Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cereb Cortex ; 30(3): 1393-1406, 2020 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-31504258

RESUMO

Neurotransmitters can be released either synchronously or asynchronously with respect to action potential timing. Synapsins (Syns) are a family of synaptic vesicle (SV) phosphoproteins that assist gamma-aminobutyric acid (GABA) release and allow a physiological excitation/inhibition balance. Consistently, deletion of either or both Syn1 and Syn2 genes is epileptogenic. In this work, we have characterized the effect of SynI knockout (KO) in the regulation of GABA release dynamics. Using patch-clamp recordings in hippocampal slices, we demonstrate that the lack of SynI impairs synchronous GABA release via a reduction of the readily releasable SVs and, in parallel, increases asynchronous GABA release. The effects of SynI deletion on synchronous GABA release were occluded by ω-AgatoxinIVA, indicating the involvement of P/Q-type Ca2+channel-expressing neurons. Using in situ hybridization, we show that SynI is more expressed in parvalbumin (PV) interneurons, characterized by synchronous release, than in cholecystokinin or SOM interneurons, characterized by a more asynchronous release. Optogenetic activation of PV and SOM interneurons revealed a specific reduction of synchronous release in PV/SynIKO interneurons associated with an increased asynchronous release in SOM/SynIKO interneurons. The results demonstrate that SynI is differentially expressed in interneuron subpopulations, where it boosts synchronous and limits asynchronous GABA release.


Assuntos
Interneurônios/fisiologia , Sinapsinas/fisiologia , Transmissão Sináptica , Ácido gama-Aminobutírico/fisiologia , Animais , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Hipocampo/fisiologia , Potenciais Pós-Sinápticos Inibidores , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal , Sinapsinas/genética
2.
J Neurosci ; 33(8): 3633-45, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23426690

RESUMO

P/Q-type voltage-gated calcium channels (Ca(v)2.1) play critical presynaptic and postsynaptic roles throughout the nervous system and have been implicated in a variety of neurological disorders. Here we report that mice with a genetic ablation of the Ca(v)2.1 pore-forming α(1A) subunit (α(1A)⁻/⁻) encoded by CACNA1a (Jun et al., 1999) suffer during postnatal development from increasing breathing disturbances that lead ultimately to death. Breathing abnormalities include decreased minute ventilation and a specific loss of sighs, which was associated with lung atelectasis. Similar respiratory alterations were preserved in the isolated in vitro brainstem slice preparation containing the pre-Bötzinger complex. The loss of Ca(v)2.1 was associated with an alteration in the functional dependency on N-type calcium channels (Ca(v)2.2). Blocking N-type calcium channels with conotoxin GVIA had only minor effects on respiratory activity in slices from control (CT) littermates, but abolished respiratory activity in all slices from α(1A)⁻/⁻ mice. The amplitude of evoked EPSPs was smaller in inspiratory neurons from α(1A)⁻/⁻ mice compared with CTs. Conotoxin GVIA abolished all EPSPs in inspiratory neurons from α(1A)⁻/⁻ mice, while the EPSP amplitude was reduced by only 30% in CT mice. Moreover, neuromodulation was significantly altered as muscarine abolished respiratory network activity in α(1A)⁻/⁻ mice but not in CT mice. We conclude that excitatory synaptic transmission dependent on N-type and P/Q-type calcium channels is required for stable breathing and sighing. In the absence of P/Q-type calcium channels, breathing, sighing, and neuromodulation are severely compromised, leading to early mortality.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Mecânica Respiratória/fisiologia , Animais , Animais Recém-Nascidos , Tronco Encefálico/fisiologia , Canais de Cálcio Tipo N/deficiência , Canais de Cálcio Tipo P/deficiência , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/deficiência , Canais de Cálcio Tipo Q/fisiologia , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Técnicas de Cultura de Órgãos , Mecânica Respiratória/genética
3.
J Neurosci ; 33(12): 5162-74, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23516282

RESUMO

Ataxia, episodic dyskinesia, and thalamocortical seizures are associated with an inherited loss of P/Q-type voltage-gated Ca(2+) channel function. P/Q-type channels are widely expressed throughout the neuraxis, obscuring identification of the critical networks underlying these complex neurological disorders. We showed recently that the conditional postnatal loss of P/Q-type channels in cerebellar Purkinje cells (PCs) in mice (purky) leads to these aberrant phenotypes, suggesting that intrinsic alteration in PC output is a sufficient pathogenic factor for disease initiation. The question arises whether P/Q-type channel deletion confined to a single upstream cerebellar synapse might induce the pathophysiological abnormality of genomically inherited P/Q-type channel disorders. PCs integrate two excitatory inputs, climbing fibers from inferior olive and parallel fibers (PFs) from granule cells (GCs) that receive mossy fiber (MF) input derived from precerebellar nuclei. In this study, we introduce a new mouse model with a selective knock-out of P/Q-type channels in rhombic-lip-derived neurons including the PF and MF pathways (quirky). We found that in quirky mice, PF-PC synaptic transmission is reduced during low-frequency stimulation. Using focal light stimulation of GCs that express optogenetic light-sensitive channels, channelrhodopsin-2, we found that modulation of PC firing via GC input is reduced in quirky mice. Phenotypic analysis revealed that quirky mice display ataxia, dyskinesia, and absence epilepsy. These results suggest that developmental alteration of patterned input confined to only one of the main afferent cerebellar excitatory synaptic pathways has a significant role in generating the neurological phenotype associated with the global genomic loss of P/Q-type channel function.


Assuntos
Ataxia/fisiopatologia , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Cerebelo/anormalidades , Epilepsia Tipo Ausência/fisiopatologia , Células de Purkinje/fisiologia , Animais , Ataxia/genética , Ataxia/patologia , Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo Q/genética , Cerebelo/fisiopatologia , Eletroencefalografia , Epilepsia Tipo Ausência/genética , Epilepsia Tipo Ausência/patologia , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes Neurológicos , Fenótipo , Transmissão Sináptica/fisiologia , Gravação de Videoteipe
4.
J Neurosci ; 33(17): 7384-92, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23616544

RESUMO

A long-held tenet of neuromuscular transmission is that calcium-dependent neurotransmitter release is mediated by N-type calcium channels in frog but P/Q-type channels in mammals. The N-type assignment in frog is based principally on pharmacological sensitivity to ω-conotoxin GVIA. Our studies show that zebrafish neuromuscular transmission is also sensitive to ω-conotoxin GVIA. However, positional cloning of a mutant line with compromised neuromuscular function identified a mutation in a P/Q- rather than N-type channel. Cloning and heterologous expression of this P/Q-type channel confirmed a block by ω-conotoxin GVIA raising the likelihood that all vertebrates, including frog, use the P/Q-type calcium channel for neuromuscular transmission. In addition, our P/Q defective mutant line offered a means of testing the ability of roscovitine, known to potentiate frog neuromuscular transmission, to mediate behavioral and functional rescue. Acute treatment led to rapid improvement of both, pointing to potential therapeutic benefit for myasthenic disorders involving calcium channel dysfunction.


Assuntos
Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Junção Neuromuscular/fisiologia , Transmissão Sináptica/fisiologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/genética , Canais de Cálcio/fisiologia , Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo Q/genética , Clonagem Molecular , Células HEK293 , Humanos , Dados de Sequência Molecular , Mutação/fisiologia , Junção Neuromuscular/genética , Transmissão Sináptica/genética , Peixe-Zebra
5.
J Neurosci ; 33(48): 18755-63, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24285882

RESUMO

At the nerve terminal, neurotransmitter release is triggered by Ca(2+) influx through voltage-gated Ca(2+) channels (VGCCs). During postnatal development, VGCC subtypes in the nerve terminal switch at many synapses. In immature rodent cerebella, N-type and P/Q-type VGCCs mediate GABAergic neurotransmission from Purkinje cells (PCs) to deep nuclear cells, but as animals mature, neurotransmission becomes entirely P/Q-type dependent. We reproduced this developmental switch in rat cerebellar slice culture to address the underlying mechanism. Chronic block of cerebellar neuronal activity with tetrodotoxin (TTX) in slice culture, or in vivo, reversed the switch, leaving neurotransmission predominantly N-type channel-dependent. Brain-derived neurotrophic factor or neurotrophin-4 rescued this TTX effect, whereas pharmacological blockade of neurotrophin receptors mimicked the TTX effect. In PC somata, unlike in presynaptic terminals, TTX had no effect on the proportion of Ca(2+) channel subtype currents. We conclude that neuronal activity activates the neurotrophin-TrkB signaling pathway, thereby causing the N-to-P/Q channel switch in presynaptic terminals.


Assuntos
Canais de Cálcio/fisiologia , Fatores de Crescimento Neural/fisiologia , Neurotransmissores/metabolismo , Transdução de Sinais/fisiologia , Anestésicos Locais/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Cerebelo/efeitos dos fármacos , Cerebelo/fisiologia , Fenômenos Eletrofisiológicos/fisiologia , Feminino , Vetores Genéticos , Imuno-Histoquímica , Técnicas In Vitro , Lentivirus/genética , Masculino , Terminações Nervosas/efeitos dos fármacos , Terminações Nervosas/metabolismo , Terminações Nervosas/fisiologia , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Wistar , Receptor trkB/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tetrodotoxina/farmacologia
6.
Am J Physiol Cell Physiol ; 307(5): C455-65, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24990647

RESUMO

At early life, the adrenal chromaffin cells respond with a catecholamine surge under hypoxic conditions. This response depends on Ca(2+) entry through voltage-activated calcium channels (VACCs). We have investigated here three unresolved questions that concern this response in rat embryo chromaffin cells (ECCs): 1) the relative contribution of L (α1D, Cav1.3), N (α1B, Cav2.2), and PQ (α1A, Cav2.1) to the whole cell Ca(2+) current (ICa); 2) the relative contribution of L and N/PQ channels to the cytosolic Ca(2+) elevations triggered by hypoxia (Δ[Ca(2+)]c); and 3) the role of L and non-L high-VACCs in the regulation of the catecholamine surge occurring during prolonged (1 min) hypoxia exposure of ECCs. Nimodipine halved peak ICa and blocked 60% the total Ca(2+) entry during a 50-ms depolarizing pulse to 0 mV (QCa). Combined ω-agatoxin IVA plus ω-conotoxin GVIA (Aga/GVIA) blocked 30% of both ICa peak and QCa. This relative proportion of L- and non-L VACCs was corroborated by Western blot that indicated 55, 23, and 25% relative expression of L, N, and PQ VACCs. Exposure of ECCs to hypoxia elicited a mild but sustained Δ[Ca(2+)]c; the area of Δ[Ca(2+)]c was blocked 50% by nifedipine and 10% by Aga/GVIA. Exposure of ECCs to 1-min hypoxia elicited an initial transient burst of amperometric secretory spikes followed by scattered spikes along the time of cell exposure to hypoxia. This bulk response was blocked 85% by nimodipine and 35% by Aga/GVIA. Histograms on secretory spike frequency vs. time indicated a faster initial inactivation when Ca(2+) entry took place through N/PQ channels; more sustained secretion but at a lower rate was associated to Ca(2+) entry through L channels. The results suggest that the HIS response may initially be controlled by L and P/Q channels, but later on, N/PQ channels inactivate and the delayed HIS response is maintained at lower rate by slow-inactivating L channels.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Catecolaminas/metabolismo , Células Cromafins/metabolismo , Animais , Sítios de Ligação/fisiologia , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio Tipo P/metabolismo , Canais de Cálcio Tipo Q/metabolismo , Hipóxia Celular/fisiologia , Células Cultivadas , Células Cromafins/fisiologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/fisiologia , Ratos
7.
J Neurochem ; 129(4): 628-36, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24521102

RESUMO

Urotensin II (U-II) is a cyclic undecapeptide that regulates cardiovascular function at central and peripheral sites. The functional role of U-II nucleus ambiguus, a key site controlling cardiac tone, has not been established, despite the identification of U-II and its receptor at this level. We report here that U-II produces an increase in cytosolic Ca(2+) concentration in retrogradely labeled cardiac vagal neurons of nucleus ambiguus via two pathways: (i) Ca(2+) release from the endoplasmic reticulum via inositol 1,4,5-trisphosphate receptor; and (ii) Ca(2+) influx through P/Q-type Ca(2+) channels. In addition, U-II depolarizes cultured cardiac parasympathetic neurons. Microinjection of increasing concentrations of U-II into nucleus ambiguus elicits dose-dependent bradycardia in conscious rats, indicating the in vivo activation of the cholinergic pathway controlling the heart rate. Both the in vitro and in vivo effects were abolished by the urotensin receptor antagonist, urantide. Our findings suggest that, in addition, to the previously reported increase in sympathetic outflow, U-II activates cardiac vagal neurons of nucleus ambiguus, which may contribute to cardioprotection.


Assuntos
Bradicardia/fisiopatologia , Tronco Encefálico/fisiopatologia , Sinalização do Cálcio/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiopatologia , Neurônios/metabolismo , Sistema Nervoso Parassimpático/fisiopatologia , Urotensinas/fisiologia , Nervo Vago/fisiopatologia , Animais , Animais Recém-Nascidos , Fibras Autônomas Pré-Ganglionares/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Bradicardia/induzido quimicamente , Tronco Encefálico/efeitos dos fármacos , Canais de Cálcio Tipo P/efeitos dos fármacos , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/efeitos dos fármacos , Canais de Cálcio Tipo Q/fisiologia , Sinalização do Cálcio/fisiologia , Feminino , Sistema de Condução Cardíaco/efeitos dos fármacos , Receptores de Inositol 1,4,5-Trifosfato/efeitos dos fármacos , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Microinjeções , Modelos Cardiovasculares , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/fisiologia , Taquicardia/induzido quimicamente , Taquifilaxia , Urotensinas/farmacologia , Urotensinas/toxicidade
8.
J Neurosci ; 32(4): 1261-72, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22279211

RESUMO

Voltage-gated Ca2+ channels (VGCCs) of the P/Q-type, which are expressed at a majority of mammalian nerve terminals, show two types of Ca2+-dependent feedback regulation-inactivation (CDI) and facilitation (CDF). Because of the nonlinear relationship between Ca2+ influx and transmitter release, CDI and CDF are powerful regulators of synaptic strength. To what extent VGCCs inactivate or facilitate during spike trains depends on the dynamics of free Ca2+ ([Ca2+]i) and the Ca2+ sensitivity of CDI and CDF, which has not been determined in nerve terminals. In this report, we took advantage of the large size of a rat auditory glutamatergic synapse--the calyx of Held--and combined voltage-clamp recordings of presynaptic Ca2+ currents (ICa(V)) with UV-light flash-induced Ca2+ uncaging and presynaptic Ca2+ imaging to study the Ca2+ requirements for CDI and CDF. We find that nearly half of the presynaptic VGCCs inactivate during 100 ms voltage steps and require several seconds to recover. This inactivation is caused neither by depletion of Ca2+ ions from the synaptic cleft nor by metabotropic feedback inhibition, because it is resistant to blockade of metabotropic and ionotropic glutamate receptors. Facilitation of ICa(V) induced by repetitive depolarizations or preconditioning voltage steps decays within tens of milliseconds. Since Ca2+ buffers only weakly affect CDI and CDF, we conclude that the Ca2+ sensors are closely associated with the channel. CDI and CDF can be induced by intracellular photo release of Ca2+ resulting in [Ca2+]i elevations in the low micromolar range, implying a surprisingly high affinity of the Ca2+ sensors.


Assuntos
Canais de Cálcio Tipo P/metabolismo , Canais de Cálcio Tipo Q/metabolismo , Cálcio/antagonistas & inibidores , Cálcio/fisiologia , Glutamatos/fisiologia , Líquido Intracelular/fisiologia , Inibição Neural/fisiologia , Terminações Pré-Sinápticas/metabolismo , Animais , Animais Recém-Nascidos , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Masculino , Técnicas de Cultura de Órgãos , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Wistar
9.
Proc Natl Acad Sci U S A ; 107(33): 14887-92, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20679232

RESUMO

The Rem, Rem2, Rad, and Gem/Kir (RGK) family of small GTP-binding proteins potently inhibits high voltage-activated (HVA) Ca(2+) channels, providing a powerful means of modulating neural, endocrine, and muscle functions. The molecular mechanisms of this inhibition are controversial and remain largely unclear. RGK proteins associate directly with Ca(2+) channel beta subunits (Ca(v)beta), and this interaction is widely thought to be essential for their inhibitory action. In this study, we investigate the molecular underpinnings of Gem inhibition of P/Q-type Ca(2+) channels. We find that a purified Gem protein markedly and acutely suppresses P/Q channel activity in inside-out membrane patches, that this action requires Ca(v)beta but not the Gem/Ca(v)beta interaction, and that Gem coimmunoprecipitates with the P/Q channel alpha(1) subunit (Ca(v)alpha(1)) in a Ca(v)beta-independent manner. By constructing chimeras between P/Q channels and Gem-insensitive low voltage-activated T-type channels, we identify a region encompassing transmembrane segments S1, S2, and S3 in the second homologous repeat of Ca(v)alpha(1) critical for Gem inhibition. Exchanging this region between P/Q and T channel Ca(v)alpha(1) abolishes Gem inhibition of P/Q channels and confers Ca(v)beta-dependent Gem inhibition to a chimeric T channel that also carries the P/Q I-II loop (a cytoplasmic region of Ca(v)alpha(1) that binds Ca(v)beta). Our results challenge the prevailing view regarding the role of Ca(v)beta in RGK inhibition of high voltage-activated Ca(2+) channels and prompt a paradigm in which Gem directly binds and inhibits Ca(v)beta-primed Ca(v)alpha(1) on the plasma membrane.


Assuntos
Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Membrana Celular/efeitos dos fármacos , Proteínas Monoméricas de Ligação ao GTP/farmacologia , Sequência de Aminoácidos , Animais , Western Blotting , Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo P/metabolismo , Canais de Cálcio Tipo Q/genética , Canais de Cálcio Tipo Q/metabolismo , Linhagem Celular , Membrana Celular/fisiologia , Feminino , Humanos , Imunoprecipitação , Cinética , Potenciais da Membrana/efeitos dos fármacos , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oócitos/fisiologia , Ligação Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Subunidades Proteicas/fisiologia , Homologia de Sequência de Aminoácidos , Xenopus laevis
11.
J Neurosci ; 31(2): 512-25, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21228161

RESUMO

Synapse formation requires the organization of presynaptic active zones, the synaptic vesicle release sites, in precise apposition to postsynaptic neurotransmitter receptor clusters; however, the molecular mechanisms responsible for these processes remain unclear. Here, we show that P/Q-type and N-type voltage-dependent calcium channels (VDCCs) play essential roles as scaffolding proteins in the organization of presynaptic active zones. The neuromuscular junction of double knock-out mice for P/Q- and N-type VDCCs displayed a normal size but had significantly reduced numbers of active zones and docked vesicles and featured an attenuation of the active-zone proteins Bassoon, Piccolo, and CAST/Erc2. Consistent with this phenotype, direct interactions of the VDCC ß1b or ß4 subunits and the active zone-specific proteins Bassoon or CAST/Erc2 were confirmed by immunoprecipitation. A decrease in the number of active zones caused by a loss of presynaptic VDCCs resembled the pathological conditions observed in the autoimmune neuromuscular disorder Lambert-Eaton myasthenic syndrome. At the synaptic cleft of double knock-out mice, we also observed a decrease of the synaptic organizer laminin ß2 protein, an extracellular ligand of P/Q- and N-type VDCCs. However, the transcription level of laminin ß2 did not decrease in double knock-out mice, suggesting that the synaptic accumulation of laminin ß2 protein required its interaction with presynaptic VDCCs. These results suggest that presynaptic VDCCs link the target-derived synapse organizer laminin ß2 to active-zone proteins and function as scaffolding proteins to anchor active-zone proteins to the presynaptic membrane.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Proteínas do Citoesqueleto/metabolismo , Laminina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Junção Neuromuscular/metabolismo , Sinapses/metabolismo , Animais , Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo Q/genética , Contagem de Células , Eletromiografia , Embrião de Mamíferos , Camundongos , Camundongos Knockout , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Nervo Frênico/metabolismo , Nervo Frênico/ultraestrutura , Subunidades Proteicas/metabolismo , Medula Espinal/citologia , Medula Espinal/metabolismo
12.
J Neurosci ; 31(4): 1313-22, 2011 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-21273416

RESUMO

Agonists at µ-opioid receptors (MORs) represent the gold standard for the treatment of severe pain. A key element of opioid analgesia is the depression of nociceptive information at the first synaptic relay in spinal pain pathways. The underlying mechanisms are, however, largely unknown. In spinal cord slices with dorsal roots attached prepared from young rats, we determined the inhibitory effect of the selective MOR agonist [d-Ala(2), N-Me-Phe(4), Gly(5)-ol]-enkephalin (DAMGO) on monosynaptic Aδ- and C-fiber-evoked EPSCs in lamina I neurons. DAMGO depressed presynaptically Aδ- and C-fiber-mediated responses, indicating that MORs are expressed on central terminals of both fiber types. We next addressed the mechanisms of presynaptic inhibition. The effect of DAMGO at both Aδ- and C-fiber terminals was mainly mediated by an inhibition of N-type voltage-dependent Ca(2+) channels (VDCCs), and to a lesser extent of P/Q-type VDCCs. Inhibition by DAMGO was not reduced by K(+) channel blockers. The rate of miniature EPSCs was reduced by DAMGO in a dose-dependent manner. The opioid also reduced Ca(2+)-dependent, ionomycin-induced EPSCs downstream of VDCCs. DAMGO had no effect on the kinetics of vesicle exocytosis in C-fiber terminals, but decreased the rate of unloading of Aδ-fiber boutons moderately, as revealed by two-photon imaging of styryl dye destaining. Together, these results suggest that binding of opioids to MORs reduces nociceptive signal transmission at central Aδ- and C-fiber synapses mainly by inhibition of presynaptic N-type VDCCs. P/Q-type VDCCs and the transmitter release machinery are targets of opioid action as well.


Assuntos
Fibras Nervosas Mielinizadas/fisiologia , Fibras Nervosas Amielínicas/fisiologia , Inibição Neural , Receptores Opioides mu/fisiologia , Medula Espinal/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Estimulação Elétrica , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Pós-Sinápticos Excitadores , Técnicas In Vitro , Masculino , Potenciais Pós-Sinápticos em Miniatura , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Amielínicas/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Medula Espinal/fisiologia , Raízes Nervosas Espinhais/efeitos dos fármacos , Raízes Nervosas Espinhais/fisiologia
13.
J Neurophysiol ; 107(8): 2212-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22279189

RESUMO

Calcium currents are critical to the intrinsic properties of neurons and the networks that contain them. These currents make attractive targets for neuromodulation. Here, we examine the serotonergic modulation of specific calcium current subtypes in neonatal (P0-5) intersegmental commissural interneurons (CINs), members of the hindlimb locomotor central pattern generator in the mouse spinal cord. Previous work in our lab showed that serotonin (5-HT) excited CINs in part by reducing a calcium current and thus indirectly reducing the calcium-activated potassium current (Diaz-Rios et al. 2007). We have determined which calcium currents are targets of serotonin modulation. Utilizing whole cell voltage clamp and toxins to specific calcium current subtypes, we found that N- and P/Q-type currents comprise over 60% of the overall calcium current. Blockade of each of these subtypes alone with either ω-conotoxin GVIA or ω-agatoxin TK was unable to occlude 5-HT's reduction of the calcium current. However, coapplication of both blockers together fully occluded 5-HT's reduction of the calcium current. Thus, 5-HT decreases both N- and P/Q-type calcium current to excite neonatal CINs.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio Tipo P/metabolismo , Canais de Cálcio Tipo Q/metabolismo , Interneurônios/fisiologia , Serotonina/fisiologia , Animais , Animais Recém-Nascidos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Interneurônios/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL
14.
Eur J Neurosci ; 36(8): 3021-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22817403

RESUMO

Electrical activity in the gamma frequency range is instrumental for temporal encoding on the millisecond scale in attentive vertebrate brains. Surprisingly, also circadian pacemaker neurons in the cockroach Rhyparobia maderae (Leucophaea maderae) employ fast spontaneous rhythmic activity in the gamma band frequency range (20-70  Hz) together with slow rhythmic activity. The ionic conductances controlling this fast spontaneous activity are still unknown. Here, Ca(2+) imaging combined with pharmacology was employed to analyse ion channels underlying spontaneous activity in dispersed circadian pacemakers of the adult accessory medulla, which controls circadian locomotor activity rhythms. Fast spontaneous Ca(2+) transients in circadian pacemakers accompany tetrodotoxin (TTX)-blockable spontaneous action potentials. In contrast to vertebrate pacemakers, the spontaneous depolarisations from rest appear to be rarely initiated via TTX-sensitive sustained Na(+) channels. Instead, they are predominantly driven by mibefradil-sensitive, low-voltage-activated Ca(2+) channels and DK-AH269-sensitive hyperpolarisation-activated, cyclic nucleotide-gated cation channels. Rhythmic depolarisations activate voltage-gated Na(+) channels and nifedipine-sensitive high-voltage-activated Ca(2+) channels. Together with Ca(2+) rises, the depolarisations open repolarising small-conductance but not large-conductance Ca(2+) -dependent K(+) channels. In contrast, we hypothesise that P/Q-type Ca(2+) channels coupled to large-conductance Ca(2+) -dependent K(+) channels are involved in input-dependent activity.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Cálcio/metabolismo , Relógios Circadianos , Neurônios/fisiologia , Canais de Potássio Cálcio-Ativados/fisiologia , Potenciais de Ação , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Baratas , Canais de Cátion Regulados por Nucleotídeos Cíclicos/antagonistas & inibidores , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Proteínas de Insetos/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Masculino , Mibefradil/farmacologia , Nifedipino/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Canais de Sódio Disparados por Voltagem/fisiologia
15.
Cerebellum ; 11(2): 449-50, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22194041

RESUMO

Cerebellar Purkinje cells (PCs) of newborn rodents are innervated by multiple climbing fibers (CFs). During the first postnatal week, single CFs are strengthened relative to other CFs on the somata of individual PCs. Then, the strengthened CFs undergo translocation to PC dendrites after P9. Elimination of the weaker CFs occurs in two distinct steps, namely the early phase from P7 to around P12 and the late phase from about P12 to around P17. Our previous study demonstrates that CF synapse elimination is severely impaired in null mutant mice lacking Ca(v)2.1, a pore-forming component of P/Q-type voltage-dependent Ca(2+) channel (VDCC). To examine the contribution of postsynaptic P/Q-type VDCC to postnatal rearrangement of CFs, we generated mice with PC-selective deletion of Ca(v)2.1 (PC-Ca(v)2.1 KO). We made whole-cell recordings from PCs in cerebellar slices and examined CF-mediated excitatory postsynaptic currents. We found that PC-Ca(v)2.1 KO PCs had severe defects in selective strengthening of single CFs during the first postnatal week and subsequent CF synapse elimination from P7. Moreover, our morphological analysis revealed that multiple CFs abnormally underwent translocation to PC dendrites in PC-Ca(v)2.1 KO mice. These results indicate that Ca(2+) influx through P/Q-type VDCC into PCs is crucial for selective strengthening of single CFs, early phase elimination and selective translocation of single strengthened CFs to PC dendrites.


Assuntos
Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Fibras Nervosas/fisiologia , Células de Purkinje/fisiologia , Animais , Animais Recém-Nascidos , Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/genética , Canais de Cálcio Tipo Q/fisiologia , Dendritos/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Camundongos , Camundongos Knockout
16.
J Neurosci ; 30(35): 11858-69, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20810905

RESUMO

Cortical axons contain a diverse range of voltage-activated ion channels, including Ca(2+) currents. Interestingly, Ca(2+) channels are not only located at presynaptic terminals, but also in the axon initial segment (AIS), suggesting a potentially important role in the regulation of action potential generation and neuronal excitability. Here, using two-photon microscopy and whole-cell patch-clamp recording, we examined the properties and role of calcium channels located in the AIS and presynaptic terminals of ferret layer 5 prefrontal cortical pyramidal cells in vitro. Subthreshold depolarization of the soma resulted in an increase in baseline and spike-triggered calcium concentration in both the AIS and nearby synaptic terminals. The increase in baseline calcium concentration rose with depolarization and fell with hyperpolarization with a time constant of approximately 1 s and was blocked by removal of Ca(2+) from the bathing medium. The increases in calcium concentration at the AIS evoked by subthreshold or suprathreshold depolarization of the soma were blocked by the P/Q-channel antagonist omega-agatoxin IVA or the N-channel antagonist omega-conotoxin GVIA or both. The presence of these channels in the AIS pyramidal cells was confirmed with immunochemistry. Block of these channels slowed axonal action potential repolarization, apparently from reduction of the activation of a Ca(2+)-activated K(+) current, and increased neuronal excitability. These results demonstrate novel mechanisms by which calcium currents may control the electrophysiological properties of axonal spike generation and neurotransmitter release in the neocortex.


Assuntos
Axônios/fisiologia , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Neocórtex/fisiologia , Terminações Pré-Sinápticas/fisiologia , Potenciais de Ação/fisiologia , Animais , Feminino , Furões , Masculino , Neocórtex/citologia
17.
J Neurosci ; 30(13): 4536-46, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20357104

RESUMO

Synaptic transmission at CNS synapses is often mediated by joint actions of multiple Ca(2+) channel subtypes, most prominently, P/Q- and N-type. We have proposed that P/Q-type Ca(2+) channels saturate type-preferring slots at presynaptic terminals, which impose a ceiling on the synaptic efficacy of the channels. To test for analogous interactions for presynaptic N-type Ca(2+) channels, we overexpressed their pore-forming Ca(V)2.2 subunit in cultured mouse hippocampal neurons, recorded excitatory synaptic transmission from transfected cells, and dissected the contributions of N-, P/Q-, and R-type channels with subtype-specific blockers. Overexpression of Ca(V)2.2 did not increase the absolute size of the EPSC even though somatic N-type current was augmented by severalfold. Thus, the strength of neurotransmission is saturated with regard to levels of Ca(2+) channel expression for both N-type and P/Q-type channels. Overexpression of Ca(2+)-impermeable Ca(V)2.2 subunits decreased EPSC size, corroborating competition for channel slots. Striking asymmetries between N- and P/Q-type channels emerged when their relative contributions were compared with channel overexpression. Overexpressed N-type channels could competitively displace P/Q-type channels from P/Q-preferring slots and take over the role of supporting transmission. The converse was not found with overexpression of P/Q-type channels, regardless of their C-terminal domain. We interpret these findings in terms of two different kinds of presynaptic slots at excitatory synapses, one accepting N-type channels but rejecting P/Q-type (N(specific)) and the other preferring P/Q-type but also accepting N-type (PQ(preferring)). The interaction between channels and slots governs the respective contributions of multiple channel types to neurotransmission and, in turn, the ability of transmission to respond to various stimulus patterns and neuromodulators.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Hipocampo/fisiologia , Sinapses/fisiologia , Potenciais de Ação , Animais , Canais de Cálcio Tipo N/biossíntese , Canais de Cálcio Tipo P/biossíntese , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/biossíntese , Canais de Cálcio Tipo Q/fisiologia , Células Cultivadas , Hipocampo/ultraestrutura , Humanos , Camundongos , Neurônios/fisiologia , Neurônios/ultraestrutura , Técnicas de Patch-Clamp , Isoformas de Proteínas/fisiologia , Subunidades Proteicas/biossíntese , Subunidades Proteicas/fisiologia , Transmissão Sináptica
18.
J Physiol ; 589(Pt 23): 5879-95, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22005682

RESUMO

Familial hemiplegic migraine type-1 (FHM1), a monogenic subtype of migraine with aura, is caused by gain-of-function mutations in Ca(V)2.1 (P/Q-type) calcium channels. The consequences of FHM1 mutations on the trigeminovascular pathway that generates migraine headache remain largely unexplored. Here we studied the calcium currents and excitability properties of two subpopulations of small-diameter trigeminal ganglion (TG) neurons from adult wild-type (WT) and R192Q FHM1 knockin (KI) mice: capsaicin-sensitive neurons without T-type calcium currents (CS) and capsaicin-insensitive neurons characterized by the expression of T-type calcium currents (CI-T). Small TG neurons retrogradely labelled from the dura are mostly CS neurons, while CI-T neurons were not present in the labelled population. CS and CI-T neurons express Ca(V)2.1 channels with different activation properties, and the Ca(V)2.1 channels are differently affected by the FHM1 mutation in the two TG neuron subtypes. In CI-T neurons from FHM1 KI mice there was a larger P/Q-type current density following mild depolarizations, a larger action potential (AP)-evoked calcium current and a longer AP duration when compared to CI-T neurons from WT mice. In striking contrast, the P/Q-type current density, voltage dependence and kinetics were not altered by the FHM1 mutation in CS neurons. The excitability properties of mutant CS neurons were also unaltered. Congruently, the FHM1 mutation did not alter depolarization-evoked CGRP release from the dura mater, while CGRP release from the trigeminal ganglion was larger in KI compared to WT mice. Our findings suggest that the facilitation of peripheral mechanisms of CGRP action, such as dural vasodilatation and nociceptor sensitization at the meninges, does not contribute to the generation of headache in FHM1.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Ataxia Cerebelar/fisiopatologia , Transtornos de Enxaqueca/fisiopatologia , Gânglio Trigeminal/fisiologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Capsaicina , Ataxia Cerebelar/genética , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos de Enxaqueca/genética , Mutação , Neurônios/fisiologia
19.
Cephalalgia ; 31(13): 1368-80, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21893556

RESUMO

INTRODUCTION: Familial hemiplegic migraine type 1 (FHM-1) is caused by mutations in the CACNA1A gene, with the R192Q mutation being the most common. Elevated calcitonin gene-related peptide (CGRP) levels in acute migraine and clinical trials using CGRP receptor antagonists suggest CGRP-related mechanisms are important in migraine. METHODS: Wild-type and R192Q knock-in mice were anaesthetized and perfused. Using immunohistochemical staining, the expression of CGRP in the trigeminocervical complex (TCC) and in the trigeminal and dorsal root ganglia was characterized. RESULTS: There was a 38% reduction in the percentage of CGRP-immunoreactive cells in the trigeminal ganglia (p < 0.001) of R192Q knock-in mice compared to wild-type animals. The size distribution profile of CGRP-immunoreactive cells within the trigeminal ganglia demonstrated no significant difference in cell diameter between the two groups (p ≥ 0.56). CGRP expression was also reduced in thoracic ganglia of R192Q knock-in mice (21% vs. 27% in wild-type group; p < 0.05), but not in other ganglia. In addition, decreased CGRP immunoreactivity was observed in the superficial laminae of the TCC in R192Q knock-in mice, when compared to the control group (p < 0.005). CONCLUSION: The data demonstrates that the FHM-1 CACNA1A mutation alters CGRP expression in the trigeminal ganglion and TCC. This suggests further study of these animals is warranted to characterize better the role of these mutations in the neurobiology of migraine.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/biossíntese , Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo Q/genética , Ataxia Cerebelar/genética , Gânglios Espinais/metabolismo , Transtornos de Enxaqueca/genética , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/fisiologia , Mutação Puntual , Medula Espinal/metabolismo , Nervo Trigêmeo/metabolismo , Núcleos do Trigêmeo/metabolismo , Substituição de Aminoácidos , Animais , Avidina/análise , Peptídeo Relacionado com Gene de Calcitonina/genética , Canais de Cálcio Tipo N , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Códon/genética , Feminino , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/análise , Gânglios Espinais/citologia , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Medula Espinal/citologia , Nervo Trigêmeo/citologia , Núcleos do Trigêmeo/citologia
20.
J Physiol ; 587(Pt 8): 1657-68, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19221121

RESUMO

The second messenger, 3',5'-cyclic adenosine monophosphate (cAMP), is known to be modulated in taste buds following exposure to gustatory and other stimuli. Which taste cell type(s) (Type I/glial-like cells, Type II/receptor cells, or Type III/presynaptic cells) undergo taste-evoked changes of cAMP and what the functional consequences of such changes are remain unknown. Using Fura-2 imaging of isolated mouse vallate taste cells, we explored how elevating cAMP alters Ca(2+) levels in identified taste cells. Stimulating taste buds with forskolin (Fsk; 1 microm) + isobutylmethylxanthine (IBMX; 100 microm), which elevates cellular cAMP, triggered Ca(2+) transients in 38% of presynaptic cells (n = 128). We used transgenic GAD-GFP mice to show that cAMP-triggered Ca(2+) responses occur only in the subset of presynaptic cells that lack glutamic acid decarboxylase 67 (GAD). We never observed cAMP-stimulated responses in receptor cells, glial-like cells or GAD-expressing presynaptic cells. The response to cAMP was blocked by the protein kinase A inhibitor H89 and by removing extracellular Ca(2+). Thus, the response to elevated cAMP is a PKA-dependent influx of Ca(2+). This Ca(2+) influx was blocked by nifedipine (an inhibitor of L-type voltage-gated Ca(2+) channels) but was unperturbed by omega-agatoxin IVA and omega-conotoxin GVIA (P/Q-type and N-type channel inhibitors, respectively). Single-cell RT-PCR on functionally identified presynaptic cells from GAD-GFP mice confirmed the pharmacological analyses: Ca(v)1.2 (an L-type subunit) is expressed in cells that display cAMP-triggered Ca(2+) influx, while Ca(v)2.1 (a P/Q subunit) is expressed in all presynaptic cells, and underlies depolarization-triggered Ca(2+) influx. Collectively, these data demonstrate cross-talk between cAMP and Ca(2+) signalling in a subclass of taste cells that form synapses with gustatory fibres and may integrate tastant-evoked signals.


Assuntos
Cálcio/fisiologia , AMP Cíclico/fisiologia , Receptores Pré-Sinápticos/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Papilas Gustativas/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo P/efeitos dos fármacos , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/efeitos dos fármacos , Canais de Cálcio Tipo Q/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Sinalização do Cálcio/fisiologia , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Glutamato Descarboxilase/biossíntese , Glutamato Descarboxilase/genética , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Endogâmicos C57BL , Inibidores de Fosfodiesterase/farmacologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores Pré-Sinápticos/efeitos dos fármacos , Receptores Pré-Sinápticos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Sistemas do Segundo Mensageiro/genética , Papilas Gustativas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA