Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39295452

RESUMO

BACKGROUND AND PURPOSE: The chemotherapy agent oxaliplatin can give rise to oxaliplatin-induced peripheral neuropathy (OIPN). Here, we investigated whether T-type calcium channels (Cav3) contribute to OIPN. EXPERIMENTAL APPROACH: We chronically treated mice with oxaliplatin and assessed pain responses and changes in expression of Cav3.2 calcium channels. We also tested the effects of T-type channel blockers on cold sensitivity in wild-type and Cav3.2 null mice. KEY RESULTS: Oxaliplatin treatment led to mechanical and cold hypersensitivity in male and female mice. Mechanical hypersensitivity persisted in Cav3.2 null mice of both sexes. Intraperitoneal or intrathecal delivery of pan T-type channel inhibitors attenuated mechanical hypersensitivity in wild-type but not Cav3.2 null mice. Remarkably cold hypersensitivity occurred in female but not male Cav3.2 null mice even without oxaliplatin treatment. Unexpectedly, intrathecal, intraplantar or intraperitoneal delivery of T-type channel inhibitors Z944 or TTA-P2 transiently induced cold hypersensitivity in both male and female wild-type mice. Acute knockdown of specific Cav3 isoforms revealed that the depletion of Cav3.1 in males and depletion of either Cav3.1 or Cav3.2 in females triggered cold hypersensitivity. Finally, reducing Cav3.2 expression by disrupting the interactions between Cav3.2 and the deubiquitinase USP5 with the small organic molecule II-2 reversed oxaliplatin-induced mechanical and cold hypersensitivity and importantly did not trigger cold allodynia. CONCLUSION AND IMPLICATIONS: Altogether, our data indicate that T-type channels differentially contribute to the regulation of cold and mechanical hypersensitivity, and raise the possibility that T-type channel blockers could promote cold allodynia.

2.
Elife ; 52016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27874832

RESUMO

Seizures are often followed by sensory, cognitive or motor impairments during the postictal phase that show striking similarity to transient hypoxic/ischemic attacks. Here we show that seizures result in a severe hypoxic attack confined to the postictal period. We measured brain oxygenation in localized areas from freely-moving rodents and discovered a severe hypoxic event (pO2 < 10 mmHg) after the termination of seizures. This event lasted over an hour, is mediated by hypoperfusion, generalizes to people with epilepsy, and is attenuated by inhibiting cyclooxygenase-2 or L-type calcium channels. Using inhibitors of these targets we separated the seizure from the resulting severe hypoxia and show that structure specific postictal memory and behavioral impairments are the consequence of this severe hypoperfusion/hypoxic event. Thus, epilepsy is much more than a disease hallmarked by seizures, since the occurrence of postictal hypoperfusion/hypoxia results in a separate set of neurological consequences that are currently not being treated and are preventable.


Assuntos
Comportamento Animal , Encéfalo/fisiopatologia , Ciclo-Oxigenase 2/metabolismo , Hipóxia/patologia , Convulsões/complicações , Animais , Ratos Long-Evans
3.
Mol Brain ; 7: 51, 2014 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-25063346

RESUMO

BACKGROUND: Manipulation of gene expression via recombinant viral vectors and creation of transgenic knock-out/in animals has revolutionized our understanding of genes that play critical roles during neuronal development and pathophysiology of neurological disorders. Recently, target-specific genetic manipulations are made possible to perform in combination with specific Cre-lines, albeit costly, labor-intensive and time consuming. Thus, alternative methods of gene manipulations to address important biological questions are highly desirable. In this study, we utilized in utero electroporation technique which involves efficient delivery of hindbrain-specific enhancer/promoter construct, Krox20 into the third ventricle of live mouse embryo to investigate green fluorescent protein (GFP) expression pattern in mouse auditory brainstem and other hindbrain neurons. RESULTS: We created a GFP/DNA construct containing a Krox20 B enhancer and ß-globin promoter to drive GFP expression in the hindbrain via injection into the third ventricle of E12 to E13.5 mice. Electrical currents were applied directly to the embryonic hindbrain to allow DNA uptake into the cell. Confocal images were then acquired from fixed brain slices to analyze GFP expression in mouse whole brain at different postnatal stages (P6-P21). By using a cell-type specific enhancer as well as region specific injection and electroporation, robust GFP expression in the cerebellum and auditory brainstem but not in the forebrain was observed. GFP expression in calyx of Held terminals was more robust in P15 mice. In contrast, GFP expression in MNTB neurons was more prevalent in >P15 compared to

Assuntos
Tronco Encefálico/embriologia , Tronco Encefálico/metabolismo , Orelha/inervação , Eletroporação/métodos , Técnicas de Transferência de Genes , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Animais , Clonagem Molecular , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Elementos Facilitadores Genéticos/genética , Feminino , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Neurônios/metabolismo , Sinapses/metabolismo
4.
Channels (Austin) ; 4(5): 375-89, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20699644

RESUMO

Low voltage-activated T-type calcium (Ca) channels contribute to the normal development of the heart and are also implicated in pathophysiological states such as cardiac hypertrophy. Functionally distinct T-type Ca channel isoforms can be generated by alternative splicing from each of three different T-type genes (Ca(V)3.1, Ca(V)3.2,Ca(V)3.3), although it remains to be described whether specific splice variants are associated with developmental states and pathological conditions. We aimed to identify and functionally characterize Ca(V)3.2 T-type Ca channel alternatively spliced variants from newborn animals and to compare with adult normotensive and spontaneously hypertensive rats (SHR). DNA sequence analysis of full-length Ca(V)3.2 cDNA generated from newborn heart tissue identified ten major regions of alternative splicing, the more common variants of which were analyzed by quantitative real-time PCR (qRT-PCR) and also subject to functional examination by whole-cell patch clamp. The main findings are that: (1) cardiac Ca(V)3.2 T-type Ca channels are subject to considerable alternative splicing, (2) there is preferential expression of Ca(V)3.2(-25) splice variant channels in newborn rat heart with a developmental shift in adult heart that results in approximately equal levels of expression of both (+25) and (-25) exon variants, (3) in the adult stage of hypertensive rats there is a both an increase in overall Ca(V)3.2 expression and a shift towards expression of Ca(V)3.2(+25) containing channels as the predominant form, and (4) alternative splicing confers a variant-specific voltage-dependent facilitation of Ca(V)3.2 channels. We conclude that Ca(V)3.2 alternative splicing generates significant T-type Ca channel structural and functional diversity with potential implications relevant to cardiac developmental and pathophysiological states.


Assuntos
Processamento Alternativo/fisiologia , Canais de Cálcio Tipo T/genética , Cardiomegalia/genética , Coração/crescimento & desenvolvimento , Fatores Etários , Animais , Animais Recém-Nascidos , Canais de Cálcio Tipo T/fisiologia , Técnicas Eletrofisiológicas Cardíacas/métodos , Técnicas de Patch-Clamp , Ratos , Ratos Endogâmicos SHR
5.
Fitoterapia ; 81(7): 826-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20472040

RESUMO

This investigation focuses on the in vitro neuroactive properties of pinostrobin, a substituted flavanone from Cajanus cajan (L.) Millsp. of the Fabaceae family. We demonstrate that pinostrobin inhibits voltage-gated sodium channels of mammalian brain (IC(50)=23 µM) based on the ability of this substance to suppress the depolarizing effects of the sodium channel-selective activator veratridine in a synaptoneurosomal preparation from mouse brain. The resting membrane potential of synaptoneurosomes was unaffected by pinostrobin. The pharmacological profile of pinostrobin resembles that of depressant drugs that block sodium channels.


Assuntos
Encéfalo/efeitos dos fármacos , Cajanus/química , Flavanonas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Animais , Encéfalo/fisiologia , Camundongos , Camundongos Endogâmicos , Fármacos Neuromusculares/farmacologia , Folhas de Planta , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/fisiologia , Veratridina/farmacologia
6.
Channels (Austin) ; 3(2): 110-21, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19242091

RESUMO

Alternative splicing is known to generate multiple functionally distinct calcium channel variants that exhibit unique spatial and temporal expression patterns. In humans, naturally occurring mutations in genes encoding calcium channel pore forming alpha(1)-subunits are associated with several severe hereditary disorders although it remains to be described whether there exists any relationship between the physiological effects of these mutations and calcium channel splice variation. In the present study, we systematically compare the biophysical effects of three type-1 familial hemiplegic migraine (FHM-1) mutations in two predominant splice variants of the neuronal Ca(V)2.1 P/Q-type channel. All three FHM-1 mutations cause a greater hyperpolarizing shift in voltage-dependent properties when expressed in the short carboxyl terminus variant (Ca(V)2.1 Delta47) compared to the long variant (Ca(V)2.1 +47). Furthermore, the FHM-1 mutations also exhibit differential splice variant-specific effects on recovery from inactivation and accumulation of inactivation during tonic and burst firing. Our findings provide important insight concerning the role of calcium channel alternatively spliced variants and the molecular pathophysiology of FHM-1 and potentially of other calcium channelopathies.


Assuntos
Processamento Alternativo/fisiologia , Canais de Cálcio Tipo N/genética , Enxaqueca com Aura/genética , Potenciais de Ação , Córtex Cerebral/química , Eletrofisiologia , Humanos , Potenciais da Membrana , Enxaqueca com Aura/fisiopatologia , Mutação
7.
J Neurosci ; 29(2): 371-80, 2009 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-19144837

RESUMO

Low-voltage-activated, or T-type, calcium (Ca(2+)) channels are believed to play an essential role in the generation of absence seizures in the idiopathic generalized epilepsies (IGEs). We describe a homozygous, missense, single nucleotide (G to C) mutation in the Ca(v)3.2 T-type Ca(2+) channel gene (Cacna1h) in the genetic absence epilepsy rats from Strasbourg (GAERS) model of IGE. The GAERS Ca(v)3.2 mutation (gcm) produces an arginine to proline (R1584P) substitution in exon 24 of Cacna1h, encoding a portion of the III-IV linker region in Ca(v)3.2. gcm segregates codominantly with the number of seizures and time in seizure activity in progeny of an F1 intercross. We have further identified two major thalamic Cacna1h splice variants, either with or without exon 25. gcm introduced into the splice variants acts "epistatically," requiring the presence of exon 25 to produce significantly faster recovery from channel inactivation and greater charge transference during high-frequency bursts. This gain-of-function mutation, the first reported in the GAERS polygenic animal model, has a novel mechanism of action, being dependent on exonic splicing for its functional consequences to be expressed.


Assuntos
Canais de Cálcio Tipo T/genética , Modelos Animais de Doenças , Epilepsia Tipo Ausência/genética , Mutação Puntual/genética , Isoformas de Proteínas/genética , Convulsões/genética , Animais , Animais Recém-Nascidos , Arginina/genética , Biofísica , Linhagem Celular Transformada , Estimulação Elétrica , Eletroencefalografia , Epilepsia Tipo Ausência/fisiopatologia , Éxons/genética , Humanos , Técnicas In Vitro , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Modelos Moleculares , Mutagênese Sítio-Dirigida/métodos , Técnicas de Patch-Clamp , Prolina/genética , Estrutura Terciária de Proteína/genética , Ratos , Ratos Transgênicos , Convulsões/fisiopatologia , Transfecção
8.
J Neurosci ; 27(46): 12577-83, 2007 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18003836

RESUMO

T-type Ca2+ channels (T-channels) are involved in the control of neuronal excitability and their gating can be modulated by a variety of redox agents. Ascorbate is an endogenous redox agent that can function as both an anti- and pro-oxidant. Here, we show that ascorbate selectively inhibits native Ca(v)3.2 T-channels in peripheral and central neurons, as well as recombinant Ca(v)3.2 channels heterologously expressed in human embryonic kidney 293 cells, by initiating the metal-catalyzed oxidation of a specific, metal-binding histidine residue in domain 1 of the channel. Our biophysical experiments indicate that ascorbate reduces the availability of Ca(v)3.2 channels over a wide range of membrane potentials, and inhibits Ca(v)3.2-dependent low-threshold-Ca2+ spikes as well as burst-firing in reticular thalamic neurons at physiologically relevant concentrations. This study represents the first mechanistic demonstration of ion channel modulation by ascorbate, and suggests that ascorbate may function as an endogenous modulator of neuronal excitability.


Assuntos
Ácido Ascórbico/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Ácido Ascórbico/metabolismo , Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio Tipo T/genética , Canais de Cálcio Tipo T/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Domínio Catalítico/efeitos dos fármacos , Domínio Catalítico/fisiologia , Linhagem Celular , Células Cultivadas , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Histidina/química , Humanos , Núcleos Intralaminares do Tálamo/efeitos dos fármacos , Núcleos Intralaminares do Tálamo/metabolismo , Ativação do Canal Iônico/fisiologia , Metais/química , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Oxirredução , Ratos
9.
J Biol Chem ; 282(29): 21043-55, 2007 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-17535809

RESUMO

T-type calcium channels play critical roles in controlling neuronal excitability, including the generation of complex spiking patterns and the modulation of synaptic plasticity, although the mechanisms and extent to which T-type Ca(2+) channels are modulated by G-protein-coupled receptors (GPCRs) remain largely unexplored. To examine specific interactions between T-type Ca(2+) channel subtypes and muscarinic acetylcholine receptors (mAChRS), the Cav3.1 (alpha(1G)), Cav3.2 (alpha(1H)), and Cav3.3 (alpha) T-type Ca(2+)(1I)channels were co-expressed with the M1 Galpha(q/11)-coupled mAChR. Perforated patch recordings demonstrate that activation of M1 receptors has a strong inhibitory effect on Cav3.3 T-type Ca(2+) currents but either no effect or a moderate stimulating effect on Cav3.1 and Cav3.2 peak current amplitudes. This differential modulation was observed for both rat and human T-type Ca(2+) channel variants. The inhibition of Cav3.3 channels by M1 receptors is reversible, use-independent, and associated with a concomitant increase in inactivation kinetics. Loss-of-function experiments with genetically encoded antagonists of Galpha and Gbetagamma proteins and gain-of-function experiments with genetically encoded Galpha subtypes indicate that M1 receptor-mediated inhibition of Cav3.3 occurs through Galpha(q/11). This is supported by experiments showing that activation of the M3 and M5 Galpha(q/11)-coupled mAChRs also causes inhibition of Cav3.3 currents, although Galpha(i)-coupled mAChRs (M2 and M4) have no effect. Examining Cav3.1-Cav3.3 chimeric channels demonstrates that two distinct regions of the Cav3.3 channel are necessary and sufficient for complete M1 receptor-mediated channel inhibition and represent novel sites not previously implicated in T-type channel modulation.


Assuntos
Canais de Cálcio Tipo T/química , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Membrana Transportadoras/química , Receptores Muscarínicos/metabolismo , Animais , Biofísica/métodos , Cálcio/metabolismo , Canais de Cálcio Tipo T/metabolismo , Linhagem Celular , Eletrofisiologia , Humanos , Cinética , Proteínas de Membrana Transportadoras/metabolismo , Técnicas de Patch-Clamp , Ratos , Transdução de Sinais , Fatores de Tempo , Transfecção
10.
Chemosphere ; 55(10): 1315-21, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15081774

RESUMO

A membrane potential assay based on synaptoneurosomes prepared from mouse brain was evaluated further for its utility in estimating saxitoxin and related bioactives. Saxitoxin concentrations quantitated in mussel extracts by the synaptoneurosomal technique correlated well with spiked concentrations in these samples (r2 = 0.995; slope=1.048). Other experiments found that the synaptoneurosomal assay can detect saxitoxin-like bioactives in zooplankton samples and the concentrations measured were consistent with preliminary estimations of saxitoxin equivalents using the [3H] saxitoxin receptor binding technique. Veratrine, a mixture of alkaloids that activate sodium channels, had similar potential as a substitute for veratridine in the synaptoneurosomal assay. The results provide additional evidence that the mouse brain synaptoneurosomal membrane potential assay has excellent capability for quantitation of saxitoxin-like activity in shellfish tissues and may also be applied to zooplankton samples.


Assuntos
Bivalves/química , Encéfalo/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos/fisiologia , Saxitoxina/análise , Sinaptossomos/efeitos dos fármacos , Animais , Fluorescência , Camundongos , Rodaminas , Saxitoxina/farmacologia , Sinaptossomos/fisiologia , Veratridina , Veratrina , Zooplâncton/química
11.
Basic Clin Pharmacol Toxicol ; 94(2): 73-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14748850

RESUMO

The cannabinoid 1 receptor antagonist AM 251 is known to block the inhibitory effects of endocannabinoids and synthetic cannabinoid agonists on transmitter release through an action at presynaptic cannabinoid 1 receptors in brain. We examined the ability of AM 251 to inhibit sodium channel-dependent functions and the binding of [3H]batrachotoxinin A 20-alpha-benzoate to sodium channels in mouse brain synaptic preparations. Depolarization of synaptoneurosomes by the sodium channel site 2-specific neurotoxin veratridine, which is abolished by tetrodotoxin, was found to be inhibited in a concentration-dependent fashion by AM 251 (IC50=8.9 microM). Veratridine-dependent (tetrodotoxin suppressible) release, of L-glutamic acid and GABA from synaptosomes was also reduced by AM 251 [IC50s=8.5 microM (L-glutamic acid), 9.2 microM (GABA)]. The binding of the radioligand [3H]batrachotoxinin A 20-alpha-benzoate to site 2 on sodium channels was displaced by AM 251 (IC50=11.2 microM). Scatchard analysis of binding showed that at its IC50, AM 251 increased (by 2.3 times) the KD of radioligand without altering Bmax, suggesting a competitive mechanism of inhibition by AM 251. Kinetic experiments indicated that AM 251 inhibits equilibrium binding by allosterically accelerating the dissociation of the [3H]-batrachotoxinin A 20-alpha-benzoate:sodium channel complex. Our data suggest that micromolar concentrations of AM 251 are capable of reducing neuronal excitability and inhibiting release of excitatory and inhibitory transmitters through blockade of voltage-sensitive sodium channels in brain.


Assuntos
Encéfalo/efeitos dos fármacos , Piperidinas/farmacologia , Pirazóis/farmacologia , Canais de Sódio/efeitos dos fármacos , Sinaptossomos/efeitos dos fármacos , Animais , Batraquiotoxinas/metabolismo , Antagonistas de Receptores de Canabinoides , Interações Medicamentosas , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Piperidinas/antagonistas & inibidores , Pirazóis/antagonistas & inibidores , Canais de Sódio/metabolismo , Sinaptossomos/metabolismo , Veratridina/farmacologia
12.
Toxicon ; 42(2): 191-8, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12906890

RESUMO

A functional pharmacologically-based assay for the brevetoxin group of sodium channel activators was developed using synaptoneurosomes isolated from the brains of CD1 mice. The assay can detect the depolarizing effect of brevetoxin congeners PbTx-2 and PbTx-3 as enhancements of the veratridine-dependent increase in fluorescence of the voltage-sensitive fluorescent probe rhodamine 6G. The assay is relatively rapid and can detect brevetoxin activity in the nanomolar range. The synaptoneurosomal assay has been used to analyse mussel tissue extracts spiked with PbTx-2, and composite toxicity, expressed as PbTx-3 equivalents in extracts of oysters naturally exposed to brevetoxins. In this latter context, the synaptoneurosomal technique was shown to compare favorably with the cytotoxicity assay, the receptor binding assay and HPLC/MS. Our results support the concept that this membrane potential assay detects brevetoxins based on their interaction with sodium channels.


Assuntos
Toxinas Marinhas/análise , Toxinas Marinhas/toxicidade , Potenciais da Membrana/efeitos dos fármacos , Oxocinas/análise , Oxocinas/toxicidade , Agonistas de Canais de Sódio , Sinaptossomos/efeitos dos fármacos , Animais , Bivalves/química , Dinoflagellida/química , Fluorescência , Corantes Fluorescentes , Masculino , Camundongos , Camundongos Endogâmicos , Ostreidae/química , Valores de Referência , Rodaminas , Sensibilidade e Especificidade , Canais de Sódio/metabolismo , Fatores de Tempo , Veratridina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA