Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 297(6): 101358, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34756888

RESUMEN

Preserving optimal mitochondrial function is critical in the heart, which is the most ATP-avid organ in the body. Recently, we showed that global deficiency of the nuclear receptor RORα in the "staggerer" mouse exacerbates angiotensin II-induced cardiac hypertrophy and compromises cardiomyocyte mitochondrial function. However, the mechanisms underlying these observations have not been defined previously. Here, we used pharmacological and genetic gain- and loss-of-function tools to demonstrate that RORα regulates cardiomyocyte mitophagy to preserve mitochondrial abundance and function. We found that cardiomyocyte mitochondria in staggerer mice with lack of functional RORα were less numerous and exhibited fewer mitophagy events than those in WT controls. The hearts of our novel cardiomyocyte-specific RORα KO mouse line demonstrated impaired contractile function, enhanced oxidative stress, increased apoptosis, and reduced autophagic flux relative to Cre(-) littermates. We found that cardiomyocyte mitochondria in "staggerer" mice with lack of functional RORα were upregulated by hypoxia, a classical inducer of mitophagy. The loss of RORα blunted mitophagy and broadly compromised mitochondrial function in normoxic and hypoxic conditions in vivo and in vitro. We also show that RORα is a direct transcriptional regulator of the mitophagy mediator caveolin-3 in cardiomyocytes and that enhanced expression of RORα increases caveolin-3 abundance and enhances mitophagy. Finally, knockdown of RORα impairs cardiomyocyte mitophagy, compromises mitochondrial function, and induces apoptosis, but these defects could be rescued by caveolin-3 overexpression. Collectively, these findings reveal a novel role for RORα in regulating mitophagy through caveolin-3 and expand our currently limited understanding of the mechanisms underlying RORα-mediated cardioprotection.


Asunto(s)
Caveolina 3/fisiología , Mitocondrias Cardíacas/fisiología , Mitofagia/fisiología , Miocitos Cardíacos/fisiología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Animales , Ratones , Mitocondrias Cardíacas/metabolismo
2.
J Neurosci ; 41(17): 3764-3776, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33731449

RESUMEN

The axon initial segment (AIS) is a specialized neuronal compartment in which synaptic input is converted into action potential (AP) output. This process is supported by a diverse complement of sodium, potassium, and calcium channels (CaV). Different classes of sodium and potassium channels are scaffolded at specific sites within the AIS, conferring unique functions, but how calcium channels are functionally distributed within the AIS is unclear. Here, we use conventional two-photon laser scanning and diffraction-limited, high-speed spot two-photon imaging to resolve AP-evoked calcium dynamics in the AIS with high spatiotemporal resolution. In mouse layer 5 prefrontal pyramidal neurons, calcium influx was mediated by a mix of CaV2 and CaV3 channels that differentially localized to discrete regions. CaV3 functionally localized to produce nanodomain hotspots of calcium influx that coupled to ryanodine-sensitive stores, whereas CaV2 localized to non-hotspot regions. Thus, different pools of CaVs appear to play distinct roles in AIS function.SIGNIFICANCE STATEMENT The axon initial segment (AIS) is the site where synaptic input is transformed into action potential (AP) output. It achieves this function through a diverse complement of sodium, potassium, and calcium channels (CaV). While the localization and function of sodium channels and potassium channels at the AIS is well described, less is known about the functional distribution of CaVs. We used high-speed two-photon imaging to understand activity-dependent calcium dynamics in the AIS of mouse neocortical pyramidal neurons. Surprisingly, we found that calcium influx occurred in two distinct domains: CaV3 generates hotspot regions of calcium influx coupled to calcium stores, whereas CaV2 channels underlie diffuse calcium influx between hotspots. Therefore, different CaV classes localize to distinct AIS subdomains, possibly regulating distinct cellular processes.


Asunto(s)
Segmento Inicial del Axón/fisiología , Segmento Inicial del Axón/ultraestructura , Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Potenciales de Acción/fisiología , Animales , Axones , Caveolina 2/efectos de los fármacos , Caveolina 2/fisiología , Caveolina 3/efectos de los fármacos , Caveolina 3/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Rianodina/farmacología , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos
3.
PLoS One ; 12(12): e0189004, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29206848

RESUMEN

OBJECTIVE: Caveolin-3 (CAV3) protein is known to be expressed specifically in various myocytes, but its physiological function remains unclear. CAV3, located at the cell membrane, may promote the sensitivity of the Akt signaling pathway, which is closely related to glucose metabolism and to cell growth and proliferation. METHODS: The CAV3 gene was stably transfected into C2C12 muscle cells, and the effects were evaluated by biochemical assays, WB and confocal microscopy for the observation of cellular glucose metabolism, growth and proliferation, and the effect of CAV3 on the Akt signaling pathway with no insulin stimulation. RESULTS: After C2C12 cells were transfected with the mouse CAV3 gene, which increased CAV3 expression, the abundance of the CAV3 and GLUT4 proteins on the cell membrane increased, but the total GLUT4 protein content of the cell was unchanged. Glucose uptake was increased, and this did not affect the glycogen synthesis, but the cell surface area and cell proliferation increased. While there were significant increases in p-Akt and p-p70s6K, which is a downstream component of Akt signaling, the level of GSK3ß protein, another component of Akt signaling did not change. CONCLUSIONS: The muscle, CAV3 protein can activate Akt signaling, increase GLUT4 protein localization in the cell membrane, increase glucose uptake, and promote myocyte growth and proliferation. CAV3 protein has a physiological role in glycometabolism, growth and proliferation, independent of insulin stimulation.


Asunto(s)
Caveolina 3/fisiología , Proliferación Celular/fisiología , Glucosa/metabolismo , Músculo Esquelético/metabolismo , Animales , Caveolina 3/genética , Línea Celular , Transportador de Glucosa de Tipo 4/metabolismo , Ratones , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Proteínas Quinasas/metabolismo , Transfección
4.
Eur Rev Med Pharmacol Sci ; 21(14): 3262-3269, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28770956

RESUMEN

OBJECTIVE: To investigate the correlations of expressions of Caveolae-3 (Cav-3) and sma and mad homologue (Smad3) with the pathogenesis and prognosis of viral myocarditis (VMC). MATERIALS AND METHODS: VMC animal models were prepared and divided into the control group, the virus group and the Shenmai group. We detected the levels of creatine kinase isoenzyme (CK-MB) in the serum that was associated with the myocardial injuries, investigated the pathological features of VMC in BALB/C mice via hematoxylin-eosin (HE) staining, measured the mRNA expressions of Cav-3 and Smad3 via Real-time polymerase chain reaction (RT-PCR) and determined the protein expressions of Cav-3 and Smad3 through Western blotting method. RESULTS: The expressions of CK-MB in the virus group and Shenmai group were significantly higher than those in the control group; in comparison with the virus group, obvious improvement was identified in the pathologic condition of the Shenmai group; also, there was a statistically significant difference in comparison of the pathologic scores of BALB/C mice between the Shenmai group and the virus group. The mRNA expressions of Cav-3 and Smad3 in the virus group and Shenmai group were significantly higher than those in the control group, and the differences had statistical significance; however, higher mRNA expressions were identified in the virus group. Besides, protein expressions of Cav-3 and Smad3 in the virus group and Shenmai group were remarkably higher than those in the control group with statistically significant differences, but those in the virus group were much higher. CONCLUSIONS: Cav-3 and Smad3 may be involved in the occurrence and development of VMC, which provides some theoretical evidence for further research into the pathogenesis of VMC and the development of clinical drugs for treatment of VMC.


Asunto(s)
Caveolina 3/fisiología , Infecciones por Coxsackievirus/etiología , Enterovirus Humano B , Miocarditis/etiología , Proteína smad3/fisiología , Animales , Caveolina 3/análisis , Forma MB de la Creatina-Quinasa/sangre , Masculino , Ratones , Ratones Endogámicos BALB C , Pronóstico , Proteína smad3/análisis
5.
Am J Physiol Heart Circ Physiol ; 309(12): H2127-36, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26497963

RESUMEN

Muscle-restricted coiled-coil protein (MURC), also referred to as cavin-4, is a member of the cavin family that works cooperatively with caveolins in caveola formation and function. Cavins are cytoplasmic proteins with coiled-coil domains and form heteromeric complexes, which are recruited to caveolae in cells expressing caveolins. Among caveolins, caveolin-3 (Cav3) is exclusively expressed in muscle cells, similar to MURC/cavin-4. In the heart, Cav3 overexpression contributes to cardiac protection, and its deficiency leads to progressive cardiomyopathy. Mutations in the MURC/cavin-4 gene have been identified in patients with dilated cardiomyopathy. In the present study, we show the role of MURC/cavin-4 as a caveolar component in the heart. In H9c2 cells, MURC/cavin-4 was localized at the plasma membrane, whereas a MURC/cavin-4 mutant lacking the coiled-coil domain (ΔCC) was primarily localized to the cytoplasm. ΔCC bound to Cav3 and impaired membrane localization of Cav3 in cardiomyocytes. Additionally, although ΔCC did not alter Cav3 mRNA expression, ΔCC decreased the Cav3 protein level. MURC/cavin-4 and ΔCC similarly induced cardiomyocyte hypertrophy; however, ΔCC showed higher hypertrophy-related fetal gene expression than MURC/cavin-4. ΔCC induced ERK activation in cardiomyocytes. Transgenic mice expressing ΔCC in the heart (ΔCC-Tg mice) showed impaired cardiac function accompanied by cardiomyocyte hypertrophy and marked interstitial fibrosis. Hearts from ΔCC-Tg mice showed a reduction of the Cav3 protein level and activation of ERK. These results suggest that MURC/cavin-4 requires its coiled-coil domain to target the plasma membrane and to stabilize Cav3 at the plasma membrane of cardiomyocytes and that MURC/cavin-4 functions as a crucial caveolar component to regulate cardiac function.


Asunto(s)
Caveolina 3/fisiología , Membrana Celular/metabolismo , Proteínas Musculares/fisiología , Miocitos Cardíacos/metabolismo , Animales , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/patología , Caveolina 3/genética , Línea Celular , Citosol/metabolismo , Fibrosis Endomiocárdica/patología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Transgénicos , Proteínas Musculares/genética , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/ultraestructura , Plásmidos/genética , Conformación Proteica , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Ultrasonografía
6.
Life Sci ; 121: 35-9, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25476831

RESUMEN

AIMS: Caveolins are structural proteins clustered in lipid-rich regions of plasma membrane involved in coordinating signal transduction in various organ systems. While caveolin-1 (Cav-1) has been shown to regulate lymphocyte activation, the role of caveolin-3 (Cav-3) in immune system signaling has not been investigated. We tested the hypothesis that Cav-3 modulates lymphocyte activation. MAIN METHODS: Lymphocyte/leukocyte subpopulations from WT and Cav-3 mice were profiled with flow cytometry. Cytokine production in quiescent and activated splenocytes from WT and Cav-3 mice was assessed with ELISA. KEY FINDINGS: Levels of T-cells, monocytes, and natural killer cells were not different between WT and KO mice, however KO mice had lower B-cell population-percentage. Functionally, activated lymphocytes from Cav-3 KO mice demonstrated significantly reduced expression of IL-2 compared to WT, while expression of TNFα, IL-6, and IL-10 was not different. Finally, expression of IL-17 was significantly reduced in T-helper cells from KO mice, while IFNγ was not, suggesting that Cav-3 is a determinant in the development of the Th-17 subpopulation. SIGNIFICANCE: This study is the first to demonstrate that Cav-3 may be a novel participant in B-cell expression, T-cell cytokine production and activation of inflammation.


Asunto(s)
Caveolina 3/fisiología , Activación de Linfocitos/fisiología , Animales , Caveolina 3/genética , Citocinas/metabolismo , Citometría de Flujo , Humanos , Sistema Inmunológico/fisiología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
7.
J Neurosci ; 34(42): 14032-45, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25319700

RESUMEN

Mitral cells express low-voltage activated Cav3.3 channels on their distal apical tuft dendrites (McKay et al., 2006; Johnston and Delaney, 2010). They also discharge Na(+)-dependent dendritic action potentials and release glutamate from these dendrites. Around resting membrane potentials, between -65 and -50 mV, Cav3.x channels are a primary determinant of cytoplasmic [Ca(2+)]. In this study using C57 mice, we present evidence that subthreshold Cav3.x-mediated Ca(2+) influx modulates action potential evoked transmitter release and directly drives asynchronous release from distal tuft dendrites. Presynaptic hyperpolarization and selective block of Cav3.x channels with Z941 (Tringham et al., 2012) reduce mitral-to-mitral EPSP amplitude, increase the coefficient of variation of EPSPs, and increase paired-pulse ratios, consistent with a reduced probability of transmitter release. Both hyperpolarization and Cav3.x channel blockade reduce steady-state cytoplasmic [Ca(2+)] in the tuft dendrite without reducing action potential evoked Ca(2+) influx, suggesting that background [Ca(2+)] modulates evoked release. We demonstrate that Cav3.x-mediated Ca(2+) influx from even one mitral cell at membrane potentials between -65 and -50 mV is sufficient to produce feedback inhibition from periglomerular neurons. Deinactivation of Cav3.x channels by hyperpolarization increases T-type Ca(2+) influx upon repolarization and increases feedback inhibition to produce subthreshold modulation of the mitral-periglomerular reciprocal circuit.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Caveolina 3/fisiología , Dendritas/fisiología , Bulbo Olfatorio/fisiología , Vías Olfatorias/fisiología , Terminales Presinápticos/fisiología , Potenciales de Acción/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Masculino , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Inhibición Neural/fisiología , Técnicas de Cultivo de Órganos
8.
Pflugers Arch ; 466(4): 645-60, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24515291

RESUMEN

Cav3 T-type channels are low-voltage-gated channels with rapid kinetics that are classified among the calcium-selective Cav1 and Cav2 type channels. Here, we outline the fundamental and unique regulators of T-type channels. An ubiquitous and proximally located "gating brake" works in concert with the voltage-sensor domain and S6 alpha-helical segment from domain II to set the canonical low-threshold and transient gating features of T-type channels. Gene splicing of optional exon 25c (and/or exon 26) in the short III-IV linker provides a developmental switch between modes of activity, such as activating in response to membrane depolarization, to channels requiring hyperpolarization input before being available to activate. Downstream of the gating brake in the I-II linker is a key region for regulating channel expression where alternative splicing patterns correlate with functional diversity of spike patterns, pacemaking rate (especially in the heart), stage of development, and animal size. A small but persistent window conductance depolarizes cells and boosts excitability at rest. T-type channels possess an ion selectivity that can resemble not only the calcium ion exclusive Cav1 and Cav2 channels but also the sodium ion selectivity of Nav1 sodium channels too. Alternative splicing in the extracellular turret of domain II generates highly sodium-permeable channels, which contribute to low-threshold sodium spikes. Cav3 channels are more ubiquitous among multicellular animals and more widespread in tissues than the more brain centric Nav1 sodium channels in invertebrates. Highly sodium-permeant Cav3 channels can functionally replace Nav1 channels in species where they are lacking, such as in Caenorhabditis elegans.


Asunto(s)
Caveolina 3/fisiología , Regulación de la Expresión Génica , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Secuencia de Aminoácidos , Animales , Cationes , Humanos , Datos de Secuencia Molecular
9.
PLoS One ; 8(9): e73887, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086300

RESUMEN

Aberrant cardiomyocyte microtubule growth is a feature of pressure overload induced cardiac hypertrophy believed to contribute to left ventricular (LV) dysfunction. Microtubule Actin Cross-linking Factor 1 (MACF1/Acf7) is a 600 kd spectraplakin that stabilizes and guides microtubule growth along actin filaments. MACF1 is expressed in the heart, but its impact on cardiac microtubules, and how this influences cardiac structure, function, and adaptation to hemodynamic overload is unknown. Here we used inducible cardiac-specific MACF1 knockout mice (MACF1 KO) to determine the impact of MACF1 on cardiac microtubules and adaptation to pressure overload (transverse aortic constriction (TAC).In adult mouse hearts, MACF1 expression was low under basal conditions, but increased significantly in response to TAC. While MACF1 KO had no observable effect on heart size or function under basal conditions, MACF1 KO exacerbated TAC induced LV hypertrophy, LV dilation and contractile dysfunction. Interestingly, subcellular fractionation of ventricular lysates revealed that MACF1 KO altered microtubule distribution in response to TAC, so that more tubulin was associated with the cell membrane fraction. Moreover, TAC induced microtubule redistribution into this cell membrane fraction in both WT and MACF1 KO mice correlated strikingly with the level of contractile dysfunction (r(2) = 0.786, p<.001). MACF1 disruption also resulted in reduction of membrane caveolin 3 levels, and increased levels of membrane PKCα and ß1 integrin after TAC, suggesting MACF1 function is important for spatial regulation of several physiologically relevant signaling proteins during hypertrophy. Together, these data identify for the first time, a role for MACF1 in cardiomyocyte microtubule distribution and in adaptation to hemodynamic overload.


Asunto(s)
Adaptación Fisiológica , Hemodinámica , Proteínas de Microfilamentos/fisiología , Microtúbulos/fisiología , Miocitos Cardíacos/fisiología , Animales , Secuencia de Bases , Caveolina 3/fisiología , Cartilla de ADN , Ecocardiografía , Integrina beta1/fisiología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa C-alfa/fisiología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Disfunción Ventricular Izquierda
10.
Circulation ; 128(11 Suppl 1): S121-9, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24030395

RESUMEN

BACKGROUND: The inhaled anesthetic sevoflurane has been demonstrated to protect against myocardial ischemia/reperfusion (MI/R) injury via mechanisms involving AMP-activated protein kinase (AMPK) and caveolin-3 (Cav-3). However, the relative contributions of AMPK and Cav-3 to sevoflurane preconditioning (SF-PreCon)-mediated cardioprotection and their precise underlying mechanisms of action remain incompletely understood. METHODS AND RESULTS: SF-PreCon (consisting of 3 cycles of 15-minute exposure to 2% sevoflurane before 30 minutes of MI) decreased MI/R injury in wild-type mice (caspase-3 activity, -29.1%; infarct size, -20.2%; and left ventricular end diastolic pressure, -33.8%). In cardiac-specific AMPKα2 dominant-negative overexpressing mice, the cardioprotective effect of SF-PreCon was largely retained (caspase-3 activity, -26.7%; infarct size, -16.7%; and left ventricular end-diastolic pressure, -25.9%; P<0.01). In contrast, SF-PreCon failed to significantly protect Cav-3 knockout mice against MI/R injury (P>0.05). SF-PreCon significantly decreased MI/R-induced superoxide generation in wild-type (-43.6%) and AMPK dominant-negative overexpressing mice (-35.5%; P<0.01) but not in Cav-3 knockout mice. SF-PreCon did not affect nicotinamide adenine dinucleotide phosphate oxidase expression but significantly inhibited cyclooxygenase-2 expression in wild-type (-38.7%) and AMPK dominant-negative overexpressing mice (-35.8%) but not in Cav-3 knockout mice. CONCLUSIONS: We demonstrate for the first time SF-PreCon mediates cardioprotection against MI/R injury via caveolin-3-dependent cyclooxygenase-2 inhibition and antioxidative effects.


Asunto(s)
Caveolina 3/fisiología , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Precondicionamiento Isquémico Miocárdico/métodos , Éteres Metílicos/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Anestésicos por Inhalación/uso terapéutico , Animales , Caveolina 3/deficiencia , Caveolina 3/genética , Células Cultivadas , Masculino , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/genética , Sevoflurano
11.
J Mol Cell Cardiol ; 61: 102-10, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23541953

RESUMEN

AIMS: Mutations in CAV3-encoding caveolin-3 (Cav3) have been implicated in type 9 long QT syndrome (LQT9) and sudden infant death syndrome (SIDS). When co-expressed with SCN5A-encoded cardiac sodium channels these mutations increased late sodium current (INa) but the mechanism was unclear. The present study was designed to address the mechanism by which the LQT9-causing mutant Cav3-F97C affects the function of caveolar SCN5A. METHODS AND RESULTS: HEK-293 cells expressing SCN5A and LQT9 mutation Cav3-F97C resulted in a 2-fold increase in late INa compared to Cav3-WT. This increase was reversed by the neural nitric oxide synthase (nNOS) inhibitor L-NMMA. Based on these findings, we hypothesized that an nNOS complex mediated the effect of Cav3 on SCN5A. A SCN5A macromolecular complex was established in HEK-293 cells by transiently expressing SCN5A, α1-syntrophin (SNTA1), nNOS, and Cav3. Compared with Cav3-WT, Cav3-F97C produced significantly larger peak INa amplitudes, and showed 3.3-fold increase in the late INa associated with increased S-nitrosylation of SCN5A. L-NMMA reversed both the Cav3-F97C induced increase in late and peak INa and decreased S-nitrosylation of SCN5A. Overexpression of Cav3-F97C in adult rat cardiomyocytes caused a significant increase in late INa compared to Cav3-WT, and prolonged the action potential duration (APD90) in a nNOS-dependent manner. CONCLUSIONS: Cav3 is identified as an important negative regulator for cardiac late INa via nNOS dependent direct S-nitrosylation of SCN5A. This provides a molecular mechanism for how Cav3 mutations increase late INa to cause LQT9. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".


Asunto(s)
Caveolina 3/fisiología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , S-Nitrosotioles/metabolismo , Animales , Células HEK293 , Humanos , Síndrome de QT Prolongado/genética , Potenciales de la Membrana , Mutación Missense , Miocitos Cardíacos/fisiología , Óxido Nítrico/metabolismo , Procesamiento Proteico-Postraduccional , Ratas , Sodio/metabolismo
12.
J Endocrinol ; 211(1): 47-54, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21746791

RESUMEN

Liberal or high-sodium (HS) intake, in conjunction with an activated renin-angiotensin-aldosterone system, increases cardiovascular (CV) damage. We tested the hypothesis that sodium intake regulates the type 1 angiotensin II receptor (AT(1)R), mineralocorticoid receptor (MR), and associated signaling pathways in heart tissue from healthy rodents. HS (1.6% Na(+)) and low-sodium (LS; 0.02% Na(+)) rat chow was fed to male healthy Wistar rats (n=7 animals per group). Protein levels were assessed by western blot and immunoprecipitation analysis. Fractionation studies showed that MR, AT(1)R, caveolin-3 (CAV-3), and CAV-1 were located in both cytoplasmic and membrane fractions. In healthy rats, consumption of an LS versus a HS diet led to decreased cardiac levels of AT(1)R and MR. Decreased sodium intake was also associated with decreased cardiac levels of CAV-1 and CAV-3, decreased immunoprecipitation of AT(1)R-CAV-3 and MR-CAV-3 complexes, but increased immunoprecipitation of AT(1)R/MR complexes. Furthermore, decreased sodium intake was associated with decreased cardiac extracellular signal-regulated kinase (ERK), phosphorylated ERK (pERK), and pERK/ERK ratio; increased cardiac striatin; decreased endothelial nitric oxide synthase (eNOS) and phosphorylated eNOS (peNOS), but increased peNOS/eNOS ratio; and decreased cardiac plasminogen activator inhibitor-1. Dietary sodium restriction has beneficial effects on the cardiac expression of factors associated with CV injury. These changes may play a role in the cardioprotective effects of dietary sodium restriction.


Asunto(s)
Corazón/efectos de los fármacos , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptores de Mineralocorticoides/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sodio en la Dieta/farmacología , Animales , Caveolina 1/efectos de los fármacos , Caveolina 1/fisiología , Caveolina 3/efectos de los fármacos , Caveolina 3/fisiología , Relación Dosis-Respuesta a Droga , Corazón/fisiología , Masculino , Modelos Animales , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1/fisiología , Receptores de Mineralocorticoides/fisiología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Transducción de Señal/fisiología
13.
Circ Res ; 108(7): 884-97, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21454796

RESUMEN

Variations in the gene encoding for the major sodium channel (Na(v)1.5) in the heart, SCN5A, has been shown to cause a number of arrhythmia syndromes (with or without structural changes in the myocardium), including the long-QT syndrome (type 3), Brugada syndrome, (progressive) cardiac conduction disease, sinus node dysfunction, atrial fibrillation, atrial standstill, and dilated cardiomyopathy. Of equal importance are variations in genes encoding for various subunits and regulatory proteins interacting with the α-subunit Na(v)1.5 and modifying its function. Based on detailed studies of genotype-phenotype relationships in these disease entities, on detailed studies of the basic electrophysiological phenotypes (heterologous expressed wild-type and mutant sodium channels and their interacting proteins), and on attempts to integrate the obtained knowledge, the past 15 years has witnessed an explosion of knowledge about these disease entities.


Asunto(s)
Arritmias Cardíacas/genética , Proteínas Musculares/genética , Mutación/genética , Fenotipo , Canales de Sodio/genética , Arritmias Cardíacas/fisiopatología , Proteínas de Unión al Calcio/fisiología , Caveolina 3/fisiología , Glicerolfosfato Deshidrogenasa/fisiología , Humanos , Proteínas de la Membrana/fisiología , Proteínas Musculares/fisiología , Canal de Sodio Activado por Voltaje NAV1.5
14.
Anesthesiology ; 112(5): 1136-45, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20418694

RESUMEN

BACKGROUND: Caveolae are small, flask-like invaginations of the plasma membrane. Caveolins are structural proteins found in caveolae that have scaffolding properties to allow organization of signaling. The authors tested the hypothesis that delayed cardiac protection induced by volatile anesthetics is caveolae or caveolin dependent. METHODS: An in vivo mouse model of ischemia-reperfusion injury with delayed anesthetic preconditioning (APC) was tested in wild-type, caveolin-1 knockout, and caveolin-3 knockout mice. Mice were exposed to 30 min of oxygen or isoflurane and allowed to recover for 24 h. After 24 h recovery, mice underwent 30-min coronary artery occlusion followed by 2 h of reperfusion at which time infarct size was determined. Biochemical assays were also performed in excised hearts. RESULTS: Infarct size as a percent of the area at risk was reduced by isoflurane in wild-type (24.0 +/- 8.8% vs. 45.1 +/- 10.1%) and caveolin-1 knockout mice (27.2 +/- 12.5%). Caveolin-3 knockout mice did not show delayed APC (41.5 +/- 5.0%). Microscopically distinct caveolae were observed in wild-type and caveolin-1 knockout mice but not in caveolin-3 knockout mice. Delayed APC increased the amount of caveolin-3 protein but not caveolin-1 protein in discontinuous sucrose-gradient buoyant fractions. In addition, glucose transporter-4 was increased in buoyant fractions, and caveolin-3/glucose transporter-4 colocalization was observed in wild-type and caveolin-1 knockout mice after APC. CONCLUSIONS: These results show that delayed APC involves translocation of caveolin-3 and glucose transporter-4 to caveolae, resulting in delayed protection in the myocardium.


Asunto(s)
Cardiotónicos/uso terapéutico , Caveolina 3/fisiología , Transportador de Glucosa de Tipo 4/fisiología , Isoflurano/uso terapéutico , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Animales , Cardiotónicos/farmacología , Caveolina 3/deficiencia , Caveolina 3/genética , Precondicionamiento Isquémico Miocárdico/métodos , Isoflurano/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/ultraestructura , Distribución Aleatoria , Factores de Tiempo
15.
Epilepsy Res ; 83(2-3): 224-34, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19124225

RESUMEN

SUMMARY: Zonisamide (ZNS) is a multi-target antiepileptic drug reported to be efficient in the treatment of both partial and generalized seizures, with T-type Ca(2+) channel blockade being one of its proposed mechanisms of action. In this study, we systematically investigated electrophysiological effects of ZNS on cloned human Ca(v)3.1-3.3 Ca(2+) channels in a heterologous HEK-293 expression system using whole cell patch-clamp technique. Concentration-response studies were performed in the range from 5 microM to 2mM for Ca(v)3.2 Ca(2+) channels exhibiting a 15.4-30.8% reduction of Ca(2+) influx within the maximum therapeutic plasma range (50-200 microM ZNS). The other T-type Ca(2+) channel entities, Ca(v)3.1 and Ca(v)3.3, were even less sensitive to ZNS. Both voltage- and concentration-dependence of inactivation kinetics remained unchanged for Ca(v)3.2 VGCC, whereas Ca(v)3.1 and Ca(v)3.3 exhibited minor, though significant reduction of inactivation-tau. Interestingly, ZNS block of Ca(v)3.2 VGCCs was not use-dependent and remained unaffected by changes in the holding potential. Steady-state inactivation studies did not display a significant shift in steady-state availability of Ca(v)3.2 channels at 100 microM ZNS (DeltaV(1/2)=3.1mV, p=0.071). Our studies indicate that ZNS is a moderate blocker of human Ca(v)3 T-type Ca(2+) channels with little or no effect on Ca(v)3.2 Ca(2+) channel inactivation kinetics, use- and state-dependence of blockade. These results suggest that T-type Ca(2+) channel inhibition only partially contributes to the anti-absence activity of ZNS antiepileptic drug.


Asunto(s)
Anticonvulsivantes/farmacología , Fenómenos Biofísicos/efectos de los fármacos , Caveolina 3/fisiología , Isoxazoles/farmacología , Potenciales de la Membrana/efectos de los fármacos , Fenómenos Biofísicos/genética , Biofisica , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Caveolina 3/genética , Línea Celular Transformada , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica/métodos , Humanos , Potenciales de la Membrana/genética , Técnicas de Placa-Clamp/métodos , Transfección/métodos , Zonisamida
16.
Acta Myol ; 27: 19-24, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19108573

RESUMEN

Caveolins, components of the uncoated invaginations of plasma membrane, regulate signal transduction and vesicular trafflicking. Loss of caveolin-3, resulting from dominant negative mutations of caveolin-3 causes autosomal dominant limb-girdle muscular dystrophy (LGMD) 1C and autosomal dominant rippling muscle disease (AD-RMD). Myostatin, a member of the muscle-specific transforming growth factor (TGF)-beta superfamily, negatively regulates skeletal muscle volume. Herein we review caveolin-3 suppressing of activation of type I myostatin receptor, thereby inhibiting subsequent intracellular signaling. In addition, a mouse model of LGMD1C has shown atrophic myopathy with enhanced myostatin signaling. Myostatin inhibition ameliorates muscular phenotype in the model mouse, accompanied by normalized myostatin signaling. Enhanced myostatin signaling by caveolin-3 mutation in human may contribute to the pathogenesis of LGMD1C. Therefore, myostatin inhibition therapy may be a promising treatment for patients with LGMD1C. More recent studies concerning regulation of TGF-beta superfamily signaling by caveolins have provided new insights into the pathogenesis of several human diseases.


Asunto(s)
Caveolina 3/fisiología , Miostatina/fisiología , Transducción de Señal/fisiología , Animales , Caveolina 3/genética , Caveolina 3/metabolismo , Modelos Animales de Enfermedad , Humanos , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/fisiopatología , Distrofia Muscular de Cinturas/terapia , Mutación , Miostatina/antagonistas & inhibidores , Miostatina/metabolismo , Fosforilación , Proteínas Smad/metabolismo , Transcripción Genética/fisiología , Factor de Crecimiento Transformador beta/metabolismo
18.
Biophys J ; 89(3): 1893-901, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15980179

RESUMEN

Caveolae are present in almost all cells and concentrate a wide variety of signaling molecules, receptors, transporters, and ion pumps. We have investigated the distribution of the ryanodine receptor, the Na(+)/Ca(2+) exchanger, the predominant Na(+) channel isoform rH1, and the L-type calcium channel, Ca(v)1.2, relative to the muscle-specific caveolin isoform, caveolin-3, in adult rat ventricular myocytes. Three-dimensional immunofluorescence images were deconvolved and analyzed. Caveolin-3 colocalizes with all of these molecules at the surface of the cell, but there is no significant colocalization between caveolin-3 and either the Na(+)/Ca(2+) exchanger or the Na(+) channel in the cell interior. The distribution of the surface colocalization indicates that the caveolae that colocalize with each molecule form distinct populations. This organization indicates that there are multiple populations of caveolae separable by location and occupants. In the interior of the cell, caveolin-3 shows a marked colocalization with a population of ryanodine receptors that are separate from those within the dyad. Because of their location, the signaling molecules contained within these caveolae may have preferred access to the neighboring nondyadic ryanodine receptors.


Asunto(s)
Caveolina 3/fisiología , Canal Liberador de Calcio Receptor de Rianodina/química , Intercambiador de Sodio-Calcio/química , Algoritmos , Animales , Proteínas de Arabidopsis , Canales de Calcio Tipo L/química , Caveolina 3/química , Caveolina 3/metabolismo , Caveolinas/química , Membrana Celular/metabolismo , Células Cultivadas , Ventrículos Cardíacos/citología , Procesamiento de Imagen Asistido por Computador , Iones/química , Masculino , Microscopía Fluorescente , Método de Montecarlo , Células Musculares/metabolismo , Contracción Miocárdica , Miocitos Cardíacos/citología , Unión Proteica , Isoformas de Proteínas , Ratas , Ratas Wistar , Transducción de Señal , Intercambiador de Sodio-Calcio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...