Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(32): 19497-19506, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32719120

RESUMEN

Understanding the genetics of susceptibility to infectious agents is of great importance to our ability to combat disease. Here, we show that voltage-gated calcium channels (VGCCs) are critical for cellular binding and entry of the New World arenaviruses Junín and Tacaribe virus, suggesting that zoonosis via these receptors could occur. Moreover, we demonstrate that α1s haploinsufficiency renders cells and mice more resistant to infection by these viruses. In addition to being more resistant to infection, haploinsufficient cells and mice required a lower dosage of VGCC antagonists to block infection. These studies underscore the importance of genetic variation in susceptibility to both viruses and pharmaceutics.


Asunto(s)
Infecciones por Arenaviridae/genética , Canales de Calcio Tipo L/genética , Resistencia a la Enfermedad/genética , Animales , Infecciones por Arenaviridae/tratamiento farmacológico , Arenavirus del Nuevo Mundo/fisiología , Agonistas de los Canales de Calcio/farmacología , Agonistas de los Canales de Calcio/uso terapéutico , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Heterocigoto , Humanos , Ratones , Ratones Mutantes , Mutación , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
2.
Eur Arch Psychiatry Clin Neurosci ; 269(8): 949-962, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30267149

RESUMEN

Genetic (G) and environmental (E) factors are involved in the etiology and course of the major psychoses (MP), i.e. major depressive disorder (MDD), bipolar disorder (BD), schizoaffective disorder (SZA) and schizophrenia (SZ). The neurobiological correlates by which these predispositions exert their influence on brain structure, function and course of illness are poorly understood. In the FOR2107 consortium, animal models and humans are investigated. A human cohort of MP patients, healthy subjects at genetic and/or environmental risk, and control subjects (N = 2500) has been established. Participants are followed up after 2 years and twice underwent extensive deep phenotyping (MR imaging, clinical course, neuropsychology, personality, risk/protective factors, biomaterials: blood, stool, urine, hair, saliva). Methods for data reduction, quality assurance for longitudinal MRI data, and (deep) machine learning techniques are employed. In the parallelised animal cluster, genetic risk was introduced by a rodent model (Cacna1c deficiency) and its interactions with environmental risk and protective factors are studied. The animals are deeply phenotyped regarding cognition, emotion, and social function, paralleling the variables assessed in humans. A set of innovative experimental projects connect and integrate data from the human and animal parts, investigating the role of microRNA, neuroplasticity, immune signatures, (epi-)genetics and gene expression. Biomaterial from humans and animals are analyzed in parallel. The FOR2107 consortium will delineate pathophysiological entities with common neurobiological underpinnings ("biotypes") and pave the way for an etiologic understanding of the MP, potentially leading to their prevention, the prediction of individual disease courses, and novel therapies in the future.


Asunto(s)
Encéfalo/patología , Trastornos Psicóticos/patología , Animales , Encéfalo/fisiopatología , Canales de Calcio Tipo L/deficiencia , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Entrevista Psicológica , Imagen por Resonancia Magnética , Masculino , MicroARNs/metabolismo , Neuroimagen , Fenotipo , Trastornos Psicóticos/etiología , Trastornos Psicóticos/fisiopatología , Ratas , Factores de Riesgo
3.
J Neurosci ; 37(42): 10038-10051, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-28899915

RESUMEN

Exploring the molecular mechanisms that drive the maturation of oligodendrocyte progenitor cells (OPCs) during the remyelination process is essential to developing new therapeutic tools to intervene in demyelinating diseases such as multiple sclerosis. To determine whether L-type voltage-gated calcium channels (L-VGCCs) are required for OPC development during remyelination, we generated an inducible conditional knock-out mouse in which the L-VGCC isoform Cav1.2 was deleted in NG2-positive OPCs (Cav1.2KO). Using the cuprizone (CPZ) model of demyelination and mice of either sex, we establish that Cav1.2 deletion in OPCs leads to less efficient remyelination of the adult brain. Specifically, Cav1.2KO OPCs mature slower and produce less myelin than control oligodendrocytes during the recovery period after CPZ intoxication. This reduced remyelination was accompanied by an important decline in the number of myelinating oligodendrocytes and in the rate of OPC proliferation. Furthermore, during the remyelination phase of the CPZ model, the corpus callosum of Cav1.2KO animals presented a significant decrease in the percentage of myelinated axons and a substantial increase in the mean g-ratio of myelinated axons compared with controls. In addition, in a mouse line in which the Cav1.2KO OPCs were identified by a Cre reporter, we establish that Cav1.2KO OPCs display a reduced maturational rate through the entire remyelination process. These results suggest that Ca2+ influx mediated by L-VGCCs in oligodendroglial cells is necessary for normal remyelination and is an essential Ca2+ channel for OPC maturation during the remyelination of the adult brain.SIGNIFICANCE STATEMENT Ion channels implicated in oligodendrocyte differentiation and maturation may induce positive signals for myelin recovery. Voltage-gated Ca2+ channels (VGCCs) are important for normal myelination by acting at several critical steps during oligodendrocyte progenitor cell (OPC) development. To determine whether voltage Ca2+ entry is involved in oligodendrocyte differentiation and remyelination, we used a conditional knockout mouse for VGCCs in OPCs. Our results indicate that VGCCs can modulate oligodendrocyte maturation in the demyelinated brain and suggest that voltage-gated Ca2+ influx in OPCs is critical for remyelination. These findings could lead to novel approaches for obtaining a better understanding of the factors that control OPC maturation in order to stimulate this pool of progenitors to replace myelin in demyelinating diseases.


Asunto(s)
Antígenos/biosíntesis , Canales de Calcio Tipo L/deficiencia , Eliminación de Gen , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Proteoglicanos/biosíntesis , Animales , Antígenos/genética , Encéfalo/metabolismo , Encéfalo/patología , Canales de Calcio Tipo L/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Vaina de Mielina/genética , Fibras Nerviosas Mielínicas/patología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Proteoglicanos/genética
4.
Hippocampus ; 28(2): 97-107, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29116659

RESUMEN

In the central nervous system, CaV 1.2 and CaV 1. 3 constitute the main L-type voltage-gated calcium channels (LTCCs) coupling membrane depolarization to gene transcription. We have previously demonstrated that inducible disruption of Cav1.2 in type-1 astrocyte-like stem cells of the adult dentate gyrus (DG) impairs hippocampal neurogenesis in a cell-autonomous fashion. To address the role of Cav1.3 channels (encoded by the Cacna1d gene), we here generated TgGLAST-CreERT2 /Cacna1dfl/fl /RCE:loxP mice which facilitate inducible deletion of Cacna1d in tandem with induction of EGFP expression in type-1 cells, allowing tracking of recombined cells and their descendants. Neurosphere cultures derived from fluorescence-activated cell sorting sorted Cacna1d-deficient (Cacna1d-/- /EGFP) hippocampal neural precursor cells (NPCs) exhibited a significant decrease in proliferative activity. Further, under differentiation conditions, Cacna1d deficiency conferred an increase in astrogenesis at the expense of neurogenesis. In like manner, type-1 cells lacking Cacna1d showed reduced proliferation in the dentate gyrus (DG) in vivo. Moreover, Cacna1d deficiency resulted in a significant decrease in the number of newly born cells adopting a neuronal fate. Finally, massive excitation induced by repeated electroconvulsive seizures rescued the proliferation defect of Cacna1d-/- /EGFP type-1 cells. Together, the effects of Cacna1d gene deletion closely recapitulate our earlier findings on the role of Cav1.2 channels expressed by type-1 cells. Similar to Cav1.2 channels, Cav1.3 channels on type-1 cells boost type-1 cell proliferation and promote subsequent neuronal fate choice.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Proliferación Celular/genética , Eliminación de Gen , Neuronas/fisiología , Animales , Canales de Calcio Tipo L/genética , Diferenciación Celular , Células Cultivadas , Giro Dentado/citología , Proteínas de Dominio Doblecortina , Estimulación Eléctrica/efectos adversos , Epilepsia/etiología , Epilepsia/patología , Epilepsia/fisiopatología , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/fisiología , Neurogénesis/genética , Neuropéptidos/metabolismo
5.
Proc Natl Acad Sci U S A ; 112(2): 602-6, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25548159

RESUMEN

Excitation-contraction (EC) coupling in skeletal muscle depends upon trafficking of CaV1.1, the principal subunit of the dihydropyridine receptor (DHPR) (L-type Ca(2+) channel), to plasma membrane regions at which the DHPRs interact with type 1 ryanodine receptors (RyR1) in the sarcoplasmic reticulum. A distinctive feature of this trafficking is that CaV1.1 expresses poorly or not at all in mammalian cells that are not of muscle origin (e.g., tsA201 cells), in which all of the other nine CaV isoforms have been successfully expressed. Here, we tested whether plasma membrane trafficking of CaV1.1 in tsA201 cells is promoted by the adapter protein Stac3, because recent work has shown that genetic deletion of Stac3 in skeletal muscle causes the loss of EC coupling. Using fluorescently tagged constructs, we found that Stac3 and CaV1.1 traffic together to the tsA201 plasma membrane, whereas CaV1.1 is retained intracellularly when Stac3 is absent. Moreover, L-type Ca(2+) channel function in tsA201 cells coexpressing Stac3 and CaV1.1 is quantitatively similar to that in myotubes, despite the absence of RyR1. Although Stac3 is not required for surface expression of CaV1.2, the principle subunit of the cardiac/brain L-type Ca(2+) channel, Stac3 does bind to CaV1.2 and, as a result, greatly slows the rate of current inactivation, with Stac2 acting similarly. Overall, these results indicate that Stac3 is an essential chaperone of CaV1.1 in skeletal muscle and that in the brain, Stac2 and Stac3 may significantly modulate CaV1.2 function.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Fibras Musculares Esqueléticas/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Línea Celular , Células Cultivadas , Acoplamiento Excitación-Contracción/fisiología , Humanos , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/fisiología
6.
J Neurosci ; 33(24): 9920-31, 2013 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-23761887

RESUMEN

Febrile seizures are associated with increased brain temperature and are often resistant to treatments with antiepileptic drugs, such as carbamazepine and phenytoin, which are sodium channel blockers. Although they are clearly correlated with the hyperthermic condition, the precise cellular mechanisms of febrile seizures remain unclear. We performed patch-clamp recordings from pyramidal cells in acute rat brain slices at temperatures up to 40°C and found that, at ≥37°C, L-type calcium channels are active at unexpectedly hyperpolarized potentials and drive intrinsic firing, which is also supported by a temperature-dependent, gadolinium-sensitive sodium conductance. Pharmacological data, RT-PCR, and the current persistence in Cav1.3 knock-out mice suggested a critical contribution of Cav1.2 subunits to the temperature-dependent intrinsic firing, which was blocked by nimodipine. Because intrinsic firing may play a critical role in febrile seizures, we tested the effect of nimodipine in an in vivo model of febrile seizures and found that this drug dramatically reduces both the incidence and duration of febrile seizures in rat pups, suggesting new possibilities of intervention for this important pathological condition.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Hipocampo/patología , Células Piramidales/fisiología , Convulsiones Febriles/patología , Temperatura , Anilidas/farmacología , Animales , Animales Recién Nacidos , Cloruro de Cadmio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Cinamatos/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Electroencefalografía , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Nimodipina/farmacología , Técnicas de Placa-Clamp , Ratas , Ratas Long-Evans , Convulsiones Febriles/genética , Convulsiones Febriles/prevención & control
7.
J Neurosci ; 32(42): 14602-16, 2012 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-23077046

RESUMEN

Synaptic refinement via the elimination of inappropriate synapses and strengthening of appropriate ones is crucially important for the establishment of specific, topographic neural circuits. The mechanisms driving these processes are poorly understood, particularly concerning inhibitory projections. Here, we address the refinement of an inhibitory topographic projection in the auditory brainstem in functional and anatomical mapping studies involving patch-clamp recordings in combination with minimal and maximal stimulation, caged glutamate photolysis, and single axon tracing. We demonstrate a crucial dependency of the refinement on Ca(V)1.3 calcium channels: Ca(V)1.3(-/-) mice displayed virtually no elimination of projections up to hearing onset. Furthermore, strengthening was strongly impaired, in line with a reduced number of axonal boutons. The mediolateral topography was less precise and the shift from a mixed GABA/glycinergic to a purely glycinergic transmission before hearing onset did not occur. Together, our findings provide evidence for a Ca(V)1.3-dependent mechanism through which both inhibitory circuit formation and determination of the neurotransmitter phenotype are achieved.


Asunto(s)
Mapeo Encefálico , Tronco Encefálico/fisiología , Canales de Calcio Tipo L/fisiología , Potenciales Postsinápticos Inhibidores/fisiología , Inhibición Neural/fisiología , Sinapsis/fisiología , Animales , Mapeo Encefálico/métodos , Tronco Encefálico/metabolismo , Canales de Calcio Tipo L/deficiencia , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibición Neural/genética , Técnicas de Cultivo de Órganos , Fenotipo , Sinapsis/genética , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
8.
Nature ; 447(7148): 1081-6, 2007 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-17558391

RESUMEN

Why dopamine-containing neurons of the brain's substantia nigra pars compacta die in Parkinson's disease has been an enduring mystery. Our studies suggest that the unusual reliance of these neurons on L-type Ca(v)1.3 Ca2+ channels to drive their maintained, rhythmic pacemaking renders them vulnerable to stressors thought to contribute to disease progression. The reliance on these channels increases with age, as juvenile dopamine-containing neurons in the substantia nigra pars compacta use pacemaking mechanisms common to neurons not affected in Parkinson's disease. These mechanisms remain latent in adulthood, and blocking Ca(v)1.3 Ca2+ channels in adult neurons induces a reversion to the juvenile form of pacemaking. Such blocking ('rejuvenation') protects these neurons in both in vitro and in vivo models of Parkinson's disease, pointing to a new strategy that could slow or stop the progression of the disease.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Modelos Animales de Enfermedad , Modelos Neurológicos , Neuronas/citología , Neuronas/patología , Enfermedad de Parkinson/patología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Envejecimiento , Animales , Antiparkinsonianos/farmacología , Calcio/metabolismo , Calcio/farmacología , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Dendritas/metabolismo , Progresión de la Enfermedad , Dopamina/metabolismo , Conductividad Eléctrica , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/prevención & control , Rotenona/farmacología , Sustancia Negra/citología , Sustancia Negra/metabolismo , Sustancia Negra/patología
9.
J Physiol ; 590(24): 6327-42, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23045342

RESUMEN

Dysregulation of L-type Ca(2+) currents in sinoatrial nodal (SAN) cells causes cardiac arrhythmia. Both Ca(v)1.2 and Ca(v)1.3 channels mediate sinoatrial L-type currents. Whether these channels exhibit differences in modulation and localization, which could affect their contribution to pacemaking, is unknown. In this study, we characterized voltage-dependent facilitation (VDF) and subcellular localization of Ca(v)1.2 and Ca(v)1.3 channels in mouse SAN cells and determined how these properties of Ca(v)1.3 affect sinoatrial pacemaking in a mathematical model. Whole cell Ba(2+) currents were recorded from SAN cells from mice carrying a point mutation that renders Ca(v)1.2 channels relatively insensitive to dihydropyridine antagonists. The Ca(v)1.2-mediated current was isolated in the presence of nimodipine (1 µm), which was subtracted from the total current to yield the Ca(v)1.3 component. With strong depolarizations (+80 mV), Ca(v)1.2 underwent significantly stronger inactivation than Ca(v)1.3. VDF of Ca(v)1.3 was evident during recovery from inactivation at a time when Ca(v)1.2 remained inactivated. By immunofluorescence, Ca(v)1.3 colocalized with ryanodine receptors in sarcomeric structures while Ca(v)1.2 was largely restricted to the delimiting plasma membrane. Ca(v)1.3 VDF enhanced recovery of pacemaker activity after pauses and positively regulated pacemaking during slow heart rate in a numerical model of mouse SAN automaticity, including preferential coupling of Ca(v)1.3 to ryanodine receptor-mediated Ca(2+) release. We conclude that strong VDF and colocalization with ryanodine receptors in mouse SAN cells are unique properties that may underlie a specific role for Ca(v)1.3 in opposing abnormal slowing of heart rate.


Asunto(s)
Relojes Biológicos , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Frecuencia Cardíaca , Nodo Sinoatrial/metabolismo , Animales , Relojes Biológicos/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/genética , Señalización del Calcio/efectos de los fármacos , Simulación por Computador , Dihidropiridinas/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Potenciales de la Membrana , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Cardiovasculares , Técnicas de Placa-Clamp , Mutación Puntual , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Sarcómeros/metabolismo , Nodo Sinoatrial/efectos de los fármacos , Factores de Tiempo
10.
Circ Res ; 107(6): 800-9, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20671241

RESUMEN

RATIONALE: Myocardial infarction (MI) leads to heart failure (HF) and premature death. The respective roles of myocyte death and depressed myocyte contractility in the induction of HF after MI have not been clearly defined and are the focus of this study. OBJECTIVES: We developed a mouse model in which we could prevent depressed myocyte contractility after MI and used it to test the idea that preventing depression of myocyte Ca(2+)-handling defects could avert post-MI cardiac pump dysfunction. METHODS AND RESULTS: MI was produced in mice with inducible, cardiac-specific expression of the ß2a subunit of the L-type Ca(2+) channel. Myocyte and cardiac function were compared in control and ß2a animals before and after MI. ß2a myocytes had increased Ca(2+) current; sarcoplasmic reticulum Ca(2+) load, contraction and Ca(2+) transients (versus controls), and ß2a hearts had increased performance before MI. After MI, cardiac function decreased. However, ventricular dilation, myocyte hypertrophy and death, and depressed cardiac pump function were greater in ß2a versus control hearts after MI. ß2a animals also had poorer survival after MI. Myocytes isolated from ß2a hearts after MI did not develop depressed Ca(2+) handling, and Ca(2+) current, contractions, and Ca(2+) transients were still above control levels (before MI). CONCLUSIONS: Maintaining myocyte contractility after MI, by increasing Ca(2+) influx, depresses rather than improves cardiac pump function after MI by reducing myocyte number.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Señalización del Calcio , Contracción Miocárdica/fisiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Animales , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Señalización del Calcio/genética , Células Cultivadas , Ratones , Ratones Transgénicos , Contracción Miocárdica/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología
11.
Circ Res ; 106(7): 1285-9, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20299662

RESUMEN

RATIONALE: In arterial myocytes, membrane depolarization-induced Ca(2+) release (DICR) from the sarcoplasmic reticulum (SR) occurs through a metabotropic pathway that leads to inositol trisphosphate synthesis independently of extracellular Ca(2+) influx. Despite the fundamental functional relevance of DICR, its molecular bases are not well known. OBJECTIVE: Biophysical and pharmacological data have suggested that L-type Ca(2+) channels could be the sensors coupling membrane depolarization to SR Ca(2+) release. This hypothesis was tested using smooth muscle-selective conditional Ca(v)1.2 knockout mice. METHODS AND RESULTS: In aortic myocytes, the decrease of Ca(2+) channel density was paralleled by the disappearance of SR Ca(2+) release induced by either depolarization or Ca(2+) channel agonists. Ca(v)1.2 channel deficiency resulted in almost abolition of arterial ring contraction evoked by DICR. Ca(2+) channel-null cells showed unaltered caffeine-induced Ca(2+) release and contraction. CONCLUSION: These data suggest that Ca(v)1.2 channels are indeed voltage sensors coupled to the metabolic cascade, leading to SR Ca(2+) release. These findings support a novel, ion-independent, functional role of L-type Ca(2+) channels linked to intracellular signaling pathways in vascular myocytes.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Señalización del Calcio , Músculo Liso Vascular/metabolismo , Retículo Sarcoplasmático/metabolismo , Vasoconstricción , Animales , Aorta/metabolismo , Cafeína/farmacología , Canales de Calcio Tipo L/genética , Señalización del Calcio/efectos de los fármacos , Potenciales de la Membrana , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Potasio/metabolismo , Retículo Sarcoplasmático/efectos de los fármacos , Sodio/metabolismo , Factores de Tiempo , Vasoconstricción/efectos de los fármacos
12.
J Neurosci ; 30(2): 491-504, 2010 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20071512

RESUMEN

We studied wild-type (WT) and Cav1.3(-/-) mouse chromaffin cells (MCCs) with the aim to determine the isoform of L-type Ca(2+) channel (LTCC) and BK channels that underlie the pacemaker current controlling spontaneous firing. Most WT-MCCs (80%) were spontaneously active (1.5 Hz) and highly sensitive to nifedipine and BayK-8644 (1,4-dihydro-2,6-dimethyl-5-nitro-4-[2-(trifluoromethyl)phenyl]-3-pyridinecarboxylic acid, methyl ester). Nifedipine blocked the firing, whereas BayK-8644 increased threefold the firing rate. The two dihydropyridines and the BK channel blocker paxilline altered the shape of action potentials (APs), suggesting close coupling of LTCCs to BK channels. WT-MCCs expressed equal fractions of functionally active Cav1.2 and Cav1.3 channels. Cav1.3 channel deficiency decreased the number of normally firing MCCs (30%; 2.0 Hz), suggesting a critical role of these channels on firing, which derived from their slow inactivation rate, sizeable activation at subthreshold potentials, and close coupling to fast inactivating BK channels as determined by using EGTA and BAPTA Ca(2+) buffering. By means of the action potential clamp, in TTX-treated WT-MCCs, we found that the interpulse pacemaker current was always net inward and dominated by LTCCs. Fast inactivating and non-inactivating BK currents sustained mainly the afterhyperpolarization of the short APs (2-3 ms) and only partially the pacemaker current during the long interspike (300-500 ms). Deletion of Cav1.3 channels reduced drastically the inward Ca(2+) current and the corresponding Ca(2+)-activated BK current during spikes. Our data highlight the role of Cav1.3, and to a minor degree of Cav1.2, as subthreshold pacemaker channels in MCCs and open new interesting features about their role in the control of firing and catecholamine secretion at rest and during sustained stimulations matching acute stress.


Asunto(s)
Médula Suprarrenal/citología , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/metabolismo , Células Cromafines/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Corteza Suprarrenal/metabolismo , Animales , Fenómenos Biofísicos/efectos de los fármacos , Fenómenos Biofísicos/genética , Biofisica , Agonistas de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/genética , Células Cultivadas , Quelantes/farmacología , Células Cromafines/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Estimulación Eléctrica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Indoles/farmacología , Ionóforos/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Venenos de Araña/farmacología , Factores de Tiempo
13.
J Physiol ; 589(Pt 13): 3275-88, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21521762

RESUMEN

Mechanisms that contribute to maintaining expression of functional ion channels at relatively constant levels following perturbations of channel biosynthesis are likely to contribute significantly to the stability of electrophysiological systems in some pathological conditions. In order to examine the robustness of L-type calcium current expression, the response to changes in Ca²âº channel Cav1.2 gene dosage was studied in adult mice. Using a cardiac-specific inducible Cre recombinase system, Cav1.2 mRNA was reduced to 11 ± 1% of control values in homozygous floxed mice and the mice died rapidly (11.9 ± 3 days) after induction of gene deletion. In these homozygous knockout mice, echocardiographic analysis showed that myocardial contractility was reduced to 14 ± 1% of control values shortly before death. For these mice, no effective compensatory changes in ion channel gene expression were triggered following deletion of both Cav1.2 alleles, despite the dramatic decay in cardiac function. In contrast to the homozygote knockout mice, following knockout of only one Cav1.2 allele, cardiac function remained unchanged, as did survival.Cav1.2mRNAexpression in the left ventricle of heterozygous knockout mice was reduced to 58 ± 3% of control values and there was a 21 ± 2% reduction in Cav1.2 protein expression. There was no significant reduction in L-type Ca²âº current density in these mice. The results are consistent with a model of L-type calcium channel biosynthesis in which there are one or more saturated steps, which act to buffer changes in both total Cav1.2 protein and L-type current expression.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Regulación de la Expresión Génica/genética , Tamización de Portadores Genéticos , Miocitos Cardíacos/fisiología , Factores de Edad , Alelos , Animales , Canales de Calcio Tipo L/biosíntesis , Canales de Calcio Tipo L/genética , Femenino , Dosificación de Gen/genética , Tamización de Portadores Genéticos/métodos , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación/genética
14.
Hippocampus ; 21(2): 133-41, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20014384

RESUMEN

Trains of action potentials in CA1 pyramidal neurons are followed by a prolonged calcium-dependent postburst afterhyperpolarization (AHP) that serves to limit further firing to a sustained depolarizing input. A reduction in the AHP accompanies acquisition of several types of learning and increases in the AHP are correlated with age-related cognitive impairment. The AHP develops primarily as the result of activation of outward calcium-activated potassium currents; however, the precise source of calcium for activation of the AHP remains unclear. There is substantial experimental evidence suggesting that calcium influx via voltage-gated L-type calcium channels (L-VGCCs) contributes to the generation of the AHP. Two L-VGCC subtypes are predominately expressed in the hippocampus, Ca(V) 1.2 and Ca(V) 1.3; however, it is not known which L-VGCC subtype is involved in generation of the AHP. This ambiguity is due in large part to the fact that at present there are no subunit-specific agonists or antagonists. Therefore, using mice in which the gene encoding Ca(V) 1.2 or Ca(V) 1.3 was deleted, we sought to determine the impact of alterations in levels of these two L-VCGG subtypes on neuronal excitability. No differences in any AHP measure were seen between neurons from Ca(V) 1.2 knockout mice and controls. However, the total area of the AHP was significantly smaller in neurons from Ca(V) 1.3 knockout mice as compared with neurons from wild-type controls. A significant reduction in the amplitude of the AHP was also seen at the 1 s time point in neurons from Ca(V) 1.3 knockout mice as compared with those from controls. Reductions in both the area and 1 s amplitude suggest the involvement of calcium influx via Ca(V) 1.3 in the slow AHP (sAHP). Thus, the results of our study demonstrate that deletion of Ca(V) 1.3, but not Ca(V) 1.2, significantly impacts the generation of the sAHP.


Asunto(s)
Región CA1 Hipocampal/fisiología , Canales de Calcio Tipo L/deficiencia , Señalización del Calcio/fisiología , Potenciales de Acción , Animales , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/fisiología , Fenómenos Electrofisiológicos , Femenino , Técnicas In Vitro , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Técnicas de Placa-Clamp
15.
J Cardiovasc Electrophysiol ; 22(8): 922-30, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21352396

RESUMEN

INTRODUCTION: Congenital heart block (CHB) is a passively acquired autoimmune disease considered to be due to the transfer of maternal autoantibodies, anti-SSA/Ro -SSB/La, to the fetus resulting in atrioventricular (AV) block and sinus bradycardia. We previously established a murine model for CHB where pups born to immunized wild-type (WT) mothers exhibited electrocardiographic abnormalities similar to those seen in CHB and demonstrated inhibition of L-type Ca channels (LTCCs) by maternal antibodies. Here, we hypothesize that overexpression of LTCC should rescue, whereas knockout of LTCC should worsen the electrocardiographic abnormalities in mice. METHODS AND RESULTS: Transgenic (TG) mice were immunized with SSA/Ro and SSB/La antigens. Pups born to immunized WT mothers had significantly greater sinus bradycardia and AV block compared to pups from nonimmunized WT. TG pups overexpressing LTCC had significantly less sinus bradycardia and AV block compared to their non-TG littermates and to pups born to immunized WT mothers. All LTCC knockout pups born to immunized mothers had sinus bradycardia, advanced degree of AV block, and decreased fetal parity. No sinus bradycardia or AV block were manifested in pups from control nonimmunized WT mothers. IgG from mothers with CHB children, but not normal IgG, completely inhibited intracellular Ca transient ([Ca](i)T) amplitude. CONCLUSIONS: Cardiac-specific overexpression of LTCC significantly reduced the incidence of AV block and sinus bradycardia in pups exposed to anti-SSA/Ro -SSB/La autoantibodies, whereas exposure of LTCC knockout pups to these autoantibodies significantly worsened the electrocardiographic abnormalities. These findings support the hypothesis that maternal antibodies inhibit LTCC and [Ca](i)T thus contributing to the development of CHB. Altogether, the results are relevant to the development of novel therapies for CHB.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Electrocardiografía , Bloqueo Cardíaco/congénito , Animales , Autoantígenos/inmunología , Canales de Calcio Tipo L/genética , Electrocardiografía/métodos , Femenino , Bloqueo Cardíaco/genética , Bloqueo Cardíaco/inmunología , Bloqueo Cardíaco/fisiopatología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Embarazo , Ribonucleoproteínas/inmunología , Antígeno SS-B
16.
Sci Rep ; 11(1): 15146, 2021 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-34312410

RESUMEN

Retinitis Pigmentosa is a genetically heterogeneous, degenerative retinal disorder characterized by gradual dysfunction and death of photoreceptors, first rods and later cones, and progressive blindness. Studies suggested that application of L-type calcium channel blockers rescues photoreceptors in paradigms related to Ca2+ overflow. To investigate whether Cav1.3 L-type channels have protective effects in the retina, we established a new mouse model by crossing rd10, modeling autosomal-recessive RP, with Cav1.3 deficient mice (rd10/Cav1.3KO). Our immunohistochemical analyses revealed an influence of Cav1.3 channels on the degenerative process of photoreceptors. The absence of Cav1.3 delayed the centre-to-periphery degeneration of rods indicated by a significantly higher number of photoreceptor rows and, consequently, of cones. In accordance with a preserved number of cones we observed a regular row of cone somas in rd10/Cav1.3-KO retinas. Surviving rod photoreceptors maintained synaptic contacts with rod bipolar cells. However, the delay in degeneration was only observed up to postnatal day 45. Although we observed a reduction in the spontaneous oscillatory retinal activity during multielectrode array analyses, measurable functional preservation was lacking in behavioural tests. In conclusion, Cav1.3 channels contribute to photoreceptor degeneration in rd10 retinas but photoreceptor temporary rescue might rather be achieved indirectly through other retinal cell layers.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/deficiencia , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/patología
17.
J Neurosci ; 29(34): 10730-40, 2009 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-19710324

RESUMEN

Hearing relies on Ca(2+) influx-triggered exocytosis in cochlear inner hair cells (IHCs). Here we studied the role of the Ca(2+) channel subunit Ca(V)beta(2) in hearing. Of the Ca(V)beta(1-4) mRNAs, IHCs predominantly contained Ca(V)beta(2). Hearing was severely impaired in mice lacking Ca(V)beta(2) in extracardiac tissues (Ca(V)beta(2)(-/-)). This involved deficits in cochlear amplification and sound encoding. Otoacoustic emissions were reduced or absent in Ca(V)beta(2)(-/-) mice, which showed strongly elevated auditory thresholds in single neuron recordings and auditory brainstem response measurements. Ca(V)beta(2)(-/-) IHCs showed greatly reduced exocytosis (by 68%). This was mostly attributable to a decreased number of membrane-standing Ca(V)1.3 channels. Confocal Ca(2+) imaging revealed presynaptic Ca(2+) microdomains albeit with much lower amplitudes, indicating synaptic clustering of fewer Ca(V)1.3 channels. The coupling of the remaining Ca(2+) influx to IHC exocytosis appeared unaffected. Extracellular recordings of sound-evoked spiking in the cochlear nucleus and auditory nerve revealed reduced spike rates in the Ca(V)beta(2)(-/-) mice. Still, sizable onset and adapted spike rates were found during suprathreshold stimulation in Ca(V)beta(2)(-/-) mice. This indicated that residual synaptic sound encoding occurred, although the number of presynaptic Ca(V)1.3 channels and exocytosis were reduced to one-third. The normal developmental upregulation, clustering, and gating of large-conductance Ca(2+) activated potassium channels in IHCs were impaired in the absence of Ca(V)beta(2). Moreover, we found the developmental efferent innervation to persist in Ca(V)beta(2)-deficient IHCs. In summary, Ca(V)beta(2) has an essential role in regulating the abundance and properties of Ca(V)1.3 channels in IHCs and, thereby, is critical for IHC development and synaptic encoding of sound.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Células Ciliadas Auditivas Internas/fisiología , Estimulación Acústica/métodos , Factores de Edad , Animales , Animales Recién Nacidos , Apamina/farmacología , Calcio/metabolismo , Canales de Calcio Tipo L/deficiencia , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Cesio/farmacología , Quelantes/farmacología , Cloruros/farmacología , Ácido Egtácico/farmacología , Estimulación Eléctrica/métodos , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Exocitosis/efectos de los fármacos , Células Ciliadas Auditivas Internas/efectos de los fármacos , Técnicas In Vitro , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Emisiones Otoacústicas Espontáneas/genética , Parvalbúminas/metabolismo , Técnicas de Placa-Clamp/métodos
18.
FEBS Lett ; 594(17): 2914-2922, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32484574

RESUMEN

Cav2.2 N-type voltage-dependent Ca2+ channel (VDCC) expressed in neurons is known to be essential for neurotransmitter release. We have shown previously that this channel is also expressed in nonexcitable microglia and plays pivotal roles in microglial functions. Here, we have examined the effects of microglia-specific knockdown (KD) of Cav2.2 channel in a mouse model of Parkinson's disease (PD). We found that the KD of Cav2.2 channel reduces the accumulation of microglia in the substantia nigra and ameliorates the behavioral deficits in PD model mice. These results are in marked contrast with those found in microglia-specific KD of Cav1.2 L-type channel, where exacerbated symptoms are observed. Our results suggest that blockade of microglial Cav2.2 N-type VDCC is beneficial for the treatment of PD.


Asunto(s)
Canales de Calcio Tipo N/genética , Neuronas Dopaminérgicas/metabolismo , Microglía/metabolismo , Trastornos Parkinsonianos/genética , Sustancia Negra/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/administración & dosificación , Animales , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo N/deficiencia , Recuento de Células , Muerte Celular/genética , Neuronas Dopaminérgicas/patología , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Desempeño Psicomotor/fisiología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Sustancia Negra/patología , Tamoxifeno/farmacología
19.
Circ Res ; 101(2): 166-75, 2007 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-17556655

RESUMEN

Calcium cycling figures prominently in excitation-contraction coupling and in various signaling cascades involved in the development of left ventricular hypertrophy. We hypothesized that genetic suppression of the L-type calcium channel accessory beta-subunit would modulate calcium current and suppress cardiac hypertrophy. A short hairpin RNA template sequence capable of mediating the knockdown of the L-type calcium channel accessory beta-subunit gene was incorporated into a lentiviral vector (PPT.CG.H1.beta(2)). Transduction of ventricular myocytes in vivo with the active short hairpin RNA partially inhibited the L-type calcium current. In neonatal rat cardiomyocytes, L-type calcium channel accessory beta-subunit gene knockdown reduced calcium transient amplitude. Similarly, [(3)H]leucine incorporation was attenuated in PPT.CG.H1.beta(2)-transduced neonatal rat cardiomyocytes compared with nonsilencing controls in a phenylephrine-induced hypertrophy model. In vivo gene transfer attenuated the hypertrophic response in an aortic-banded rat model of left ventricular hypertrophy, with reduced left ventricular wall thickness and heart weight/body weight ratios in PPT.CG.H1.beta(2)-injected rats at four weeks post transduction. Fractional shortening was preserved in rats treated with PPT.CG.H1.beta(2). These findings indicate that knockdown of L-type calcium channel accessory beta-subunit is capable of attenuating the hypertrophic response both in vitro and in vivo without compromising systolic performance. Suppression of the calcium channel beta subunit may represent a novel and useful therapeutic strategy for left ventricular hypertrophy.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/metabolismo , Silenciador del Gen , Terapia Genética , Hipertrofia Ventricular Izquierda/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Humanos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/terapia , Lentivirus , Miocitos Cardíacos/patología , Tamaño de los Órganos/genética , Subunidades de Proteína/deficiencia , Subunidades de Proteína/metabolismo , Ratas , Transducción Genética
20.
Circ Res ; 100(1): 112-20, 2007 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-17110593

RESUMEN

Cytoskeletal proteins are known to sculpt the structural architecture of cells. However, their role as bridges linking the functional crosstalk of different ion channels is unknown. Here, we demonstrate that a small conductance Ca(2+)-activated K(+) channels (SK2 channel), present in a variety of cells, where they integrate changes in intracellular Ca(2+) concentration [Ca(2+)(i)] with changes in K(+) conductance and membrane potential, associate with L-type Ca(2+) channels; Ca(v)1.3 and Ca(v)1.2 through a physical bridge, alpha-actinin2 in cardiac myocytes. SK2 channels do not physically interact with L-type Ca(2+) channels, instead, the 2 channels colocalize via their interaction with alpha-actinin2 cytoskeletal protein. The association of SK2 channel with alpha-actinin2 localizes the channel to the entry of external Ca(2+) source, which regulate the channel function. Furthermore, we demonstrated that the functions of SK2 channels in atrial myocytes are critically dependent on the normal expression of Ca(v)1.3 Ca(2+) channels. Null deletion of Ca(v)1.3 channel results in abnormal function of SK2 channel and prolongation of repolarization and atrial arrhythmias. Our study provides insight into the molecular mechanisms of the coupling of SK2 channel with voltage-gated Ca(2+) channel, and represents the first report linking the coupling of 2 different types of ion channels via cytoskeletal proteins.


Asunto(s)
Actinina/metabolismo , Canales de Calcio Tipo L/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Animales , Canales de Calcio Tipo L/deficiencia , Línea Celular , Conductividad Eléctrica , Atrios Cardíacos , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Miocardio/citología , Miocardio/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Distribución Tisular , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA