Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(45): e2305959120, 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37903280

RESUMEN

TRAAK channels are mechano-gated two-pore-domain K+ channels. Up to now, activity of these channels has been reported in neurons but not in skeletal muscle, yet an archetype of tissue challenged by mechanical stress. Using patch clamp methods on isolated skeletal muscle fibers from adult zebrafish, we show here that single channels sharing properties of TRAAK channels, i.e., selective to K+ ions, of 56 pS unitary conductance in the presence of 5 mM external K+, activated by membrane stretch, heat, arachidonic acid, and internal alkaline pH, are present in enzymatically isolated fast skeletal muscle fibers from adult zebrafish. The kcnk4b transcript encoding for TRAAK channels was cloned and found, concomitantly with activity of mechano-gated K+ channels, to be absent in zebrafish fast skeletal muscles at the larval stage but arising around 1 mo of age. The transfer of the kcnk4b gene in HEK cells and in the adult mouse muscle, that do not express functional TRAAK channels, led to expression and activity of mechano-gated K+ channels displaying properties comparable to native zebrafish TRAAK channels. In whole-cell voltage-clamp and current-clamp conditions, membrane stretch and heat led to activation of macroscopic K+ currents and to acceleration of the repolarization phase of action potentials respectively, suggesting that heat production and membrane deformation associated with skeletal muscle activity can control muscle excitability through TRAAK channel activation. TRAAK channels may represent a teleost-specific evolutionary product contributing to improve swimming performance for escaping predators and capturing prey at a critical stage of development.


Asunto(s)
Calor , Pez Cebra , Animales , Ratones , Chlorocebus aethiops , Pez Cebra/genética , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético , Células COS
2.
Diabetologia ; 63(11): 2471-2481, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32840676

RESUMEN

AIMS/HYPOTHESIS: Disrupted intracellular Ca2+ handling is known to play a role in diabetic cardiomyopathy but it has also been postulated to contribute to obesity- and type 2 diabetes-associated skeletal muscle dysfunction. Still, there is so far very limited functional insight into whether, and if so to what extent, muscular Ca2+ homeostasis is affected in this situation, so as to potentially determine or contribute to muscle weakness. In differentiated muscle, force production is under the control of the excitation-contraction coupling process: upon plasma membrane electrical activity, the CaV1.1 voltage sensor/Ca2+ channel in the plasma membrane triggers opening of the ryanodine receptor Ca2+ release channel in the sarcoplasmic reticulum (SR) membrane. Opening of the ryanodine receptor triggers the rise in cytosolic Ca2+, which activates contraction while Ca2+ uptake by the SR ATPase Ca2+-pump promotes relaxation. These are the core mechanisms underlying the tight control of muscle force by neuronal electrical activity. This study aimed at characterising their inherent physiological function in a diet-induced mouse model of obesity and type 2 diabetes. METHODS: Intact muscle fibres were isolated from mice fed either with a standard chow diet or with a high-fat, high-sucrose diet generating obesity, insulin resistance and glucose intolerance. Properties of muscle fibres were investigated with a combination of whole-cell voltage-clamp electrophysiology and confocal fluorescence imaging. The integrity and density of the plasma membrane network (transverse tubules) that carries the membrane excitation throughout the muscle fibres was assessed with the dye Di-8-ANEPPS. CaV1.1 Ca2+ channel activity was studied by measuring the changes in current across the plasma membrane elicited by voltage-clamp depolarising pulses of increasing amplitude. SR Ca2+ release through ryanodine receptors was simultaneously detected with the Ca2+-sensitive dye Rhod-2 in the cytosol. CaV1.1 voltage-sensing activity was separately characterised from the properties of intra-plasma-membrane charge movement produced by short voltage-clamp depolarising pulses. Spontaneous Ca2+ release at rest was assessed with the Ca2+-sensitive dye Fluo-4. The rate of SR Ca2+ uptake was assessed from the time course of cytosolic Ca2+ recovery after the end of voltage excitation using the Ca2+-sensitive dye Fluo-4FF. The response to a fatigue-stimulation protocol was determined from the time course of decline of the peak Fluo-4FF Ca2+ transients elicited by 30 trains of 5-ms-long depolarising pulses delivered at 100 Hz. RESULTS: The transverse tubule network architecture and density were well preserved in the fibres from the obese mice. The CaV1.1 Ca2+ current and voltage-sensing properties were also largely unaffected with mean values for maximum conductance and maximum amount of charge of 234 ± 12 S/F and 30.7 ± 1.6 nC/µF compared with 196 ± 13 S/F and 32.9 ± 2.0 nC/µF in fibres from mice fed with the standard diet, respectively. Voltage-activated SR Ca2+ release through ryanodine receptors also exhibited very similar properties in the two groups with mean values for maximum rate of Ca2+ release of 76.0 ± 6.5 and 78.1 ± 4.4 µmol l-1 ms-1, in fibres from control and obese mice, respectively. The response to a fatigue protocol was also largely unaffected in fibres from the obese mice, and so were the rate of cytosolic Ca2+ removal and the spontaneous Ca2+ release activity at rest. CONCLUSIONS/INTERPRETATION: The functional properties of the main mechanisms involved in the control of muscle Ca2+ homeostasis are well preserved in muscle fibres from obese mice, at the level of both the plasma membrane and of the SR. We conclude that intracellular Ca2+ handling and excitation-contraction coupling in skeletal muscle fibres are not primary targets of obesity and type 2 diabetes. Graphical abstract.


Asunto(s)
Calcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Músculo Esquelético/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Señalización del Calcio/fisiología , Membrana Celular/metabolismo , Ratones , Ratones Obesos
3.
Proc Natl Acad Sci U S A ; 113(50): 14432-14437, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911767

RESUMEN

Mutations in the gene encoding the phosphoinositide 3-phosphatase myotubularin (MTM1) are responsible for a pediatric disease of skeletal muscle named myotubular myopathy (XLMTM). Muscle fibers from MTM1-deficient mice present defects in excitation-contraction (EC) coupling likely responsible for the disease-associated fatal muscle weakness. However, the mechanism leading to EC coupling failure remains unclear. During normal skeletal muscle EC coupling, transverse (t) tubule depolarization triggers sarcoplasmic reticulum (SR) Ca2+ release through ryanodine receptor channels gated by conformational coupling with the t-tubule voltage-sensing dihydropyridine receptors. We report that MTM1 deficiency is associated with a 60% depression of global SR Ca2+ release over the full range of voltage sensitivity of EC coupling. SR Ca2+ release in the diseased fibers is also slower than in normal fibers, or delayed following voltage activation, consistent with the contribution of Ca2+-gated ryanodine receptors to EC coupling. In addition, we found that SR Ca2+ release is spatially heterogeneous within myotubularin-deficient muscle fibers, with focally defective areas recapitulating the global alterations. Importantly, we found that pharmacological inhibition of phosphatidylinositol 3-kinase (PtdIns 3-kinase) activity rescues the Ca2+ release defects in isolated muscle fibers and increases the lifespan and mobility of XLMTM mice, providing proof of concept for the use of PtdIns 3-kinase inhibitors in myotubular myopathy and suggesting that unbalanced PtdIns 3-kinase activity plays a critical role in the pathological process.


Asunto(s)
Señalización del Calcio/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia , Androstadienos/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Acoplamiento Excitación-Contracción/efectos de los fármacos , Acoplamiento Excitación-Contracción/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/fisiología , Miopatías Estructurales Congénitas/tratamiento farmacológico , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/fisiopatología , Técnicas de Placa-Clamp , Proteínas Tirosina Fosfatasas no Receptoras/genética , Wortmanina
4.
Am J Physiol Cell Physiol ; 315(5): C776-C779, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30281323

RESUMEN

High metabolic activity and existence of a large transmembrane inward electrochemical gradient for H+ at rest promote intracellular acidification of skeletal muscle. Exchangers and cotransports efficiently contend against accumulation of intracellular H+ and associated deleterious effects on muscle functions. Voltage-gated H+ channels have also been found to represent another H+ extrusion pathway in cultured muscle cells. Up to now, the skeletal muscle cell was therefore the unique vertebrate excitable cell in which voltage-gated H+ currents have been described. In this study, we show that, unlike cultured cells, single mouse muscle fibers do not generate H+ currents in response to depolarization. In contrast, expression of human voltage-gated H+ channels in mouse muscle gives rise to robust outward voltage-gated H+ currents. This result excludes that inappropriate experimental conditions may have failed to reveal voltage-gated H+ currents in control muscle. This work therefore demonstrates that fully differentiated mammalian muscle fibers do not express functional voltage-gated H+ channels and consequently can no longer be considered as the only vertebrate excitable cells exhibiting voltage-gated H+ currents.


Asunto(s)
Canales Iónicos/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Activación del Canal Iónico/genética , Ratones , Músculo Esquelético/citología , Fármacos Neuromusculares Despolarizantes/farmacología , Técnicas de Placa-Clamp
5.
J Physiol ; 596(11): 2019-2027, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29572832

RESUMEN

Patients suffering from type 1 hypokalaemic periodic paralysis (HypoPP1) experience attacks of muscle paralysis associated with hypokalaemia. The disease arises from missense mutations in the gene encoding the α1 subunit of the dihydropyridine receptor (DHPR), a protein complex anchored in the tubular membrane of skeletal muscle fibres which controls the release of Ca2+ from sarcoplasmic reticulum and also functions as a Ca2+ channel. The vast majority of mutations consist of the replacement of one of the outer arginines in S4 segments of the α1 subunit by neutral residues. Early studies have shown that muscle fibres from HypoPP1 patients are abnormally depolarized at rest in low K+ to the point of inducing muscle inexcitability. The relationship between HypoPP1 mutations and depolarization has long remained unknown. More recent investigations conducted in the closely structurally related voltage-gated Na+ and K+ channels have shown that comparable S4 arginine substitutions gave rise to elevated inward currents at negative potentials called gating pore currents. Experiments performed in muscle fibres from different models revealed such an inward resting current through HypoPP1 mutated Ca2+ channels. In mouse fibres transfected with HypoPP1 mutated channels, the elevated resting current was found to carry H+ for the R1239H arginine-to-histidine mutation in a S4 segment and Na+ for the V876E HypoPP1 mutation, which has the peculiarity of not being located in S4 segments. Muscle paralysis probably results from the presence of a gating pore current associated with hypokalaemia for both mutations, possibly aggravated by external acidosis for the R1239H mutation.


Asunto(s)
Canales de Calcio/fisiología , Cationes Monovalentes/metabolismo , Parálisis Periódica Hipopotasémica/fisiopatología , Activación del Canal Iónico , Músculo Esquelético/fisiología , Animales , Humanos
6.
Cell Tissue Res ; 371(2): 309-323, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29018970

RESUMEN

Andersen's syndrome (AS) is a rare autosomal disorder that has been defined by the triad of periodic paralysis, cardiac arrhythmia, and developmental anomalies. AS has been directly linked to over 40 different autosomal dominant negative loss-of-function mutations in the KCNJ2 gene, encoding for the tetrameric strong inward rectifying K+ channel KIR2.1. While KIR2.1 channels have been suggested to contribute to setting the resting membrane potential (RMP) and to control the duration of the action potential (AP) in skeletal and cardiac muscle, the mechanism by which AS mutations produce such complex pathophysiological symptoms is poorly understood. Thus, we use an adenoviral transduction strategy to study in vivo subcellular distribution of wild-type (WT) and AS-associated mutant KIR2.1 channels in mouse skeletal muscle. We determined that WT and D71V AS mutant KIR2.1 channels are localized to the sarcolemma and the transverse tubules (T-tubules) of skeletal muscle fibers, while the ∆314-315 AS KIR2.1 mutation prevents proper trafficking of the homo- or hetero-meric channel complexes. Whole-cell voltage-clamp recordings in individual skeletal muscle fibers confirmed the reduction of inwardly rectifying K+ current (IK1) after transduction with ∆314-315 KIR2.1 as compared to WT channels. Analysis of skeletal muscle function revealed reduced force generation during isometric contraction as well as reduced resistance to muscle fatigue in extensor digitorum longus muscles transduced with AS mutant KIR2.1. Together, these results suggest that KIR2.1 channels may be involved in the excitation-contraction coupling process required for proper skeletal muscle function. Our findings provide clues to mechanisms associated with periodic paralysis in AS.


Asunto(s)
Síndrome de Andersen/genética , Técnicas de Silenciamiento del Gen , Músculo Esquelético/patología , Mutación/genética , Canales de Potasio de Rectificación Interna/genética , Adenoviridae/metabolismo , Síndrome de Andersen/patología , Síndrome de Andersen/fisiopatología , Animales , Células COS , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Activación del Canal Iónico , Contracción Isométrica , Ratones , Fatiga Muscular , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/fisiopatología
7.
J Physiol ; 595(20): 6417-6428, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28857175

RESUMEN

KEY POINTS: Missense mutations in the gene encoding the α1 subunit of the skeletal muscle voltage-gated Ca2+ channel induce type 1 hypokalaemic periodic paralysis, a poorly understood neuromuscular disease characterized by episodic attacks of paralysis associated with low serum K+ . Acute expression of human wild-type and R1239H HypoPP1 mutant α1 subunits in mature mouse muscles showed that R1239H fibres displayed Ca2+ currents of reduced amplitude and larger resting leak inward current increased by external acidification. External acidification also produced intracellular acidification at a higher rate in R1239H fibres and inhibited inward rectifier K+ currents. These data suggest that the R1239H mutation induces an elevated leak H+ current at rest flowing through a gating pore and could explain why paralytic attacks preferentially occur during the recovery period following muscle exercise. ABSTRACT: Missense mutations in the gene encoding the α1 subunit of the skeletal muscle voltage-gated Ca2+ channel induce type 1 hypokalaemic periodic paralysis, a poorly understood neuromuscular disease characterized by episodic attacks of paralysis associated with low serum K+ . The present study aimed at identifying the changes in muscle fibre electrical properties induced by acute expression of the R1239H hypokalaemic periodic paralysis human mutant α1 subunit of Ca2+ channels in a mature muscle environment to better understand the pathophysiological mechanisms involved in this disorder. We transferred genes encoding wild-type and R1239H mutant human Ca2+ channels into hindlimb mouse muscle by electroporation and combined voltage-clamp and intracellular pH measurements on enzymatically dissociated single muscle fibres. As compared to fibres expressing wild-type α1 subunits, R1239H mutant-expressing fibres displayed Ca2+ currents of reduced amplitude and a higher resting leak inward current that was increased by external acidification. External acidification also produced intracellular acidification at a higher rate in R1239H fibres and inhibited inward rectifier K+ currents. These data indicate that the R1239H mutation induces an elevated leak H+ current at rest flowing through a gating pore created by the mutation and that external acidification favours onset of muscle paralysis by potentiating H+ depolarizing currents and inhibiting resting inward rectifier K+ currents. Our results could thus explain why paralytic attacks preferentially occur during the recovery period following intense muscle exercise.


Asunto(s)
Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/fisiología , Parálisis Periódica Hipopotasémica , Fibras Musculares Esqueléticas/fisiología , Animales , Concentración de Iones de Hidrógeno , Masculino , Ratones , Mutación Missense , Técnicas de Placa-Clamp
8.
J Physiol ; 595(24): 7369-7382, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29071728

RESUMEN

KEY POINTS: Dynamin 2 is a ubiquitously expressed protein involved in membrane trafficking processes. Mutations in the gene encoding dynamin 2 are responsible for a congenital myopathy associated with centrally located nuclei in the muscle fibres. Using muscle fibres from a mouse model of the most common mutation responsible for this disease in humans, we tested whether altered Ca2+ signalling and excitation-contraction coupling contribute to muscle weakness. The plasma membrane network that carries the electrical excitation is moderately perturbed in the diseased muscle fibres. The excitation-activated Ca2+ input fluxes across both the plasma membrane and the membrane of the sarcoplasmic reticulum are defective in the diseased fibres, which probably contributes to muscle weakness in patients. ABSTRACT: Mutations in the gene encoding dynamin 2 (DNM2) are responsible for autosomal dominant centronuclear myopathy (AD-CNM). We studied the functional properties of Ca2+ signalling and excitation-contraction (EC) coupling in muscle fibres isolated from a knock-in (KI) mouse model of the disease, using confocal imaging and the voltage clamp technique. The transverse-tubule network organization appeared to be unaltered in the diseased fibres, although its density was reduced by ∼10% compared to that in control fibres. The density of Ca2+ current through CaV1.1 channels and the rate of voltage-activated sarcoplasmic reticulum Ca2+ release were reduced by ∼60% and 30%, respectively, in KI vs. control fibres. In addition, Ca2+ release in the KI fibres reached its peak value 10-50 ms later than in control ones. Activation of Ca2+ transients along the longitudinal axis of the fibres was more heterogeneous in the KI than in the control fibres, with the difference being exacerbated at intermediate membrane voltages. KI fibres exhibited spontaneous Ca2+ release events that were almost absent from control fibres. Overall, the results of the present study demonstrate that Ca2+ signalling and EC coupling exhibit a number of dysfunctions likely contributing to muscle weakness in DNM2-related AD-CNM.


Asunto(s)
Dinamina II/genética , Acoplamiento Excitación-Contracción , Fibras Musculares Esqueléticas/metabolismo , Miopatías Estructurales Congénitas/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Células Cultivadas , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/fisiología , Mutación Missense , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/fisiopatología
9.
Development ; 140(22): 4602-13, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24131632

RESUMEN

The myotendinous junction (MTJ) is the major site of force transfer in skeletal muscle, and defects in its structure correlate with a subset of muscular dystrophies. Col22a1 encodes the MTJ component collagen XXII, the function of which remains unknown. Here, we have cloned and characterized the zebrafish col22a1 gene and conducted morpholino-based loss-of-function studies in developing embryos. We showed that col22a1 transcripts localize at muscle ends when the MTJ forms and that COLXXII protein integrates the junctional extracellular matrix. Knockdown of COLXXII expression resulted in muscular dystrophy-like phenotype, including swimming impairment, curvature of embryo trunk/tail, strong reduction of twitch-contraction amplitude and contraction-induced muscle fiber detachment, and provoked significant activation of the survival factor Akt. Electron microscopy and immunofluorescence studies revealed that absence of COLXXII caused a strong reduction of MTJ folds and defects in myoseptal structure. These defects resulted in reduced contractile force and susceptibility of junctional extracellular matrix to rupture when subjected to repeated mechanical stress. Co-injection of sub-phenotypic doses of morpholinos against col22a1 and genes of the major muscle linkage systems showed a synergistic gene interaction between col22a1 and itga7 (α7ß1 integrin) that was not observed with dag1 (dystroglycan). Finally, pertinent to a conserved role in humans, the dystrophic phenotype was rescued by microinjection of recombinant human COLXXII. Our findings indicate that COLXXII contributes to the stabilization of myotendinous junctions and strengthens skeletal muscle attachments during contractile activity.


Asunto(s)
Colágeno/genética , Técnicas de Silenciamiento del Gen , Distrofia Muscular Animal/patología , Tendones/patología , Pez Cebra/genética , Animales , Supervivencia Celular/efectos de los fármacos , Colágeno/metabolismo , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Embrión no Mamífero/ultraestructura , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Integrinas/metabolismo , Mamíferos , Microinyecciones , Morfolinos/farmacología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Debilidad Muscular/metabolismo , Debilidad Muscular/patología , Distrofia Muscular Animal/embriología , Distrofia Muscular Animal/genética , Fenotipo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Tendones/efectos de los fármacos , Tendones/metabolismo , Tendones/ultraestructura
10.
J Physiol ; 593(21): 4781-97, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26383921

RESUMEN

Muscle contraction is triggered by Ca(2+) ions released from the sarcoplasmic reticulum (SR) in response to depolarization of skeletal muscle fibres. Muscle activation is also associated with a voltage-activated trans-sarcolemmal Ca(2+) influx early identified as a current flowing through L-type Ca(2+) channels. Because removal of external Ca(2+) does not impede fibres from contracting, a negligible role was given to this voltage-activated Ca(2+) entry, although the decline of Ca(2+) release is more pronounced in the absence of Ca(2+) during long-lasting activation. Furthermore, it is not clearly established whether Ca(2+) exclusively flows through L-type channels or in addition through a parallel voltage-activated pathway distinct from L-type channels. Here, by monitoring the quenching of fura-2 fluorescence resulting from Mn(2+) influx in voltage-controlled mouse and zebrafish isolated muscle fibres, we show that the L-type current is the only contributor to Ca(2+) influx during long-lasting depolarizations in skeletal muscle. Calibration of the Mn(2+) quenching signal allowed us to estimate a mean Mn(2+) current of 0.31 ± 0.06 A F(-1) flowing through L-type channels during a train of action potentials. Measurements of SR Ca(2+) changes with fluo-5N in response to depolarization revealed that an elevated voltage-activated Ca(2+) current potentiated SR Ca(2+) loading and addition of external Mn(2+) produced quenching of fluo-5N in the SR, indicating that voltage-activated Ca(2+) /Mn(2+) influx contributes to SR Ca(2+) /Mn(2+) loading.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Músculo Esquelético/metabolismo , Retículo Sarcoplasmático/metabolismo , Potenciales de Acción , Animales , Calcio/metabolismo , Células Cultivadas , Manganeso/metabolismo , Contracción Muscular , Músculo Esquelético/fisiología , Pez Cebra
11.
J Physiol ; 593(17): 3849-63, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26108786

RESUMEN

Increased plasma osmolarity induces intracellular water depletion and cell shrinkage (CS) followed by activation of a regulatory volume increase (RVI). In skeletal muscle, the hyperosmotic shock-induced CS is accompanied by a small membrane depolarization responsible for a release of Ca(2+) from intracellular pools. Hyperosmotic shock also induces phosphorylation of STE20/SPS1-related proline/alanine-rich kinase (SPAK). TRPV2 dominant negative expressing fibres challenged with hyperosmotic shock present a slower membrane depolarization, a diminished Ca(2+) response, a smaller RVI response, a decrease in SPAK phosphorylation and defective muscle function. We suggest that hyperosmotic shock induces TRPV2 activation, which accelerates muscle cell depolarization and allows the subsequent Ca(2+) release from the sarcoplasmic reticulum, activation of the Na(+) -K(+) -Cl(-) cotransporter by SPAK, and the RVI response. Increased plasma osmolarity induces intracellular water depletion and cell shrinkage followed by activation of a regulatory volume increase (RVI). In skeletal muscle, this is accompanied by transverse tubule (TT) dilatation and by a membrane depolarization responsible for a release of Ca(2+) from intracellular pools. We observed that both hyperosmotic shock-induced Ca(2+) transients and RVI were inhibited by Gd(3+) , ruthenium red and GsMTx4 toxin, three inhibitors of mechanosensitive ion channels. The response was also completely absent in muscle fibres overexpressing a non-permeant, dominant negative (DN) mutant of the transient receptor potential, V2 isoform (TRPV2) ion channel, suggesting the involvement of TRPV2 or of a TRP isoform susceptible to heterotetramerization with TRPV2. The release of Ca(2+) induced by hyperosmotic shock was increased by cannabidiol, an activator of TRPV2, and decreased by tranilast, an inhibitor of TRPV2, suggesting a role for the TRPV2 channel itself. Hyperosmotic shock-induced membrane depolarization was impaired in TRPV2-DN fibres, suggesting that TRPV2 activation triggers the release of Ca(2+) from the sarcoplasmic reticulum by depolarizing TTs. RVI requires the sequential activation of STE20/SPS1-related proline/alanine-rich kinase (SPAK) and NKCC1, a Na(+) -K(+) -Cl(-) cotransporter, allowing ion entry and driving osmotic water flow. In fibres overexpressing TRPV2-DN as well as in fibres in which Ca(2+) transients were abolished by the Ca(2+) chelator BAPTA, the level of P-SPAK(Ser373) in response to hyperosmotic shock was reduced, suggesting a modulation of SPAK phosphorylation by intracellular Ca(2+) . We conclude that TRPV2 is involved in osmosensation in skeletal muscle fibres, acting in concert with P-SPAK-activated NKCC1.


Asunto(s)
Canales de Calcio/fisiología , Fibras Musculares Esqueléticas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Miembro 2 de la Familia de Transportadores de Soluto 12/fisiología , Canales Catiónicos TRPV/fisiología , Animales , Calcio , Tamaño de la Célula , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Concentración Osmolar , Presión Osmótica , Fosforilación
12.
J Gen Physiol ; 155(4)2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36820799

RESUMEN

Tight control of skeletal muscle contractile activation is secured by the excitation-contraction (EC) coupling protein complex, a molecular machinery allowing the plasma membrane voltage to control the activity of the ryanodine receptor Ca2+ release channel in the sarcoplasmic reticulum (SR) membrane. This machinery has been shown to be intimately linked to the plasma membrane protein pannexin-1 (Panx1). We investigated whether the prescription drug probenecid, a widely used Panx1 blocker, affects Ca2+ signaling, EC coupling, and muscle force. The effect of probenecid was tested on membrane current, resting Ca2+, and SR Ca2+ release in isolated mouse muscle fibers, using a combination of whole-cell voltage-clamp and Ca2+ imaging, and on electrically triggered contraction of isolated muscles. Probenecid (1 mM) induces SR Ca2+ leak at rest and reduces peak voltage-activated SR Ca2+ release and contractile force by 40%. Carbenoxolone, another Panx1 blocker, also reduces Ca2+ release, but neither a Panx1 channel inhibitory peptide nor a purinergic antagonist affected Ca2+ release, suggesting that probenecid and carbenoxolone do not act through inhibition of Panx1-mediated ATP release and consequently altered purinergic signaling. Probenecid may act by altering Panx1 interaction with the EC coupling machinery, yet the implication of another molecular target cannot be excluded. Since probenecid has been used both in the clinic and as a masking agent for doping in sports, these results should encourage evaluation of possible effects on muscle function in treated individuals. In addition, they also raise the question of whether probenecid-induced altered Ca2+ homeostasis may be shared by other tissues.


Asunto(s)
Calcio , Probenecid , Ratones , Animales , Probenecid/metabolismo , Probenecid/farmacología , Calcio/metabolismo , Carbenoxolona/metabolismo , Carbenoxolona/farmacología , Fibras Musculares Esqueléticas/metabolismo , Contracción Muscular , Músculo Esquelético/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Conexinas/metabolismo
13.
J Physiol ; 590(23): 6027-36, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23006480

RESUMEN

Contraction of skeletal muscle is triggered by the release of Ca(2+) from the sarcoplasmic reticulum (SR) in response to depolarization of the muscle membrane. Depolarization is known to elicit a conformational change of the dihydropyridine receptor (DHPR) in the tubular membrane that controls in a time- and voltage-dependent manner the opening of the ryanodine receptor (RyR), the SR Ca(2+) release channel. At rest, it is assumed that RyRs are kept in a closed state imposed by the repressive action of DHPRs; however, a direct control of the RyR gating by the DHPR has up to now never been demonstrated in resting adult muscle. In this study, we monitored slow changes in SR Ca(2+) content using the Ca(2+) indicator fluo-5N loaded in the SR of voltage-clamped mouse muscle fibres. We first show that external Ca(2+) removal induced a reversible SR Ca(2+) efflux at -80 mV and prevented SR Ca(2+) refilling following depolarization-evoked SR Ca(2+) depletion. The dihydropyridine compound nifedipine induced similar effects. The rate of SR Ca(2+) efflux was also shown to be controlled in a time- and voltage-dependent manner within a membrane potential range more negative than -50 mV. Finally, intracellular addition of ryanodine produced an irreversible SR Ca(2+) efflux and kept the SR in a highly depleted state following depolarization-evoked SR Ca(2+) depletion. The fact that resting SR Ca(2+) efflux is modulated by conformational changes of DHPRs induced by external Ca(2+), nifedipine and voltage demonstrates that DHPRs exert an active control on gating of RyRs in resting skeletal muscle.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Fibras Musculares Esqueléticas/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Animales , Calcio/fisiología , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Retículo Sarcoplasmático/fisiología
14.
Proc Natl Acad Sci U S A ; 106(44): 18763-8, 2009 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-19846786

RESUMEN

Skeletal muscle contraction is triggered by the excitation-contraction (E-C) coupling machinery residing at the triad, a membrane structure formed by the juxtaposition of T-tubules and sarcoplasmic reticulum (SR) cisternae. The formation and maintenance of this structure is key for muscle function but is not well characterized. We have investigated the mechanisms leading to X-linked myotubular myopathy (XLMTM), a severe congenital disorder due to loss of function mutations in the MTM1 gene, encoding myotubularin, a phosphoinositide phosphatase thought to have a role in plasma membrane homeostasis and endocytosis. Using a mouse model of the disease, we report that Mtm1-deficient muscle fibers have a decreased number of triads and abnormal longitudinally oriented T-tubules. In addition, SR Ca(2+) release elicited by voltage-clamp depolarizations is strongly depressed in myotubularin-deficient muscle fibers, with myoplasmic Ca(2+) removal and SR Ca(2+) content essentially unaffected. At the molecular level, Mtm1-deficient myofibers exhibit a 3-fold reduction in type 1 ryanodine receptor (RyR1) protein level. These data reveal a critical role of myotubularin in the proper organization and function of the E-C coupling machinery and strongly suggest that defective RyR1-mediated SR Ca(2+) release is responsible for the failure of muscle function in myotubular myopathy.


Asunto(s)
Metabolismo de los Lípidos , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia , Retículo Sarcoplasmático/enzimología , Retículo Sarcoplasmático/patología , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Regulación de la Expresión Génica , Homeostasis/genética , Activación del Canal Iónico , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/ultraestructura , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Retículo Sarcoplasmático/ultraestructura
15.
J Gen Physiol ; 154(9)2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-35767225

RESUMEN

The zebrafish has emerged as a very relevant animal model for probing the pathophysiology of human skeletal muscle disorders. This vertebrate animal model displays a startle response characterized by high-frequency swimming activity powered by contraction of fast skeletal muscle fibers excited at extremely high frequencies, critical for escaping predators and capturing prey. Such intense muscle performance requires extremely fast properties of the contractile machinery but also of excitation-contraction coupling, the process by which an action potential spreading along the sarcolemma induces a change in configuration of the dihydropyridine receptors, resulting in intramembrane charge movements, which in turn triggers the release of Ca2+ from the sarcoplasmic reticulum. However, thus far, the fastest Ca2+ transients evoked by vertebrate muscle fibers has been described in muscles used to produce sounds, such as those in the toadfish swim bladder, but not in muscles used for locomotion. By performing intracellular Ca2+ measurements under voltage control in isolated fast skeletal muscle fibers from adult zebrafish and mouse, we demonstrate that fish fast muscle fibers display superfast kinetics of action potentials, intramembrane charge movements, and action potential-evoked Ca2+ transient, allowing fusion and fused sustained Ca2+ transients at frequencies of excitation much higher than in mouse fast skeletal muscle fibers and comparable to those recorded in muscles producing sounds. The present study is the first demonstration of superfast kinetics of excitation-contraction coupling in skeletal muscle allowing superfast locomotor behaviors in a vertebrate.


Asunto(s)
Calcio , Pez Cebra , Animales , Ratones , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/fisiología , Retículo Sarcoplasmático
16.
Matrix Biol ; 109: 1-18, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35278627

RESUMEN

The myotendinous junction (MTJ) is essential for the integrity of the musculoskeletal unit. Here, we show that gene ablation of the MTJ marker col22a1 in zebrafish results in MTJ dysfunction but with variable degrees of expression and distinct phenotypic classes. While most individuals reach adulthood with no overt muscle phenotype (class 1), a subset of the progeny displays severe movement impairment and die before metamorphosis (class 2). Yet all mutants display muscle weakness due to ineffective muscle force transmission that is ultimately detrimental for class-specific locomotion-related functions. Movement impairment at the critical stage of swimming postural learning causes class 2 larval death by compromising food intake. In class 1 adults, intensive exercise is required to uncover a decline in muscle performance, accompanied by higher energy demand and mitochondrial adaptation. This study underscores COL22A1 as a candidate gene for myopathies associated with dysfunctional force transmission and anticipates a phenotypically heterogeneous disease.


Asunto(s)
Tendones , Pez Cebra , Animales , Locomoción , Músculo Esquelético , Fenotipo , Postura , Pez Cebra/genética
17.
J Gen Physiol ; 153(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33538764

RESUMEN

In intact muscle fibers, functional properties of ryanodine receptor (RYR)-mediated sarcoplasmic reticulum (SR) Ca2+ release triggered by activation of the voltage sensor CaV1.1 have so far essentially been addressed with diffusible Ca2+-sensitive dyes. Here, we used a domain (T306) of the protein triadin to target the Ca2+-sensitive probe GCaMP6f to the junctional SR membrane, in the immediate vicinity of RYR channels, within the triad region. Fluorescence of untargeted GCaMP6f was distributed throughout the muscle fibers and experienced large Ca2+-dependent changes, with obvious kinetic delays, upon application of voltage-clamp depolarizing pulses. Conversely, T306-GCaMP6f localized to the triad and generated Ca2+-dependent fluorescence transients of lower amplitude and faster kinetics for low and intermediate levels of Ca2+ release than those of untargeted GCaMP6f. By contrast, model simulation of the spatial gradients of Ca2+ following Ca2+ release predicted limited kinetic differences under the assumptions that the two probes were present at the same concentration and suffered from identical kinetic limitations. At the spatial level, T306-GCaMP6f transients within distinct regions of a same fiber yielded a uniform time course, even at low levels of Ca2+ release activation. Similar observations were made using GCaMP6f fused to the γ1 auxiliary subunit of CaV1.1. Despite the probe's limitations, our results point out the remarkable synchronicity of voltage-dependent Ca2+ release activation and termination among individual triads and highlight the potential of the approach to visualize activation or closure of single groups of RYR channels. We anticipate targeting of improved Ca2+ sensors to the triad will provide illuminating insights into physiological normal RYR function and its dysfunction under stress or pathological conditions.


Asunto(s)
Calcio , Canal Liberador de Calcio Receptor de Rianodina , Calcio/metabolismo , Señalización del Calcio , Colorantes/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
18.
J Gen Physiol ; 153(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34636893

RESUMEN

One of the most important functions of skeletal muscle is to respond to nerve stimuli by contracting. This function ensures body movement but also participates in other important physiological roles, like regulation of glucose homeostasis. Muscle activity is closely regulated to adapt to different demands and shows a plasticity that relies on both transcriptional activity and nerve stimuli. These two processes, both dependent on depolarization of the plasma membrane, have so far been regarded as separated and independent processes due to a lack of evidence of common protein partners or molecular mechanisms. In this study, we reveal intimate functional interactions between the process of excitation-induced contraction and the process of excitation-induced transcriptional activity in skeletal muscle. We show that the plasma membrane voltage-sensing protein CaV1.1 and the ATP-releasing channel Pannexin-1 (Panx1) regulate each other in a reciprocal manner, playing roles in both processes. Specifically, knockdown of CaV1.1 produces chronically elevated extracellular ATP concentrations at rest, consistent with disruption of the normal control of Panx1 activity. Conversely, knockdown of Panx1 affects not only activation of transcription but also CaV1.1 function on the control of muscle fiber contraction. Altogether, our results establish the presence of bidirectional functional regulations between the molecular machineries involved in the control of contraction and transcription induced by membrane depolarization of adult muscle fibers. Our results are important for an integrative understanding of skeletal muscle function and may impact our understanding of several neuromuscular diseases.


Asunto(s)
Canales de Calcio Tipo L , Acoplamiento Excitación-Contracción , Canales de Calcio Tipo L/metabolismo , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo
19.
J Neuromuscul Dis ; 8(s2): S243-S255, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34633328

RESUMEN

Mutations in the Anoctamin 5 (Ano5) gene that result in the lack of expression or function of ANO5 protein, cause Limb Girdle Muscular Dystrophy (LGMD) 2L/R12, and Miyoshi Muscular Dystrophy (MMD3). However, the dystrophic phenotype observed in patient muscles is not uniformly recapitulated by ANO5 knockout in animal models of LGMD2L. Here we describe the generation of a mouse model of LGMD2L generated by targeted out-of-frame deletion of the Ano5 gene. This model shows progressive muscle loss, increased muscle weakness, and persistent bouts of myofiber regeneration without chronic muscle inflammation, which recapitulates the mild to moderate skeletal muscle dystrophy reported in the LGMD2L patients. We show that these features of ANO5 deficient muscle are not associated with a change in the calcium-activated sarcolemmal chloride channel activity or compromised in vivo regenerative myogenesis. Use of this mouse model allows conducting in vivo investigations into the functional role of ANO5 in muscle health and for preclinical therapeutic development for LGMD2L.


Asunto(s)
Anoctaminas/genética , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/genética , Animales , Canales de Cloruro/genética , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Debilidad Muscular/genética , Distrofia Muscular de Cinturas/patología , Mutación , Fenotipo
20.
J Physiol ; 588(Pt 15): 2945-60, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20547679

RESUMEN

A number of G-protein-coupled receptors are expressed in skeletal muscle but their roles in muscle physiology and downstream effector systems remain poorly investigated. Here we explored the functional importance of the G-protein betagamma (Gbetagamma) signalling pathway on voltage-controlled Ca(2+) homeostasis in single isolated adult skeletal muscle fibres. A GFP-tagged Gbeta(1)gamma(2) dimer was expressed in vivo in mice muscle fibres. The GFP fluorescence pattern was consistent with a Gbeta(1)gamma(2) dimer localization in the transverse-tubule membrane. Membrane current and indo-1 fluorescence measurements performed under voltage-clamp conditions reveal a drastic reduction of both L-type Ca(2+) current density and of peak amplitude of the voltage-activated Ca(2+) transient in Gbeta(1)gamma(2)-expressing fibres. These effects were not observed upon expression of Gbeta(2)gamma(2), Gbeta(3)gamma(2) or Gbeta(4)gamma(2). Our data suggest that the G-protein beta(1)gamma(2) dimer may play an important regulatory role in skeletal muscle excitation-contraction coupling.


Asunto(s)
Potenciales de Acción/fisiología , Canales de Calcio Tipo L/fisiología , Calcio/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Activación del Canal Iónico/fisiología , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/fisiología , Animales , Células Cultivadas , Dimerización , Expresión Génica/fisiología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA