metabolismo , Suero/metabolismo
14.
Proc Natl Acad Sci U S A ; 109(50): 20614-9, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23185011

RESUMEN

Duchenne muscular dystrophy (DMD) is a devastating genetic muscular disorder of childhood marked by progressive debilitating muscle weakness and wasting, and ultimately death in the second or third decade of life. Wnt7a signaling through its receptor Fzd7 accelerates and augments regeneration by stimulating satellite stem cell expansion through the planar cell polarity pathway, as well as myofiber hypertrophy through the AKT/mammalian target of rapamycin (mTOR) anabolic pathway. We investigated the therapeutic potential of the secreted factor Wnt7a for focal treatment of dystrophic DMD muscles using the mdx mouse model, and found that Wnt7a treatment efficiently induced satellite cell expansion and myofiber hypertrophy in treated mucles in mdx mice. Importantly, Wnt7a treatment resulted in a significant increase in muscle strength, as determined by generation of specific force. Furthermore, Wnt7a reduced the level of contractile damage, likely by inducing a shift in fiber type toward slow-twitch. Finally, we found that Wnt7a similarly induced myotube hypertrophy and a shift in fiber type toward slow-twitch in human primary myotubes. Taken together, our findings suggest that Wnt7a is a promising candidate for development as an ameliorative treatment for DMD.


Asunto(s)
Distrofia Muscular Animal/tratamiento farmacológico , Proteínas Wnt/uso terapéutico , Animales , Electroquimioterapia , Técnicas de Silenciamiento del Gen , Terapia Genética , Humanos , Factores de Transcripción MEF2 , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Contracción Muscular/fisiología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Distrofia Muscular Animal/fisiopatología , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Plásmidos/administración & dosificación , Plásmidos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regeneración/fisiología , Transducción de Señal , Proteínas Wnt/genética , Proteínas Wnt/fisiología
15.
Physiol Genomics ; 46(11): 385-97, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24714718

RESUMEN

Hyperkalemic periodic paralysis (HyperKPP) is characterized by myotonic discharges that occur between episodic attacks of paralysis. Individuals with HyperKPP rarely suffer respiratory distress even though diaphragm muscle expresses the same defective Na(+) channel isoform (NaV1.4) that causes symptoms in limb muscles. We tested the hypothesis that the extent of the HyperKPP phenotype (low force generation and shift toward oxidative type I and IIA fibers) in muscle is a function of 1) the NaV1.4 channel content and 2) the Na(+) influx through the defective channels [i.e., the tetrodotoxin (TTX)-sensitive Na(+) influx]. We measured NaV1.4 channel protein content, TTX-sensitive Na(+) influx, force generation, and myosin isoform expression in four muscles from knock-in mice expressing a NaV1.4 isoform corresponding to the human M1592V mutant. The HyperKPP flexor digitorum brevis muscle showed no contractile abnormalities, which correlated well with its low NaV1.4 protein content and by far the lowest TTX-sensitive Na(+) influx. In contrast, diaphragm muscle expressing the HyperKPP mutant contained high levels of NaV1.4 protein and exhibited a TTX-sensitive Na(+) influx that was 22% higher compared with affected extensor digitorum longus (EDL) and soleus muscles. Surprisingly, despite this high burden of Na(+) influx, the contractility phenotype was very mild in mutant diaphragm compared with the robust abnormalities observed in EDL and soleus. This study provides evidence that HyperKPP phenotype does not depend solely on the NaV1.4 content or Na(+) influx and that the diaphragm does not depend solely on Na(+)-K(+) pumps to ameliorate the phenotype.


Asunto(s)
Contracción Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Mutación/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética , Parálisis Periódica Hiperpotasémica/genética , Sodio/metabolismo , Animales , Humanos , Ratones , Miosinas/genética , Miosinas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.4/metabolismo , Parálisis Periódica Hiperpotasémica/metabolismo , Potasio/metabolismo
16.
J Cell Sci ; 125(Pt 9): 2105-14, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22627029

RESUMEN

ATP provides the energy in our muscles to generate force, through its use by myosin ATPases, and helps to terminate contraction by pumping Ca(2+) back into the sarcoplasmic reticulum, achieved by Ca(2+) ATPase. The capacity to use ATP through these mechanisms is sufficiently high enough so that muscles could quickly deplete ATP. However, this potentially catastrophic depletion is avoided. It has been proposed that ATP is preserved not only by the control of metabolic pathways providing ATP but also by the regulation of the processes that use ATP. Considering that contraction (i.e. myosin ATPase activity) is triggered by release of Ca(2+), the use of ATP can be attenuated by decreasing Ca(2+) release within each cell. A lower level of Ca(2+) release can be accomplished by control of membrane potential and by direct regulation of the ryanodine receptor (RyR, the Ca(2+) release channel in the terminal cisternae). These highly redundant control mechanisms provide an effective means by which ATP can be preserved at the cellular level, avoiding metabolic catastrophe. This Commentary will review some of the known mechanisms by which this regulation of Ca(2+) release and contractile response is achieved, demonstrating that skeletal muscle fatigue is a consequence of attenuation of contractile activation; a process that allows avoidance of metabolic catastrophe.


Asunto(s)
Calcio/metabolismo , Acoplamiento Excitación-Contracción/fisiología , Fatiga Muscular/fisiología , Músculo Esquelético/fisiología , Miosinas/metabolismo , Actinas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calmodulina , Humanos , Canales Iónicos/metabolismo , Potenciales de la Membrana/fisiología , Fosforilación , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/fisiología
17.
FASEB J ; 27(10): 4213-25, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23825224

RESUMEN

Exercise substantially improves metabolic health, making the elicited mechanisms important targets for novel therapeutic strategies. Uncoupling protein 3 (UCP3) is a mitochondrial inner membrane protein highly selectively expressed in skeletal muscle. Here we report that moderate UCP3 overexpression (roughly 3-fold) in muscles of UCP3 transgenic (UCP3 Tg) mice acts as an exercise mimetic in many ways. UCP3 overexpression increased spontaneous activity (∼40%) and energy expenditure (∼5-10%) and decreased oxidative stress (∼15-20%), similar to exercise training in wild-type (WT) mice. The increase in complete fatty acid oxidation (FAO; ∼30% for WT and ∼70% for UCP3 Tg) and energy expenditure (∼8% for WT and 15% for UCP3 Tg) in response to endurance training was higher in UCP3 Tg than in WT mice, showing an additive effect of UCP3 and endurance training on these two parameters. Moreover, increases in circulating short-chain acylcarnitines in response to acute exercise in untrained WT mice were absent with training or in UCP3 Tg mice. UCP3 overexpression had the same effect as training in decreasing long-chain acylcarnitines. Outcomes coincided with a reduction in muscle carnitine acetyltransferase activity that catalyzes the formation of acylcarnitines. Overall, results are consistent with the conclusions that circulating acylcarnitines could be used as a marker of incomplete muscle FAO and that UCP3 is a potential target for the treatment of prevalent metabolic diseases in which muscle FAO is affected.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Resistencia Física , Animales , Biomarcadores , Ingestión de Alimentos , Metabolismo Energético , Canales Iónicos/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/genética , Músculo Esquelético/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Condicionamiento Físico Animal , Proteína Desacopladora 3
18.
Skelet Muscle ; 14(1): 8, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38671506

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is associated with impaired muscle regeneration, progressive muscle weakness, damage, and wasting. While the cause of DMD is an X-linked loss of function mutation in the gene encoding dystrophin, the exact mechanisms that perpetuate the disease progression are unknown. Our laboratory has demonstrated that pannexin 1 (Panx1 in rodents; PANX1 in humans) is critical for the development, strength, and regeneration of male skeletal muscle. In normal skeletal muscle, Panx1 is part of a multiprotein complex with dystrophin. We and others have previously shown that Panx1 levels and channel activity are dysregulated in various mouse models of DMD. METHODS: We utilized myoblast cell lines derived from DMD patients to assess PANX1 expression and function. To investigate how Panx1 dysregulation contributes to DMD, we generated a dystrophic (mdx) mouse model that lacks Panx1 (Panx1-/-/mdx). In depth characterization of this model included histological analysis, as well as locomotor, and physiological tests such as muscle force and grip strength assessments. RESULTS: Here, we demonstrate that PANX1 levels and channel function are reduced in patient-derived DMD myoblast cell lines. Panx1-/-/mdx mice have a significantly reduced lifespan, and decreased body weight due to lean mass loss. Their tibialis anterior were more affected than their soleus muscles and displayed reduced mass, myofiber loss, increased centrally nucleated myofibers, and a lower number of muscle stem cells compared to that of Panx1+/+/mdx mice. These detrimental effects were associated with muscle and locomotor functional impairments. In vitro, PANX1 overexpression in patient-derived DMD myoblasts improved their differentiation and fusion. CONCLUSIONS: Collectively, our findings suggest that PANX1/Panx1 dysregulation in DMD exacerbates several aspects of the disease. Moreover, our results suggest a potential therapeutic benefit to increasing PANX1 levels in dystrophic muscles.


Asunto(s)
Conexinas , Ratones Endogámicos mdx , Músculo Esquelético , Distrofia Muscular de Duchenne , Proteínas del Tejido Nervioso , Animales , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Conexinas/genética , Conexinas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Humanos , Ratones , Mioblastos/metabolismo , Línea Celular , Fuerza Muscular , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Noqueados
19.
Hum Mol Genet ; 20(17): 3478-93, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21659335

RESUMEN

A therapeutic approach for Duchenne muscular dystrophy (DMD) is to up-regulate utrophin in skeletal muscle in an effort to compensate for the lack of dystrophin. We previously hypothesized that promotion of the slow, oxidative myogenic program, which triggers utrophin up-regulation, can attenuate the dystrophic pathology in mdx animals. Since treatment of healthy mice with the AMP-activated protein kinase (AMPK) activator 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR) enhances oxidative capacity and elicits a fast-to-slow fiber-type transition, we evaluated the effects of chronic AMPK stimulation on skeletal muscle phenotype and utrophin expression in mdx mice. Daily AICAR administration (500 mg/kg/day, 30 days) of 5-7-week-old mdx animals induced an elevation in mitochondrial cytochrome c oxidase enzyme activity, an increase in myosin heavy-chain type IIa-positive fibers and slower twitch contraction kinetics in the fast, glycolytic extensor digitorum longus muscle. Utrophin expression was significantly enhanced in response to AICAR, which occurred coincident with an elevated ß-dystroglycan expression along the sarcolemma. These adaptations were associated with an increase in sarcolemmal structural integrity under basal conditions, as well as during damaging eccentric contractions ex vivo. Notably, peroxisome proliferator-activated receptor γ co-activator-1α (PGC-1α) and silent information regulator two ortholog 1 protein contents were significantly higher in muscle from mdx mice compared with wild-type littermates and AICAR further increased PGC-1α expression. Our data show that AICAR-evoked muscle plasticity results in beneficial phenotypic adaptations in mdx mice and suggest that the contextually novel application of this compound for muscular dystrophy warrants further study.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Ribonucleótidos/farmacología , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular , Distroglicanos/genética , Distroglicanos/metabolismo , Ratones , Ratones Endogámicos mdx , PPAR gamma/genética , PPAR gamma/metabolismo , Sarcolema/genética , Sarcolema/metabolismo
20.
Am J Physiol Cell Physiol ; 302(1): C110-21, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21940670

RESUMEN

In the present study, we evaluated how a pharmacologically induced phenotype shift in dystrophic skeletal muscle would affect subsequent intracellular signaling in response to a complementary, adaptive physiological stimulus. mdx mice were treated with the AMP-activated protein kinase (AMPK) activator 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR; 500 mg·kg(-1)·day(-1)) for 30 days, and then one-half of the animals were subjected to a bout of treadmill running to induce acute AMPK and p38 MAPK signaling. The mRNA levels of phenotypic modifiers, including peroxisome proliferator-activated receptor-δ (PPARδ), PPARγ coactivator-1α (PGC-1α), receptor interacting protein 140 (RIP 140), and silent information regulator two ortholog 1 (SIRT1) were assessed in skeletal muscle, as well as the expression of the protein arginine methyltransferase genes PRMT1 and CARM1. We found unique AMPK and p38 phosphorylation and expression signatures between dystrophic and healthy muscle. In dystrophic skeletal muscle, treadmill running induced PPARδ, PGC-1α, and SIRT1 mRNAs, three molecules that promote the slow, oxidative myogenic program. In the mdx animals that received the chronic AICAR treatment, running-elicited AMPK and p38 phosphorylation was attenuated compared with vehicle-treated mice. Similarly, acute stress-evoked expression of PPARδ, PGC-1α, and SIRT1 was also blunted by chronic pharmacological AMPK stimulation. Skeletal muscle PRMT1 and CARM1 protein contents were higher in mdx mice compared with wild-type littermates. The acute running-evoked induction of PRMT1 and CARM1 mRNAs was also attenuated by the AICAR treatment. Our data demonstrate that prior pharmacological conditioning is a salient determinant in how dystrophic muscle adapts to subsequent complementary, acute physiological stress stimuli. These results provide insight into possible therapeutic applications of synthetic agonists in neuromuscular diseases, such as during chronic administration to Duchenne muscular dystrophy patients.


Asunto(s)
Proteínas Quinasas Activadas por AMP/administración & dosificación , Adaptación Fisiológica/fisiología , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/fisiopatología , Transducción de Señal/fisiología , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Condicionamiento Físico Animal/métodos , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA