Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
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
2.
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
3.
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
4.
Sci Rep ; 9(1): 9138, 2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31235768

RESUMEN

Cav1.2 channels are an L-type voltage-dependent Ca2+ channel, which is specifically blocked by calcium antagonists. Voltage-dependent Ca2+ channels are generally considered to be functional only in excitable cells like neurons and muscle cells, but recently they have been reported to also be functional in non-excitable cells like microglia, which are key players in the innate immune system and have been shown to be involved in the pathophysiology of Parkinson's disease. Here, we show that Cav1.2 channels are expressed in microglia, and that calcium antagonists enhanced the neuroinflammatory M1 transition and inhibited neuroprotective M2 transition of microglia in vitro. Moreover, intensive degeneration of dopaminergic neurons and accompanying behavioural deficits were observed in microglia-specific Cav1.2 knockdown mice intoxicated with MPTP, a neurotoxin that induces Parkinson's disease-like symptoms, suggesting detrimental effects of microglial Cav1.2 blockade on Parkinson's disease. Therefore, microglial Cav1.2 channel may have neuroprotective roles under physiological conditions and may also contribute to recovery from disease conditions.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Técnicas de Silenciamiento del Gen , Microglía/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Ratones , Microglía/patología , Enfermedad de Parkinson/metabolismo
5.
Behav Brain Res ; 367: 35-52, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30902660

RESUMEN

The risk gene CACNA1C is strongly implicated in the etiology of all major psychiatric disorders, such as depressive disorder, bipolar disorder, autism spectrum disorder, and schizophrenia. These disorders feature high levels of comorbidity and share an overlap of symptoms; in particular, deficits in social functioning are common. Intriguingly, sex-dependent effects of CACNA1C single nucleotide polymorphisms on prevalence, health outcomes, and psychological traits have been reported, typically suggesting that women are more affected by CACNA1C mutations than men. In rodents, genetic modifications specifically targeting Cacna1c have repeatedly been linked to deficits in social behavior in male mice and rats but many studies neglect the sex-dependent effects observed in humans. Our study focused on the role of Cacna1c in regulating social behavior and communication in adult female rats. We compared social and non-social behavior together with concomitant emission of pro-social 50-kHz ultrasonic vocalizations (USV) associated with positive affect in constitutive heterozygous (Cacna1c+/-) rats to wildtype (Cacna1c+/+) littermate controls. Our results indicate that partial Cacna1c depletion leads to strongly reduced emission of 50-kHz USV and mild social deficits during female direct reciprocal social interaction. Detailed temporal analyses revealed most prominent reductions of 50-kHz USV during non-social behavior, suggesting that reduced positive affect occurs in a social context in Cacna1c+/- rats but is not specifically linked to social behavior. Finally, we observed increased self-grooming behavior in Cacna1c+/- rats, consistent with an autism-like phenotype. Our findings in rats thus support a role of Cacna1c in regulating behavioral phenotypes with relevance for several neuropsychiatric disorders.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Relaciones Interpersonales , Conducta Social , Vocalización Animal/fisiología , Animales , Canales de Calcio Tipo L/deficiencia , Modelos Animales de Enfermedad , Femenino , Trastornos Mentales/genética , Ratas , Ratas Sprague-Dawley
6.
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
7.
Cell Rep ; 23(13): 3891-3904, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29949772

RESUMEN

Formation of synapses between motor neurons and muscles is initiated by clustering of acetylcholine receptors (AChRs) in the center of muscle fibers prior to nerve arrival. This AChR patterning is considered to be critically dependent on calcium influx through L-type channels (CaV1.1). Using a genetic approach in mice, we demonstrate here that either the L-type calcium currents (LTCCs) or sarcoplasmic reticulum (SR) calcium release is necessary and sufficient to regulate AChR clustering at the onset of neuromuscular junction (NMJ) development. The combined lack of both calcium signals results in loss of AChR patterning and excessive nerve branching. In the absence of SR calcium release, the severity of synapse formation defects inversely correlates with the magnitude of LTCCs. These findings highlight the importance of activity-dependent calcium signaling in early neuromuscular junction formation and indicate that both LTCC and SR calcium release individually support proper innervation of muscle by regulating AChR patterning and motor axon outgrowth.


Asunto(s)
Calcio/metabolismo , Unión Neuromuscular/fisiología , Proyección Neuronal/fisiología , Receptores Colinérgicos/metabolismo , Animales , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Diafragma/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Fetal , Ratones , Ratones Noqueados , Neuronas Motoras/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
8.
PLoS One ; 13(3): e0194428, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29543863

RESUMEN

In skeletal muscle the coordinated actions of two mechanically coupled Ca2+ channels-the 1,4-dihydropyridine receptor (Cav1.1) and the type 1 ryanodine receptor (RYR1)-underlie the molecular mechanism of rapid cytosolic [Ca2+] increase leading to contraction. While both [Ca2+]i and contractile activity have been implicated in the regulation of myogenesis, less is known about potential specific roles of Cav1.1 and RYR1 in skeletal muscle development. In this study, we analyzed the histology and the transcriptomic changes occurring at E14.5 -the end of primary myogenesis and around the onset of intrauterine limb movement, and at E18.5 -the end of secondary myogenesis, in WT, RYR1-/-, and Cav1.1-/- murine limb skeletal muscle. At E14.5 the muscle histology of both mutants exhibited initial alterations, which became much more severe at E18.5. Immunohistological analysis also revealed higher levels of activated caspase-3 in the Cav1.1-/- muscles at E14.5, indicating an increase in apoptosis. With WT littermates as controls, microarray analyses identified 61 and 97 differentially regulated genes (DEGs) at E14.5, and 493 and 1047 DEGs at E18.5, in RYR1-/- and Cav1.1-/- samples, respectively. Gene enrichment analysis detected no overlap in the affected biological processes and pathways in the two mutants at E14.5, whereas at E18.5 there was a significant overlap of DEGs in both mutants, affecting predominantly processes linked to muscle contraction. Moreover, the E18.5 vs. E14.5 comparison revealed multiple genotype-specific DEGs involved in contraction, cell cycle and miRNA-mediated signaling in WT, neuronal and bone development in RYR1-/-, and lipid metabolism in Cav1.1-/- samples. Taken together, our study reveals discrete changes in the global transcriptome occurring in limb skeletal muscle from E14.5 to E18.5 in WT, RYR1-/- and Cav1.1-/- mice. Our results suggest distinct functional roles for RYR1 and Cav1.1 in skeletal primary and secondary myogenesis.


Asunto(s)
Canales de Calcio Tipo L/genética , Regulación del Desarrollo de la Expresión Génica , Músculo Esquelético/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Transcriptoma , Animales , Canales de Calcio Tipo L/deficiencia , Ontología de Genes , Miembro Posterior/embriología , Miembro Posterior/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos/genética , Músculo Esquelético/embriología , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Factores de Tiempo
9.
Channels (Austin) ; 12(1): 51-57, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28949795

RESUMEN

Recently, we reported that homozygous deletion of alternative exon 33 of CaV1.2 calcium channel in the mouse resulted in ventricular arrhythmias arising from increased CaV1.2Δ33 ICaL current density in the cardiomyocytes. We wondered whether heterozygous deletion of exon 33 might produce cardiac phenotype in a dose-dependent manner, and whether the expression levels of RNA splicing factors known to regulate alternative splicing of exon 33 might change in human heart failure. Unexpectedly, we found that exon 33+/- cardiomyocytes showed similar CaV1.2 channel properties as wild-type cardiomyocyte, even though CaV1.2Δ33 channels exhibit a gain-in-function. In human hearts, we found that the mRNA level of splicing factor Rbfox1, but not Rbfox2, was downregulated in dilated cardiomyopathy, and CACNA1C mRNA level was dramatically decreased in the both of dilated and ischemic cardiomyopathy. These data imply Rbfox1 may be involved in the development of cardiomyopathies via regulating the alternative splicing of CaV1.2 exon 33. (149 words).


Asunto(s)
Canales de Calcio Tipo L/genética , Exones/genética , Insuficiencia Cardíaca/genética , Factores de Empalme de ARN/genética , Animales , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/metabolismo , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Empalme de ARN/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
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
11.
Brain Struct Funct ; 223(1): 111-130, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28733833

RESUMEN

The phenotype of calcium channel subunit (CACN) α2δ3 knockout (KO) mice includes sensory cross-activation and deficient pain perception. Sensory cross-activation defines the activation of a sensory cortical region by input from another modality due to reorganization in the brain such as after sensory loss. To obtain mechanistic insight into both phenomena, we employed a comprehensive battery of neuroanatomical techniques. While CACNα2δ3 was ubiquitously expressed in wild-type mice, it was absent in α2δ3 KO animals. Immunostaining of α1A, α1B, and α1E revealed upregulation of N-type and R-type, but not P/Q-type Cav2 channels in cortical neurons of CACNα2δ3 KO mice. Compared to wild-type mice, axonal processes in somatosensory cortex were enhanced, and dendritic processes reduced, in CACNα2δ3 KO mice. Immunohistochemical and MRI analyses, investigating morphology, thalamocortical and intra-/intercortical trajectories, revealed a disparity between projection and commissural fibers with reduction of the number of spatial specificity of thalamocortical projections. L1cam staining revealed wide-ranging projections of thalamocortical fibers reaching both somatosensory/motor and visual cortical areas. Activation (c-fos+) of excitatory and inhibitory neurons suggested that deficient pain perception in α2δ3 KO mice is unlikely to result from cortical disinhibition. Collectively, our data demonstrate that knock out of CACN α2δ3 results in some structural abnormalities whose functional implications converge to dedifferentiation of sensory activation.


Asunto(s)
Encéfalo/patología , Canales de Calcio Tipo L/deficiencia , Regulación de la Expresión Génica/genética , Percepción del Dolor/fisiología , Trastornos Somatosensoriales/genética , Trastornos Somatosensoriales/patología , Vibrisas/inervación , Acetiltransferasas/metabolismo , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/ultraestructura , Mapeo Encefálico , Canales de Calcio Tipo L/genética , Glutamato Descarboxilasa/metabolismo , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vías Nerviosas/diagnóstico por imagen , Proteínas de Neurofilamentos/metabolismo , Dimensión del Dolor , Estimulación Física , Proteínas Proto-Oncogénicas c-fos/metabolismo
12.
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
13.
eNeuro ; 3(6)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27957527

RESUMEN

L-type voltage-gated calcium channels (LVGCCs) have been implicated in various forms of learning, memory, and synaptic plasticity. Within the hippocampus, the LVGCC subtype, CaV1.2 is prominently expressed throughout the dentate gyrus. Despite the apparent high levels of CaV1.2 expression in the dentate gyrus, the role of CaV1.2 in hippocampal- and dentate gyrus-associated forms of learning remain unknown. To address this question, we examined alternate forms of hippocampal-dependent associative and spatial memory in mice lacking the mouse ortholog of CACNA1C (Cacna1c), which encodes CaV1.2, with dentate gyrus function implicated in difficult forms of each task. We found that while the deletion of CaV1.2 did not impair the acquisition of fear of a conditioned context, mice lacking CaV1.2 exhibited deficits in the ability to discriminate between two contexts, one in which the mice were conditioned and one in which they were not. Similarly, CaV1.2 knock-out mice exhibited normal acquisition and recall of the location of the hidden platform in a standard Morris water maze, but were unable to form a memory of the platform location when the task was made more difficult by restricting the number of available spatial cues. Within the dentate gyrus, pan-neuronal deletion of CaV1.2 resulted in decreased cell proliferation and the numbers of doublecortin-positive adult-born neurons, implicating CaV1.2 in adult neurogenesis. These results suggest that CaV1.2 is important for dentate gyrus-associated tasks and may mediate these forms of learning via a role in adult neurogenesis and cell proliferation within the dentate gyrus.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Giro Dentado/metabolismo , Memoria/fisiología , Neurogénesis/fisiología , Neuronas/metabolismo , Células Madre Adultas/metabolismo , Células Madre Adultas/patología , Animales , Canales de Calcio Tipo L/genética , Condicionamiento Clásico/fisiología , Giro Dentado/patología , Discriminación en Psicología/fisiología , Proteínas de Dominio Doblecortina , Miedo/fisiología , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuronas/patología , Neuropéptidos/metabolismo , Percepción Espacial/fisiología
14.
Sci Rep ; 5: 13688, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26381090

RESUMEN

The preferential degeneration of Substantia nigra dopamine midbrain neurons (SN DA) causes the motor-symptoms of Parkinson's disease (PD). Voltage-gated L-type calcium channels (LTCCs), especially the Cav1.3-subtype, generate an activity-related oscillatory Ca(2+) burden in SN DA neurons, contributing to their degeneration and PD. While LTCC-blockers are already in clinical trials as PD-therapy, age-dependent functional roles of Cav1.3 LTCCs in SN DA neurons remain unclear. Thus, we analysed juvenile and adult Cav1.3-deficient mice with electrophysiological and molecular techniques. To unmask compensatory effects, we compared Cav1.3 KO mice with pharmacological LTCC-inhibition. LTCC-function was not necessary for SN DA pacemaker-activity at either age, but rather contributed to their pacemaker-precision. Moreover, juvenile Cav1.3 KO but not WT mice displayed adult wildtype-like, sensitised inhibitory dopamine-D2-autoreceptor (D2-AR) responses that depended upon both, interaction of the neuronal calcium sensor NCS-1 with D2-ARs, and on voltage-gated T-type calcium channel (TTCC) activity. This functional KO-phenotype was accompanied by cell-specific up-regulation of NCS-1 and Cav3.1-TTCC mRNA. Furthermore, in wildtype we identified an age-dependent switch of TTCC-function from contributing to SN DA pacemaker-precision in juveniles to pacemaker-frequency in adults. This novel interplay of Cav1.3 L-type and Cav3.1 T-type channels, and their modulation of SN DA activity-pattern and D2-AR-sensitisation, provide new insights into flexible age- and calcium-dependent activity-control of SN DA neurons and its pharmacological modulation.


Asunto(s)
Autorreceptores/metabolismo , Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Neuronas Dopaminérgicas/metabolismo , Receptores de Dopamina D2/metabolismo , Sustancia Negra/metabolismo , Factores de Edad , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Regulación de la Expresión Génica , Masculino , Potenciales de la Membrana , Ratones , Ratones Noqueados , Proteínas Sensoras del Calcio Neuronal/genética , Proteínas Sensoras del Calcio Neuronal/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo
15.
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
16.
PLoS One ; 8(10): e78598, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24205277

RESUMEN

Voltage-gated Ca(V)2.1 (P/Q-type) Ca²âº channels located at the presynaptic membrane are known to control a multitude of Ca²âº-dependent cellular processes such as neurotransmitter release and synaptic plasticity. Our knowledge about their contributions to complex cognitive functions, however, is restricted by the limited adequacy of existing transgenic Ca(V)2.1 mouse models. Global Ca(V)2.1 knock-out mice lacking the α1 subunit Cacna1a gene product exhibit early postnatal lethality which makes them unsuitable to analyse the relevance of Ca(V)2.1 Ca²âº channels for complex behaviour in adult mice. Consequently we established a forebrain specific Ca(V)2.1 knock-out model by crossing mice with a floxed Cacna1a gene with mice expressing Cre-recombinase under the control of the NEX promoter. This novel mouse model enabled us to investigate the contribution of Ca(V)2.1 to complex cognitive functions, particularly learning and memory. Electrophysiological analysis allowed us to test the specificity of our conditional knock-out model and revealed an impaired synaptic transmission at hippocampal glutamatergic synapses. At the behavioural level, the forebrain-specific Ca(V)2.1 knock-out resulted in deficits in spatial learning and reference memory, reduced recognition memory, increased exploratory behaviour and a strong attenuation of circadian rhythmicity. In summary, we present a novel conditional Ca(V)2.1 knock-out model that is most suitable for analysing the in vivo functions of Ca(V)2.1 in the adult murine forebrain.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Canales de Calcio Tipo L/genética , Cognición/fisiología , Técnicas de Inactivación de Genes , Prosencéfalo/metabolismo , Animales , Conducta Animal/fisiología , Aprendizaje por Laberinto/fisiología , Ratones , Reconocimiento en Psicología/fisiología
17.
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
18.
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
19.
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
20.
J Clin Invest ; 122(1): 280-90, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22133878

RESUMEN

Antagonists of L-type Ca²âº channels (LTCCs) have been used to treat human cardiovascular diseases for decades. However, these inhibitors can have untoward effects in patients with heart failure, and their overall therapeutic profile remains nebulous given differential effects in the vasculature when compared with those in cardiomyocytes. To investigate this issue, we examined mice heterozygous for the gene encoding the pore-forming subunit of LTCC (calcium channel, voltage-dependent, L type, α1C subunit [Cacna1c mice; referred to herein as α1C⁻/⁺ mice]) and mice in which this gene was loxP targeted to achieve graded heart-specific gene deletion (termed herein α1C-loxP mice). Adult cardiomyocytes from the hearts of α1C⁻/⁺ mice at 10 weeks of age showed a decrease in LTCC current and a modest decrease in cardiac function, which we initially hypothesized would be cardioprotective. However, α1C⁻/⁺ mice subjected to pressure overload stimulation, isoproterenol infusion, and swimming showed greater cardiac hypertrophy, greater reductions in ventricular performance, and greater ventricular dilation than α1C⁺/⁺ controls. The same detrimental effects were observed in α1C-loxP animals with a cardiomyocyte-specific deletion of one allele. More severe reductions in α1C protein levels with combinatorial deleted alleles produced spontaneous cardiac hypertrophy before 3 months of age, with early adulthood lethality. Mechanistically, our data suggest that a reduction in LTCC current leads to neuroendocrine stress, with sensitized and leaky sarcoplasmic reticulum Ca²âº release as a compensatory mechanism to preserve contractility. This state results in calcineurin/nuclear factor of activated T cells signaling that promotes hypertrophy and disease.


Asunto(s)
Canales de Calcio Tipo L/deficiencia , Cardiomegalia/etiología , Insuficiencia Cardíaca/etiología , Animales , Calcineurina/metabolismo , Canales de Calcio Tipo L/genética , Señalización del Calcio , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Técnicas de Silenciamiento del Gen , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Heterocigoto , Homocigoto , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Sistemas Neurosecretores/metabolismo , Estrés Fisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA