Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34948337

RESUMO

Voltage-gated Na+ (Nav) channels are the primary molecular determinant of the action potential. Among the nine isoforms of the Nav channel α subunit that have been described (Nav1.1-Nav1.9), Nav1.1, Nav1.2, and Nav1.6 are the primary isoforms expressed in the central nervous system (CNS). Crucially, these three CNS Nav channel isoforms display differential expression across neuronal cell types and diverge with respect to their subcellular distributions. Considering these differences in terms of their localization, the CNS Nav channel isoforms could represent promising targets for the development of targeted neuromodulators. However, current therapeutics that target Nav channels lack selectivity, which results in deleterious side effects due to modulation of off-target Nav channel isoforms. Among the structural components of the Nav channel α subunit that could be pharmacologically targeted to achieve isoform selectivity, the C-terminal domains (CTD) of Nav channels represent promising candidates on account of displaying appreciable amino acid sequence divergence that enables functionally unique protein-protein interactions (PPIs) with Nav channel auxiliary proteins. In medium spiny neurons (MSNs) of the nucleus accumbens (NAc), a critical brain region of the mesocorticolimbic circuit, the PPI between the CTD of the Nav1.6 channel and its auxiliary protein fibroblast growth factor 14 (FGF14) is central to the generation of electrical outputs, underscoring its potential value as a site for targeted neuromodulation. Focusing on this PPI, we previously developed a peptidomimetic derived from residues of FGF14 that have an interaction site on the CTD of the Nav1.6 channel. In this work, we show that whereas the compound displays dose-dependent effects on the activity of Nav1.6 channels in heterologous cells, the compound does not affect Nav1.1 or Nav1.2 channels at comparable concentrations. In addition, we show that the compound correspondingly modulates the action potential discharge and the transient Na+ of MSNs of the NAc. Overall, these results demonstrate that pharmacologically targeting the FGF14 interaction site on the CTD of the Nav1.6 channel is a strategy to achieve isoform-selective modulation, and, more broadly, that sites on the CTDs of Nav channels interacted with by auxiliary proteins could represent candidates for the development of targeted therapeutics.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Neurônios/metabolismo , Peptidomiméticos/farmacologia , Domínios Proteicos , Animais , Fatores de Crescimento de Fibroblastos/metabolismo , Células HEK293 , Humanos , Camundongos , Simulação de Acoplamento Molecular , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiologia , Ligação Proteica
2.
J Neurophysiol ; 124(2): 510-524, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32667253

RESUMO

Vestibular afferent neurons convey information from hair cells in the peripheral vestibular end organs to central nuclei. Primary vestibular afferent neurons can fire action potentials at high rates and afferent firing patterns vary with the position of nerve terminal endings in vestibular neuroepithelia. Terminals contact hair cells as small bouton or large calyx endings. To investigate the role of Na+ currents (INa) in firing mechanisms, we investigated biophysical properties of INa in calyx-bearing afferents. Whole cell patch-clamp recordings were made from calyx terminals in thin slices of gerbil crista at different postnatal ages: immature [postnatal day (P)5-P8, young (P13-P15), and mature (P30-P45)]. A large transient Na+ current (INaT) was completely blocked by 300 nM tetrodotoxin (TTX) in mature calyces. In addition, INaT was accompanied by much smaller persistent Na+ currents (INaP) and distinctive resurgent Na+ currents (INaR), which were also blocked by TTX. ATX-II, a toxin that slows Na+ channel inactivation, enhanced INaP in immature and mature calyces. 4,9-Anhydro-TTX (4,9-ah-TTX), which selectively blocks Nav1.6 channels, abolished the enhanced INa in mature, but not immature, calyces. Therefore, Nav1.6 channels mediate a component of INaT and INaP in mature calyces, but are minimally expressed at early postnatal days. INaR was expressed in less than one-third of calyces at P6-P8, but expression increased with development, and in mature cristae INaR was frequently found in peripheral calyces. INaR served to increase the availability of Na+ channels following brief membrane depolarizations. In current clamp, the rate and regularity of action potential firing decreased in mature peripheral calyces following 4,9-ah-TTX application. Therefore, Nav1.6 channels are upregulated during development, contribute to INaT, INaP, and INaR, and may regulate excitability by enabling higher mean discharge rates in a subpopulation of mature calyx afferents.NEW & NOTEWORTHY Action potential firing patterns differ between groups of afferent neurons innervating vestibular epithelia. We investigated the biophysical properties of Na+ currents in specialized vestibular calyx afferent terminals during postnatal development. Mature calyces express Na+ currents with transient, persistent, and resurgent components. Nav1.6 channels contribute to resurgent Na+ currents and may enhance firing in peripheral calyx afferents. Understanding Na+ channels that contribute to vestibular nerve responses has implications for developing new treatments for vestibular dysfunction.


Assuntos
Potenciais de Ação/fisiologia , Células Ciliadas Vestibulares/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Sódio , Tetrodotoxina/farmacologia , Nervo Vestibular/fisiologia , Potenciais de Ação/efeitos dos fármacos , Fatores Etários , Animais , Gerbillinae , Células Ciliadas Vestibulares/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Nervo Vestibular/efeitos dos fármacos
3.
Toxicology ; 440: 152488, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32387285

RESUMO

Exposure to pyrethroids, a popular insecticide class that targets voltage-gated Na+ (Nav) channels, has been correlated to an increase in diagnosis of neurodevelopmental disorders, such as attention deficit hyperactive disorder (ADHD), in children. Dysregulation of medium spiny neurons (MSNs) firing in the nucleus accumbens (NAc) is thought to play a critical role in the pathophysiology of ADHD and other neurodevelopmental disorders. The Nav1.6 channel is the primary molecular determinant of MSN firing and is sensitive to modification by pyrethroids. Building on previous studies demonstrating that deltamethrin (DM), a commonly used pyrethroid, leads to use-dependent enhancement of sodium currents, we characterized the effect of the toxin on long-term inactivation (LTI) of the Nav1.6 channel, a parameter known to affect neuronal firing, and characterized changes in MSN intrinsic excitability. We employed whole-cell patch-clamp electrophysiology to measure sodium currents in HEK-293 cells stably expressing Nav1.6 channels and intrinsic excitability of MSNs in the brain slice preparation. We found that in response to repetitive stimulation acute exposure to 10 µM DM potentiated a build-up of residual sodium currents and modified availability of Nav1.6 by inducing LTI. In the NAc, DM modified MSN intrinsic excitability increasing evoked action potential firing frequency and inducing aberrant action potentials with low amplitude and depolarized voltage threshold, phenotypes that could be explained by DM induced changes on the Nav1.6 channel. These results provide a potential initial mechanism of toxicity of DM that could lead to disruption of the NAc circuitry overtime, increasing the risk of ADHD and other neurodevelopmental disorders.


Assuntos
Inseticidas/toxicidade , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nitrilas/toxicidade , Núcleo Accumbens/efeitos dos fármacos , Piretrinas/toxicidade , Bloqueadores dos Canais de Sódio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Accumbens/citologia , Técnicas de Patch-Clamp , Sódio/metabolismo
4.
Biochem Biophys Res Commun ; 526(3): 786-792, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32268959

RESUMO

Hyperactivity in cochlear nucleus (CN) is one of the major neural correlates for tinnitus induction, yet the molecular factors that participate in the neuronal hyperexcitability remain unclear. The present study showed that acute and chronic administrations of salicylate were both capable of inducing reversible tinnitus in rats. The number of GAD 65/67-immunoreactive neurons in the AVCN and DCN was decreased, while the number of VGLUT 1/2-immunoreactive neurons in the AVCN and DCN was increased when rats were experiencing tinnitus, providing evidence for excitatory-inhibitory imbalance in CN is correlated with tinnitus. Interestingly, the expression level of Nav1.6, an important subtype of voltage-gated sodium channels was significantly increased in the DCN and AVCN of rats experiencing tinnitus, the up-regulation of Nav1.6 was returned to normal level following the disappearance of tinnitus. Double-labeling experiments revealed that Nav1.6 expression was down-regulated in the GAD 65/67-positive neurons in the DCN and AVCN of rats experiencing tinnitus. Notably, the percentage of co-localization of Nav1.6 and NeuN-labeling fusiform neurons was markedly increased in the DCN during tinnitus. These findings uncover the tinnitus-associated alteration in Nav1.6, a potential key contributor that can lead to hyperexcitability in CN and contribute to salicylate-induced tinnitus.


Assuntos
Núcleo Coclear/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Ácido Salicílico/metabolismo , Animais , Escala de Avaliação Comportamental , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Zumbido/metabolismo , Regulação para Cima
5.
Ann Neurol ; 87(3): 339-346, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31943325

RESUMO

OBJECTIVE: SCN8A encephalopathy is a developmental and epileptic encephalopathy (DEE) caused by de novo gain-of-function mutations of sodium channel Nav 1.6 that result in neuronal hyperactivity. Affected individuals exhibit early onset drug-resistant seizures, developmental delay, and cognitive impairment. This study was carried out to determine whether reducing the abundance of the Scn8a transcript with an antisense oligonucleotide (ASO) would delay seizure onset and prolong survival in a mouse model of SCN8A encephalopathy. METHODS: ASO treatment was tested in a conditional mouse model with Cre-dependent expression of the pathogenic patient SCN8A mutation p.Arg1872Trp (R1872W). This model exhibits early onset of seizures, rapid progression, and 100% penetrance. An Scn1a +/- haploinsufficient mouse model of Dravet syndrome was also treated. ASO was administered by intracerebroventricular injection at postnatal day 2, followed in some cases by stereotactic injection at postnatal day 30. RESULTS: We observed a dose-dependent increase in length of survival from 15 to 65 days in the Scn8a-R1872W/+ mice treated with ASO. Electroencephalographic recordings were normal prior to seizure onset. Weight gain and activity in an open field were unaffected, but treated mice were less active in a wheel running assay. A single treatment with Scn8a ASO extended survival of Dravet syndrome mice from 3 weeks to >5 months. INTERPRETATION: Reduction of Scn8a transcript by 25 to 50% delayed seizure onset and lethality in mouse models of SCN8A encephalopathy and Dravet syndrome. Reduction of SCN8A transcript is a promising approach to treatment of intractable childhood epilepsies. Ann Neurol 2020;87:339-346.


Assuntos
Encefalopatias/prevenção & controle , Epilepsias Mioclônicas/prevenção & controle , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Animais , Encefalopatias/complicações , Encefalopatias/mortalidade , Relação Dose-Resposta a Droga , Epilepsias Mioclônicas/complicações , Epilepsias Mioclônicas/mortalidade , Feminino , Infusões Intraventriculares , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Canal de Sódio Disparado por Voltagem NAV1.6/administração & dosagem , Oligonucleotídeos Antissenso/farmacologia , Convulsões/complicações , Convulsões/prevenção & controle
6.
Biochem Biophys Res Commun ; 521(2): 340-346, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31668811

RESUMO

Prostate cancer is the most highly diagnosed cancer in men worldwide. It is characterized by high proliferation, great invasion and metastatic potential. Sodium channel subtypes have been identified as highly expressed in different prostate cancer cell lines. In this study, we have screened the negatively charged fractions of Androctonus australis (Aa) scorpion venom to identify active peptides on DU145 prostate cancer cells proliferation. The most active compound was identified to be the sodium channel peptide AaHIV with an IC50 value of 15 µM. At this concentration, AaHIV had low effect on the adhesion of DU145 cells to fibronectin. When compared to other Na+ channel Aa toxins, AaHIV was found to be 2 times more active than AaHI and AaHII on DU145 cells proliferation and slightly less active than AaHII on their adhesion. The three peptides are inactive on DU145 cells migration. AaHIV was found to be 16 times more active than veratridine, asteroidal alkaloid from plants of the lily family widely used as a sodium channel activator. Electrophysiological experiments showed that the AaHIV toxin activates Nav1.6 channel, suggesting that this sodium channel subtype is implicated in the proliferation of DU145 prostate cancer cells.


Assuntos
Neoplasias da Próstata/patologia , Venenos de Escorpião/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Escorpiões , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo
7.
Sci Rep ; 9(1): 16890, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31729429

RESUMO

Multiple voltage-gated Na+ (Nav) channelopathies can be ascribed to subtle changes in the Nav macromolecular complex. Fibroblast growth factor 14 (FGF14) is a functionally relevant component of the Nav1.6 channel complex, a causative link to spinocerebellar ataxia 27 (SCA27) and an emerging risk factor for neuropsychiatric disorders. Yet, how this protein:channel complex is regulated in the cell is still poorly understood. To search for key cellular pathways upstream of the FGF14:Nav1.6 complex, we have developed, miniaturized and optimized an in-cell assay in 384-well plates by stably reconstituting the FGF14:Nav1.6 complex using the split-luciferase complementation assay. We then conducted a high-throughput screening (HTS) of 267 FDA-approved compounds targeting known mediators of cellular signaling. Of the 65 hits initially detected, 24 were excluded based on counter-screening and cellular toxicity. Based on target analysis, potency and dose-response relationships, 5 compounds were subsequently repurchased for validation and confirmed as hits. Among those, the tyrosine kinase inhibitor lestaurtinib was highest ranked, exhibiting submicromolar inhibition of FGF14:Nav1.6 assembly. While providing evidence for a robust in-cell HTS platform that can be adapted to search for any channelopathy-associated regulatory proteins, these results lay the potential groundwork for repurposing cancer drugs for neuropsychopharmacology.


Assuntos
Antineoplásicos , Ensaios de Triagem em Larga Escala/métodos , Mapas de Interação de Proteínas/fisiologia , Agonistas do Canal de Sódio Disparado por Voltagem/isolamento & purificação , Bloqueadores do Canal de Sódio Disparado por Voltagem/isolamento & purificação , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Fatores de Crescimento de Fibroblastos/agonistas , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/química , Células HEK293 , Humanos , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/química , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Ligação Proteica , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo
8.
Toxicon ; 157: 87-92, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30468759

RESUMO

The scorpion venom is a cocktail of many components. Its composition can exhibit a level of plasticity in response to different behavioral and environmental factors, leading to intraspecific variation. The toxicity and specificity of scorpion venoms appear to be taxon-dependent, due to a co-evolutionary interaction with prey and predators, which shaped the composition at the molecular level. The venom regeneration by the venom glands is an asynchronous process, in which particular components are expressed at different stages and at different rates. According to this, it can be reasonably assumed that the regeneration of toxicity in the venom is also asynchronous. In this work, we studied the toxicity regeneration dynamics by the scorpion Centruroides limpidus after full venom depletion by electrical stimulation. For this, we evaluated the toxicity of venom samples extracted at different days post depletion, against insects (crickets) and mammals (humans, by assessing the venom activity on the human voltage-dependent Na+ channel Nav1.6). The regeneration of toxicity against humans lagged behind that against crickets (13 vs 10 days, respectively). Thirteen days after depletion the venom seems to be replenished. Our results show asynchrony in the regeneration of species-specific toxic activity in the venom of Centruroides limpidus. The understanding of the venom regeneration kinetics for the different scorpion species will help to design venom extraction protocols that could maximize the yield and quality of the collected venoms.


Assuntos
Glândulas Exócrinas/fisiologia , Venenos de Escorpião/biossíntese , Venenos de Escorpião/toxicidade , Escorpiões/fisiologia , Animais , Estimulação Elétrica/métodos , Gryllidae/efeitos dos fármacos , Células HEK293 , Humanos , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos
9.
Biomed Pharmacother ; 106: 930-940, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30119265

RESUMO

BACKGROUND: Oxaliplatin-induced cold allodynia is a frequent complication appearing in patients treated with this anti-tumor drug. Since, there are no clear algorithms to overcome this painful condition effectively, it is important to establish novel strategies for its treatment. AIM: In this study, the ability of pregabalin and ambroxol, used as single drugs or in combinations administered in a time-shifted manner to attenuate cold allodynia was assessed in the mouse cold plate test. The hot plate test was additionally used to assess antinociceptive properties of ambroxol in the acute, thermally-induced pain model. Locomotor activity and motor coordination of mice were also evaluated. In silico studies were undertaken to predict potential binding of ambroxol to sodium channel (Nav) subtypes whose overexpression is implicated in the development of oxaliplatin-induced neuropathic pain. KEY FINDINGS: A hyperadditive antiallodynic effect of combined sub-analgesic ambroxol and pregabalin was demonstrated in oxaliplatin-treated mice. This effect was particularly strong when these drugs were given 4 h apart. Both drugs used in combination reduced animals' locomotor activity, but they did not impair motor coordination in the rotarod test. Ambroxol did not show antinociceptive properties in the hot plate test. The molecular docking studies predicted that in mice ambroxol might bind to Nav1.6 and Nav1.9 rather than Nav1.7 and Nav1.8. SIGNIFICANCE: Time-shifted co-administration of sub-analgesic doses of ambroxol and pregabalin effectively attenuates oxaliplatin-induced cold allodynia. Molecular docking model predicts preferential binding of ambroxol to mouse Nav1.6, Nav1.9 channels. This mechanism, if confirmed in vitro, might explain pharmacological activities observed in vivo.


Assuntos
Ambroxol/administração & dosagem , Analgésicos/administração & dosagem , Temperatura Baixa , Hiperalgesia/prevenção & controle , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.9/efeitos dos fármacos , Oxaliplatina , Limiar da Dor/efeitos dos fármacos , Pregabalina/administração & dosagem , Ambroxol/metabolismo , Analgésicos/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Sítios de Ligação , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Quimioterapia Combinada , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Simulação de Acoplamento Molecular , Atividade Motora/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.9/metabolismo , Ligação Proteica , Teste de Desempenho do Rota-Rod , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
10.
J Med Chem ; 61(4): 1730-1736, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29378414

RESUMO

We report the chemical synthesis of scorpion toxin Cn2, a potent and highly selective activator of the human voltage-gated sodium channel NaV1.6. In an attempt to decouple channel activation from channel binding, we also synthesized the first analogue of this toxin, Cn2[E15R]. This mutation caused uncoupling of the toxin's excitatory and depressant activities, effectively resulting in a NaV1.6 inhibitor. In agreement with the in vitro observations, Cn2[E15R] is antinociceptive in mouse models of NaV1.6-mediated pain.


Assuntos
Analgésicos/síntese química , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Mutação Puntual , Venenos de Escorpião/química , Toxinas Biológicas/síntese química , Animais , Humanos , Camundongos , Proteínas Mutantes/farmacologia , Dor/tratamento farmacológico , Mutação Puntual/fisiologia , Escorpiões , Toxinas Biológicas/genética
11.
Naunyn Schmiedebergs Arch Pharmacol ; 390(12): 1255-1270, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28905186

RESUMO

Tricyclic antidepressants (TCAs) and duloxetine are used to treat neuropathic pain. However, the mechanisms underlying their analgesic effects remain unclear. Although many investigators have shown inhibitory effects of antidepressants on voltage-gated sodium channels (Nav) as a possible mechanism of analgesia, to our knowledge, no one has compared effects on the diverse variety of sodium channel α subunits. We investigated the effects of antidepressants on sodium currents in Xenopus oocytes expressing Nav1.2, Nav1.3, Nav1.6, Nav1.7, and Nav1.8 with a ß1 subunit by using whole-cell, two-electrode, voltage clamp techniques. We also studied the role of the ß3 subunit on the effect of antidepressants on Nav1.3. All antidepressants inhibited sodium currents in an inactivated state induced by all five α subunits with ß1. The inhibitory effects were more potent for Nav1.3, Nav1.7, and Nav1.8, which are distributed in dorsal root ganglia, than Nav1.2 and Nav1.6, which are distributed primarily in the central nervous system. The effect of amitriptyline on Nav1.7 with ß1 was most potent with a half-maximal inhibitory concentration (IC50) 4.6 µmol/L. IC50 for amitriptyline on Nav1.3 coexpressed with ß1 was lowered from 8.4 to 4.5 µmol/L by coexpression with ß3. Antidepressants predominantly inhibited the sodium channels expressed in dorsal root ganglia, and amitriptyline has the most potent inhibitory effect. This is the first evidence, to our knowledge, showing the diverse effects of antidepressants on various α subunits. Moreover, the ß3 subunit appears important for inhibition of Nav1.3. These findings may aid better understanding of the mechanisms underlying the pain relieving effects of antidepressants.


Assuntos
Antidepressivos/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.3/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.8/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Amitriptilina/farmacologia , Animais , Antidepressivos Tricíclicos/farmacologia , Cloridrato de Duloxetina/farmacologia , Gânglios Espinais/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.2/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Oócitos , Técnicas de Patch-Clamp , Ratos , Xenopus
12.
Brain ; 139(Pt 8): 2164-81, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27267376

RESUMO

Mutations in brain isoforms of voltage-gated sodium channels have been identified in patients with distinct epileptic phenotypes. Clinically, these patients often do not respond well to classic anti-epileptics and many remain refractory to treatment. Exogenous as well as endogenous cannabinoids have been shown to target voltage-gated sodium channels and cannabidiol has recently received attention for its potential efficacy in the treatment of childhood epilepsies. In this study, we further investigated the ability of cannabinoids to modulate sodium currents from wild-type and epilepsy-associated mutant voltage-gated sodium channels. We first determined the biophysical consequences of epilepsy-associated missense mutations in both Nav1.1 (arginine 1648 to histidine and asparagine 1788 to lysine) and Nav1.6 (asparagine 1768 to aspartic acid and leucine 1331 to valine) by obtaining whole-cell patch clamp recordings in human embryonic kidney 293T cells with 200 µM Navß4 peptide in the pipette solution to induce resurgent sodium currents. Resurgent sodium current is an atypical near threshold current predicted to increase neuronal excitability and has been implicated in multiple disorders of excitability. We found that both mutations in Nav1.6 dramatically increased resurgent currents while mutations in Nav1.1 did not. We then examined the effects of anandamide and cannabidiol on peak transient and resurgent currents from wild-type and mutant channels. Interestingly, we found that cannabidiol can preferentially target resurgent sodium currents over peak transient currents generated by wild-type Nav1.6 as well as the aberrant resurgent and persistent current generated by Nav1.6 mutant channels. To further validate our findings, we examined the effects of cannabidiol on endogenous sodium currents from striatal neurons, and similarly we found an inhibition of resurgent and persistent current by cannabidiol. Moreover, current clamp recordings show that cannabidiol reduces overall action potential firing of striatal neurons. These findings suggest that cannabidiol could be exerting its anticonvulsant effects, at least in part, through its actions on voltage-gated sodium channels, and resurgent current may be a promising therapeutic target for the treatment of epilepsy syndromes.


Assuntos
Anticonvulsivantes/farmacologia , Canabidiol/farmacologia , Epilepsia/tratamento farmacológico , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Neostriado/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Ácidos Araquidônicos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Endocanabinoides/farmacologia , Epilepsia/genética , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1 , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Técnicas de Patch-Clamp , Alcamidas Poli-Insaturadas/farmacologia
13.
Neurotherapeutics ; 13(1): 192-7, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26252990

RESUMO

Mutations in SCN8A are associated with epilepsy and intellectual disability. SCN8A encodes for sodium channel Nav1.6, which is located in the brain. Gain-of-function missense mutations in SCN8A are thought to lead to increased firing of excitatory neurons containing Nav1.6, and therefore to lead to increased seizure susceptibility. We hypothesized that sodium channel blockers could have a beneficial effect in patients with SCN8A-related epilepsy by blocking the overactive Nav1.6 and thereby counteracting the effect of the mutation. Herein, we describe 4 patients with a missense SCN8A mutation and epilepsy who all show a remarkably good response on high doses of phenytoin and loss of seizure control when phenytoin medication was reduced, while side effects were relatively mild. In 2 patients, repeated withdrawal of phenytoin led to the reoccurrence of seizures. Based on the findings in these patients and the underlying molecular mechanism we consider treatment with (high-dose) phenytoin as a possible treatment option in patients with difficult-to-control seizures due to an SCN8A mutation.


Assuntos
Anticonvulsivantes/uso terapêutico , Epilepsia/genética , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Fenitoína/uso terapêutico , Criança , Pré-Escolar , Epilepsia/tratamento farmacológico , Feminino , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Resultado do Tratamento
14.
Mar Drugs ; 13(2): 984-95, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25686275

RESUMO

Tetrodotoxin (TTX) is a potent neurotoxin found in a number of marine creatures including the pufferfish, where it is synthesized by bacteria and accumulated through the food chain. It is a potent and selective blocker of some types of voltage-gated Na+ channel (NaV channel). 4,9-Anhydrotetrodotoxin (4,9-anhydroTTX) was purified from a crude mixture of TTX analogues (such as TTX, 4-epiTTX, 6-epiTTX, 11-oxoTTX and 11-deoxyTTX) by the use of liquid chromatography-fluorescence detection (LC-FLD) techniques. Recently, it has been reported that 4,9-anhydroTTX selectively blocks the activity of NaV1.6 channels with a blocking efficacy 40-160 times higher than that for other TTX-sensitive NaV1.x channel isoforms. However, little attention has been paid to the molecular properties of the α-subunit in NaV1.6 channels and the characteristics of binding of 4,9-anhydroTTX. From a functional point of view, it is important to determine the relative expression of NaV1.6 channels in a wide variety of tissues. The aim of this review is to discuss briefly current knowledge about the pharmacology of 4,9-anhydroTTX, and provide an analysis of the molecular structure of native NaV1.6 channels. In addition, chemical aspects of 4,9-anhydroTTX are briefly covered.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/análogos & derivados , Animais , Humanos , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Bloqueadores dos Canais de Sódio/síntese química , Tetrodotoxina/síntese química , Tetrodotoxina/farmacologia
15.
Anesthesiology ; 121(3): 620-31, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24809977

RESUMO

BACKGROUND: The neurosteroids allopregnanolone and pregnanolone are potent positive modulators of γ-aminobutyric acid type A receptors. Antinociceptive effects of allopregnanolone have attracted much attention because recent reports have indicated the potential of allopregnanolone as a therapeutic agent for refractory pain. However, the analgesic mechanisms of allopregnanolone are still unclear. Voltage-gated sodium channels (Nav) are thought to play important roles in inflammatory and neuropathic pain, but there have been few investigations on the effects of allopregnanolone on sodium channels. METHODS: Using voltage-clamp techniques, the effects of allopregnanolone sulfate (APAS) and pregnanolone sulfate (PAS) on sodium current were examined in Xenopus oocytes expressing Nav1.2, Nav1.6, Nav1.7, and Nav1.8 α subunits. RESULTS: APAS suppressed sodium currents of Nav1.2, Nav1.6, and Nav1.7 at a holding potential causing half-maximal current in a concentration-dependent manner, whereas it markedly enhanced sodium current of Nav1.8 at a holding potential causing maximal current. Half-maximal inhibitory concentration values for Nav1.2, Nav1.6, and Nav1.7 were 12 ± 4 (n = 6), 41 ± 2 (n = 7), and 131 ± 15 (n = 5) µmol/l (mean ± SEM), respectively. The effects of PAS were lower than those of APAS. From gating analysis, two compounds increased inactivation of all α subunits, while they showed different actions on activation of each α subunit. Moreover, two compounds showed a use-dependent block on Nav1.2, Nav1.6, and Nav1.7. CONCLUSION: APAS and PAS have diverse effects on sodium currents in oocytes expressing four α subunits. APAS inhibited the sodium currents of Nav1.2 most strongly.


Assuntos
Pregnanolona/farmacologia , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Animais , Feminino , Canal de Sódio Disparado por Voltagem NAV1.2/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.8/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Canais de Sódio Disparados por Voltagem/fisiologia , Xenopus laevis
16.
Eur J Pharmacol ; 727: 158-66, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24486399

RESUMO

n-butyl-p-aminobenzoate (BAB), a local anesthetic, is administered epidurally in cancer patients to treat pain that is poorly controlled by other drugs that have a number of adverse effects. The purpose of the study was to unravel the mechanisms underlying the apparent selective pain suppressant effect of BAB. We used the whole-cell patch-clamp technique to record Na(+) currents and action potentials (APs) in dissociated, nociceptive dorsal root ganglion (DRG) cells from rats, two types of peripheral sensory neuron Na(+) channels (Nav1.7 and Nav1.8), and the motor neuron-specific Na(+) channel (Nav1.6) expressed in HEK293 cells. BAB (1-100µM) inhibited, in a concentration-dependent manner, the depolarization evoked repetitive firing in DRG cells, the three types of Na(+) current expressed in HEK293 cells, and the TTXr Na(+) current of the DRG neurons. BAB induced a use-dependent block that caused a shift of the inactivation curve in the hyperpolarizing direction. BAB enhanced the onset of slow inactivation of Nav1.7 and Nav1.8 currents but not of Nav1.6 currents. At clinically relevant concentrations (1-100µM), BAB is thus a more potent inhibitor of peripheral TTX-sensitive TTXs, Nav1.7 and TTX-resistant NaV1.8 Na(+) channels than of motor neuron axonal Nav1.6 Na(+) channels. BAB had similar effects on the TTXr Na(+) channels of rat DRG neurons and Nav1.8 channels expressed in HEK293 cells. The observed selectivity of BAB in treating cancer pain may be due to an enhanced and selective responsiveness of Na(+) channels in nociceptive neurons to this local anesthetic.


Assuntos
Anestésicos Locais/farmacologia , Benzocaína/análogos & derivados , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Potenciais de Ação , Animais , Benzocaína/farmacologia , Relação Dose-Resposta a Droga , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Masculino , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.8/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Neurônios/metabolismo , Ratos Sprague-Dawley , Fatores de Tempo , Transfecção , Canais de Sódio Disparados por Voltagem/genética , Canais de Sódio Disparados por Voltagem/metabolismo
17.
Epilepsy Res ; 106(1-2): 17-28, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23886654

RESUMO

Voltage-gated Na(+) channels control neuronal excitability and are the primary target for the majority of anti-epileptic drugs. This study investigates the (sub)cellular expression patterns of three important brain-associated Na(+) channel α subunits: NaV1.1, NaV1.2 and NaV1.6 during epileptogenesis (induced by kainic acid) using time points that cover the period from induction to the chronic phase of epilepsy. NaV1.1 immunoreactivity was persistently reduced at 1 day, 3 weeks and 2 months after SE in CA1 and CA3. About 50% of the NaV1.1-positive interneurons was lost at one day after SE in all regions investigated. In the hilus a similar reduction in NeuN-positive neurons was found, while in the CA1 and CA3 region the loss in NeuN-positive neurons only reached 15% in the chronic phase of epilepsy. This implies a stronger shift in the balance between excitation and inhibition toward excitation in the CA1 and CA3 region than in the hilus. NaV1.2 immunoreactivity in the inner molecular layer of the dentate gyrus was lower than control at 1 day after SE. It increased at 3 weeks and 2 months after SE in the inner molecular layer and overlapped with sprouted mossy fibers. NaV1.6 immunoreactivity in the dendritic region of CA1 and CA3 was persistently reduced at all time-points during epileptogenesis. Some astrocytes expressed NaV1.1 and NaV1.6 at 3 weeks after SE. Expression data alone are not sufficient to explain changes in network stability, or infer causality in epileptogenesis. These results demonstrate that hippocampal sub-regional expression of NaV1.1, NaV1.2 and NaV1.6 Na(+) channel α subunits is altered during epileptogenesis in a time and location specific way. This implies that understanding epileptogenesis has to take into account several distinct and type-specific changes in sodium channel expression.


Assuntos
Convulsivantes , Epilepsia/induzido quimicamente , Epilepsia/metabolismo , Agonistas de Aminoácidos Excitatórios , Hipocampo/metabolismo , Ácido Caínico , Canal de Sódio Disparado por Voltagem NAV1.1/biossíntese , Canal de Sódio Disparado por Voltagem NAV1.2/biossíntese , Canal de Sódio Disparado por Voltagem NAV1.6/biossíntese , Animais , Região CA1 Hipocampal/metabolismo , Região CA3 Hipocampal/metabolismo , Interpretação Estatística de Dados , Eletrodos Implantados , Eletroencefalografia/efeitos dos fármacos , Epilepsia/patologia , Imunofluorescência , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Imuno-Histoquímica , Interneurônios/metabolismo , Masculino , Canal de Sódio Disparado por Voltagem NAV1.1/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.2/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/fisiopatologia , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/fisiopatologia
18.
J Neurophysiol ; 110(5): 1144-57, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23741036

RESUMO

During epileptogenesis a series of molecular and cellular events occur, culminating in an increase in neuronal excitability, leading to seizure initiation. The entorhinal cortex has been implicated in the generation of epileptic seizures in both humans and animal models of temporal lobe epilepsy. This hyperexcitability is due, in part, to proexcitatory changes in ion channel activity. Sodium channels play an important role in controlling neuronal excitability, and alterations in their activity could facilitate seizure initiation. We sought to investigate whether medial entorhinal cortex (mEC) layer II neurons become hyperexcitable and display proexcitatory behavior of Na channels during epileptogenesis. Experiments were conducted 7 days after electrical induction of status epilepticus (SE), a time point during the latent period of epileptogenesis and before the onset of seizures. mEC layer II stellate neurons from post-SE animals were hyperexcitable, eliciting action potentials at higher frequencies compared with control neurons. Na channel currents recorded from post-SE neurons revealed increases in Na current amplitudes, particularly persistent and resurgent currents, as well as depolarized shifts in inactivation parameters. Immunocytochemical studies revealed increases in voltage-gated Na (Nav) 1.6 isoform levels. The toxin 4,9-anhydro-tetrodotoxin, which has greater selectivity for Nav1.6 over other Na channel isoforms, suppressed neuronal hyperexcitability, reduced macroscopic Na currents, persistent and resurgent Na current densities, and abolished depolarized shifts in inactivation parameters in post-SE neurons. These studies support a potential role for Nav1.6 in facilitating the hyperexcitability of mEC layer II neurons during epileptogenesis.


Assuntos
Córtex Entorrinal/fisiopatologia , Epilepsia/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Neurônios/fisiologia , Animais , Técnicas In Vitro , Masculino , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Ratos , Ratos Sprague-Dawley , Sódio/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/análogos & derivados , Tetrodotoxina/farmacologia , Fatores de Tempo
19.
Pflugers Arch ; 464(5): 493-502, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22986623

RESUMO

Patch-clamp experiments were performed to investigate the molecular properties of resurgent-like currents in single smooth muscle cells dispersed from mouse vas deferens, utilizing both Na(V)1.6-null mice (Na(V)1.6(-/-)), lacking the expression of the Scn8a Na(+) channel gene, and their wild-type littermates (Na(V)1.6(+/+)). Na(V)1.6 immunoreactivity was clearly visible in dispersed smooth muscle cells obtained from Na(V)1.6(+/+), but not Na(V)1.6(-/-), vas deferens. Following a depolarization to +30 mV from a holding potential of -70 mV (to produce maximal inactivation of the Na(+) current), repolarization to voltages between -60 and +20 mV elicited a tetrodotoxin (TTX)-sensitive inward current in Na(V)1.6(+/+), but not Na(V)1.6(-/-), vas deferens myocytes. The resurgent-like current in Na(V)1.6(+/+) vas deferens myocytes peaked at approximately -20 mV in the current-voltage relationship. The peak amplitude of the resurgent-like current remained at a constant level when the membrane potential was repolarized to -20 mV following the application of depolarizing rectangular pulses to more positive potentials than +20 mV. 4,9-Anhydrotetrodotoxin (4,9-anhydroTTX), a selective Na(V)1.6 blocking toxin, purified from a crude mixture of TTX analogues by LC-FLD techniques, reversibly suppressed the resurgent-like currents. ß-Pompilidotoxin, a voltage-gated Na(+) channel activator, evoked sustained resurgent-like currents in Na(V)1.6(+/+) but not Na(V)1.6(-/-) murine vas deferens myocytes. These results strongly indicate that, primarily, resurgent-like currents are generated as a result of Na(V)1.6 channel activity.


Assuntos
Potenciais de Ação , Miócitos de Músculo Liso/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Potenciais de Ação/genética , Animais , Proteínas de Insetos/farmacologia , Masculino , Camundongos , Camundongos Mutantes , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Tetrodotoxina/farmacologia , Ducto Deferente/citologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Venenos de Vespas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA