Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(51): 20831-6, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24302765

RESUMEN

Dysferlinopathies, most commonly limb girdle muscular dystrophy 2B and Miyoshi myopathy, are degenerative myopathies caused by mutations in the DYSF gene encoding the protein dysferlin. Studies of dysferlin have focused on its role in the repair of the sarcolemma of skeletal muscle, but dysferlin's association with calcium (Ca(2+)) signaling proteins in the transverse (t-) tubules suggests additional roles. Here, we reveal that dysferlin is enriched in the t-tubule membrane of mature skeletal muscle fibers. Following experimental membrane stress in vitro, dysferlin-deficient muscle fibers undergo extensive functional and structural disruption of the t-tubules that is ameliorated by reducing external [Ca(2+)] or blocking L-type Ca(2+) channels with diltiazem. Furthermore, we demonstrate that diltiazem treatment of dysferlin-deficient mice significantly reduces eccentric contraction-induced t-tubule damage, inflammation, and necrosis, which resulted in a concomitant increase in postinjury functional recovery. Our discovery of dysferlin as a t-tubule protein that stabilizes stress-induced Ca(2+) signaling offers a therapeutic avenue for limb girdle muscular dystrophy 2B and Miyoshi myopathy patients.


Asunto(s)
Señalización del Calcio , Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Estrés Fisiológico , Animales , Antihipertensivos/farmacología , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Membrana Celular/patología , Diltiazem/farmacología , Disferlina , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Contracción Muscular/efectos de los fármacos , Contracción Muscular/genética , Fibras Musculares Esqueléticas/patología , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Necrosis/genética , Necrosis/metabolismo , Necrosis/patología
2.
Nat Cell Biol ; 26(6): 991-1002, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38866970

RESUMEN

The contribution of three-dimensional genome organization to physiological ageing is not well known. Here we show that large-scale chromatin reorganization distinguishes young and old bone marrow progenitor (pro-) B cells. These changes result in increased interactions at the compartment level and reduced interactions within topologically associated domains (TADs). The gene encoding Ebf1, a key B cell regulator, switches from compartment A to B with age. Genetically reducing Ebf1 recapitulates some features of old pro-B cells. TADs that are most reduced with age contain genes important for B cell development, including the immunoglobulin heavy chain (Igh) locus. Weaker intra-TAD interactions at Igh correlate with altered variable (V), diversity (D) and joining (J) gene recombination. Our observations implicate three-dimensional chromatin reorganization as a major driver of pro-B cell phenotypes that impair B lymphopoiesis with age.


Asunto(s)
Envejecimiento , Linfocitos B , Ensamble y Desensamble de Cromatina , Cadenas Pesadas de Inmunoglobulina , Linfopoyesis , Animales , Envejecimiento/genética , Envejecimiento/metabolismo , Linfocitos B/metabolismo , Linfopoyesis/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Transactivadores/metabolismo , Transactivadores/genética , Cromatina/metabolismo , Cromatina/genética , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/inmunología , Ratones Endogámicos C57BL , Ratones , Diferenciación Celular , Ratones Noqueados
3.
J Mol Cell Cardiol ; 58: 172-81, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23220288

RESUMEN

X-ROS signaling is a novel redox signaling pathway that links mechanical stress to changes in [Ca(2+)]i. This pathway is activated rapidly and locally within a muscle cell under physiological conditions, but can also contribute to Ca(2+)-dependent arrhythmia in the heart and to the dystrophic phenotype in the heart and skeletal muscle. Upon physiologic cellular stretch, microtubules serve as mechanotransducers to activate NADPH oxidase 2 in the transverse tubules and sarcolemmal membranes to produce reactive oxygen species (ROS). In the heart, the ROS acts locally to activate ryanodine receptor Ca(2+) release channels in the junctional sarcoplasmic reticulum, increasing the Ca(2+) spark rate and "tuning" excitation-contraction coupling. In the skeletal muscle, where Ca(2+) sparks are not normally observed, the X-ROS signaling process is muted. However in muscular dystrophies, such as Duchenne Muscular Dystrophy and dysferlinopathy, X-ROS signaling operates at a high level and contributes to myopathy. Importantly, Ca(2+) permeable stretch-activated channels are activated by X-ROS and contribute to skeletal muscle pathology. Here we review X-ROS signaling and mechanotransduction in striated muscle, and highlight important questions to drive future work on stretch-dependent signaling. We conclude that X-ROS provides an exciting mechanism for the mechanical control of redox and Ca(2+) signaling, but much work is needed to establish its contribution to physiologic and pathophysiologic processes in diverse cell systems.


Asunto(s)
Calcio/metabolismo , Músculo Esquelético/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Señalización del Calcio , Humanos , Músculo Esquelético/patología , Miocitos Cardíacos , Sarcolema/metabolismo , Retículo Sarcoplasmático/metabolismo , Transducción de Señal
4.
J Cell Sci ; 124(Pt 21): 3619-30, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045734

RESUMEN

Small ankyrin 1 (sAnk1; Ank1.5) is a ~20 kDa protein of striated muscle that concentrates in the network compartment of the sarcoplasmic reticulum (nSR). We used siRNA targeted to sAnk1 to assess its role in organizing the sarcoplasmic reticulum (SR) of skeletal myofibers in vitro. siRNA reduced sAnk1 mRNA and protein levels and disrupted the organization of the remaining sAnk1. Sarcomeric proteins were unchanged, but two other proteins of the nSR, SERCA and sarcolipin, decreased significantly in amount and segregated into distinct structures containing sarcolipin and sAnk1, and SERCA, respectively. Exogenous sAnk1 restored SERCA to its normal distribution. Ryanodine receptors and calsequestrin in the junctional SR, and L-type Ca(2+) channels in the transverse tubules were not reduced, although their striated organization was mildly altered. Consistent with the loss of SERCA, uptake and release of Ca(2+) were significantly inhibited. Our results show that sAnk1 stabilizes the nSR and that its absence causes the nSR to fragment into distinct membrane compartments.


Asunto(s)
Ancirinas/metabolismo , Músculo Esquelético/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Ancirinas/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteolípidos/genética , Proteolípidos/metabolismo , Ratas , Retículo Sarcoplasmático/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
5.
J Biol Chem ; 285(53): 41686-700, 2010 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-21041300

RESUMEN

Although the function of protein kinase D1 (PKD) in cardiac cells has remained enigmatic, recent work has shown that PKD phosphorylates the nuclear regulators HDAC5/7 (histone deacetylase 5/7) and CREB, implicating this kinase in the development of dysfunction seen in heart failure. Additional studies have shown that PKD also phosphorylates multiple sarcomeric substrates to regulate myofilament function. Initial studies examined PKD through adenoviral vector expression of wild type PKD, constitutively active PKD (caPKD), or dominant negative PKD in cultured adult rat ventricular myocytes. Confocal immunofluorescent images of these cells reveal a predominant distribution of all PKD forms in a non-nuclear, Z-line localized, striated reticular pattern, suggesting the importance of PKD in Ca(2+) signaling in heart. Consistent with an established role of PKD in targeting cardiac troponin I (cTnI), caPKD expression led to a marked decrease in contractile myofilament Ca(2+) sensitivity with an unexpected electrical stimulus dependence to this response. This desensitization was accompanied by stimulus-dependent increases in cTnI phosphorylation in control and caPKD cells with a more pronounced effect in the latter. Electrical stimulation also provoked phosphorylation of regulatory site Ser(916) on PKD. The functional importance of this phospho-Ser(916) event is demonstrated in experiments with a phosphorylation-defective mutant, caPKD-S916A, which is functionally inactive and blocks stimulus-dependent increases in cTnI phosphorylation. Dominant negative PKD expression resulted in sensitization of the myofilaments to Ca(2+) and blocked stimulus-dependent increases in cTnI phosphorylation. Taken together, these data reveal that localized PKD may play a role as a dynamic regulator of Ca(2+) sensitivity of contraction in cardiac myocytes.


Asunto(s)
Calcio/metabolismo , Regulación Enzimológica de la Expresión Génica , Miocardio/enzimología , Proteína Quinasa C/fisiología , Animales , Sitios de Unión , Señalización del Calcio , Ventrículos Cardíacos/citología , Masculino , Células Musculares/citología , Contracción Muscular , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal
6.
Biophys J ; 99(8): 2705-14, 2010 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-20959112

RESUMEN

Skeletal muscle stores Ca²(+) in the sarcoplasmic reticulum (SR) and releases it to initiate contraction, but the concentration of luminal Ca²(+) in the SR ([Ca²(+)](SR)) and the amount that is released by physiological or pharmacological stimulation has been difficult to measure. Here we present a novel, yet simple and direct, method that provides the first quantitative estimates of static content and dynamic changes in [Ca²(+)](SR) in mammalian skeletal muscle, to our knowledge. The method uses fluo-5N loaded into the SR of single, mammalian skeletal muscle cells (murine flexor digitorum brevis myofibers) and confocal imaging to detect and calibrate the signals. Using this method, we have determined that [Ca²(+)](SR, free) is 390 µM. 4-Chloro-m-cresol, an activator of the skeletal muscle ryanodine receptor, reduces [Ca²(+)](SR, free) to ∼8 µM, when values are corrected for background fluorescence from cytoplasmic pools of dye. Prolonged electrical stimulation (10 s) at 50 Hz releases 88% of the SR Ca²(+) content, whereas stimulation at 1 Hz (10 s) releases only 20%. Our results lay the foundation for molecular modeling of the dynamics of luminal SR Ca²(+) and for future studies of the role of SR Ca²(+) in healthy and diseased mammalian muscle.


Asunto(s)
Calcio/metabolismo , Músculo Esquelético/citología , Retículo Sarcoplasmático/metabolismo , Animales , Calibración , Cresoles/farmacología , Estimulación Eléctrica , Pie , Ratones , Movimiento/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Retículo Sarcoplasmático/efectos de los fármacos
7.
J Mol Cell Cardiol ; 48(2): 379-86, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19818794

RESUMEN

Cardiac contraction is initiated by the release of Ca(2+) from intracellular stores in response to an action potential, in a process known as "excitation-contraction coupling" (ECC). Here we investigate the maturation of ECC in the rat heart during postnatal development. We provide new information on how proteins of the sarcoplasmic reticulum (SR) and the t-tubules (TTs) assemble to form the structures that support EC coupling during postnatal development. We show that the surface membrane protein, caveolin-3 (Cav3), is a good protein marker for TTs in ventricular myocytes and compared it quantitatively to junctophilin-2 (JP2), a protein found on the SR at sites of SR-TT junctions, or couplons. Although JP2 and Cav3 associate primarily with the SR and TTs, respectively, we found that they occupy the appropriate sites at maturing structures in synchrony, as visualized with high resolution, quantitative 3-dimensional imaging. We also found the surprising result that while both ryanodine receptor type 2, (RyR2) and JP2 proteins are localized to the same membrane and sub-compartments, they assume their positions at very different rates: RyR2 moves to the SR membrane at the Z-disc very early in development while JP2 only appears in the SR membrane as the TTs mature. Our data suggest that, although RyR2 appears to be prepositioned at the sites ultimately occupied by dyad junctions, JP2 arrives at these sites in synchrony with the development of the TTs at the Z-discs. Finally, we report that EC coupling efficiency changes with development, in concert with these structural changes. Thus we provide the first well-integrated information that links the developing organization of proteins underlying EC coupling (RyR2, DHPR, Cav3 and JP2) to the developing efficacy of EC coupling.


Asunto(s)
Acoplamiento Excitación-Contracción/fisiología , Corazón/crecimiento & desarrollo , Contracción Miocárdica/fisiología , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Calcio/metabolismo , Caveolina 3/metabolismo , Activación del Canal Iónico , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Transporte de Proteínas , Ratas , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/fisiología
8.
Biochem Pharmacol ; 168: 224-236, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31306645

RESUMEN

The constitutive androstane receptor (CAR) plays an important role in hepatic drug metabolism and detoxification but has recently been projected as a potential drug target for metabolic disorders due to its repression of lipogenesis and gluconeogenesis. Thus, identification of physiologically-relevant CAR modulators has garnered significant interest. Here, we adapted the previously characterized human CAR (hCAR) nuclear translocation assay in human primary hepatocytes (HPH) to a high-content format and screened an FDA-approved drug library containing 978 compounds. Comparison of hCAR nuclear translocation results with the Tox21 hCAR luciferase reporter assay database in 643 shared compounds revealed significant overlap between these two assays, with approximately half of hCAR agonists also mediating nuclear translocation. Further validation of these compounds in HPH and/or using published data from literature demonstrated that hCAR translocation exhibits a higher correlation with the induction of hCAR target genes, such as CYP2B6, than the luciferase assay. In addition, some CAR antagonists which repress CYP2B6 mRNA expression in HPH, such as sorafenib, rimonabant, and CINPA1, were found to translocate hCAR to the nucleus of HPH. Notably, both the translocation assay and the luciferase assay identified mosapride citrate (MOS), a gastroprokinetic agent that is known to reduce fasting blood glucose levels in humans, as a novel hCAR activator. Further studies with MOS in HPH uncovered that MOS can repress the expression of gluconeogenic genes and decrease glucose output from hepatocytes, providing a previously unidentified liver-specific mechanism by which MOS modulates blood glucose levels.


Asunto(s)
Benzamidas/farmacología , Gluconeogénesis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Morfolinas/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptor de Androstano Constitutivo , Gluconeogénesis/fisiología , Humanos
9.
Ann N Y Acad Sci ; 1047: 99-111, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16093488

RESUMEN

Ca(2+) sparks in heart muscle are activated on depolarization by the influx of Ca(2+) through dihydropyridine receptors in the sarcolemmal (SL) and transverse tubule (TT) membranes. The cardiac action potential is thus able to synchronize the [Ca(2+)](i) transient as Ca(2+) release is activated throughout the cell. Increases in the amount of Ca(2+) within the sarcoplasmic reticulum (SR) underlie augmented Ca(2+) release globally and an increase in the sensitivity of the ryanodine receptors (RyRs) to be triggered by the local [Ca(2+)](i). In a similar manner, phosphorylation of the RyRs by protein kinase A (PKA) increases the sensitivity of the RyRs to be activated by local [Ca(2+)](i). Heart failure and other cardiac diseases are associated with changes in SR Ca(2+) content, phosphorylation state of the RyRs, [Ca(2+)](i) signaling defects and arrhythmias. Additional changes in transverse tubules and nearby junctional SR may contribute to alterations in local Ca(2+) signaling. Here we briefly discuss how TT organization can influence Ca(2+) signaling and how changes in SR Ca(2+) release triggering can influence excitation-contraction (EC) coupling. High speed imaging methods are used in combination with single cell patch clamp experiments to investigate how abnormal Ca(2+) signaling may be regulated in health and disease. Three issues are examined in this presentation: (1) normal Ca(2+)-induced Ca(2+) release and Ca(2+) sparks, (2) abnormal SR Ca(2+) release in disease, and (3) the triggering and propagation of waves of elevated [Ca(2+)](i).


Asunto(s)
Señalización del Calcio/fisiología , Miocitos Cardíacos/metabolismo , Sarcómeros/fisiología , Retículo Sarcoplasmático/fisiología , Animales , Electrofisiología , Humanos , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Sarcómeros/ultraestructura , Retículo Sarcoplasmático/ultraestructura
11.
PLoS One ; 6(12): e27036, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22164205

RESUMEN

In skeletal muscle, the release of calcium (Ca(2+)) by ryanodine sensitive sarcoplasmic reticulum (SR) Ca(2+) release channels (i.e., ryanodine receptors; RyR1s) is the primary determinant of contractile filament activation. Much attention has been focused on calsequestrin (CASQ1) and its role in SR Ca(2+) buffering as well as its potential for modulating RyR1, the L-type Ca(2+) channel (dihydropyridine receptor, DHPR) and other sarcolemmal channels through sensing luminal [Ca(2+)]. The genetic ablation of CASQ1 expression results in significant alterations in SR Ca(2+) content and SR Ca(2+) release especially during prolonged activation. While these findings predict a significant loss-of-function phenotype in vivo, little information on functional status of CASQ1 null mice is available. We examined fast muscle in vivo and in vitro and identified significant deficits in functional performance that indicate an inability to sustain contractile activation. In single CASQ1 null skeletal myofibers we demonstrate a decrease in voltage dependent RyR Ca(2+) release with single action potentials and a collapse of the Ca(2+) release with repetitive trains. Under voltage clamp, SR Ca(2+) release flux and total SR Ca(2+) release are significantly reduced in CASQ1 null myofibers. The decrease in peak Ca(2+) release flux appears to be solely due to elimination of the slowly decaying component of SR Ca(2+) release, whereas the rapidly decaying component of SR Ca(2+) release is not altered in either amplitude or time course in CASQ1 null fibers. Finally, intra-SR [Ca(2+)] during ligand and voltage activation of RyR1 revealed a significant decrease in the SR[Ca(2+)](free) in intact CASQ1 null fibers and a increase in the release and uptake kinetics consistent with a depletion of intra-SR Ca(2+) buffering capacity. Taken together we have revealed that the genetic ablation of CASQ1 expression results in significant functional deficits consistent with a decrease in the slowly decaying component of SR Ca(2+) release.


Asunto(s)
Calcio/metabolismo , Calsecuestrina/genética , Ratones Transgénicos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Calcio/química , Electrodos , Electrofisiología/métodos , Cinética , Ratones , Modelos Biológicos , Contracción Muscular , Músculo Esquelético/metabolismo , Fenotipo , Condicionamiento Físico Animal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA