Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 41(44): 9141-9162, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34544832

RESUMO

The potassium channel Kv1.6 has recently been implicated as a major modulatory channel subunit expressed in primary nociceptors. Furthermore, its expression at juxtaparanodes of myelinated primary afferents is induced following traumatic nerve injury as part of an endogenous mechanism to reduce hyperexcitability and pain-related hypersensitivity. In this study, we compared two mouse models of constitutive Kv1.6 knock-out (KO) achieved by different methods: traditional gene trap via homologous recombination and CRISPR-mediated excision. Both Kv1.6 KO mouse lines exhibited an unexpected reduction in sensitivity to noxious heat stimuli, to differing extents: the Kv1.6 mice produced via gene trap had a far more significant hyposensitivity. These mice (Kcna6lacZ ) expressed the bacterial reporter enzyme LacZ in place of Kv1.6 as a result of the gene trap mechanism, and we found that their central primary afferent presynaptic terminals developed a striking neurodegenerative phenotype involving accumulation of lipid species, development of "meganeurites," and impaired transmission to dorsal horn wide dynamic range neurons. The anatomic defects were absent in CRISPR-mediated Kv1.6 KO mice (Kcna6-/-) but were present in a third mouse model expressing exogenous LacZ in nociceptors under the control of a Nav1.8-promoted Cre recombinase. LacZ reporter enzymes are thus intrinsically neurotoxic to sensory neurons and may induce pathologic defects in transgenic mice, which has confounding implications for the interpretation of gene KOs using lacZ Nonetheless, in Kcna6-/- mice not affected by LacZ, we demonstrated a significant role for Kv1.6 regulating acute noxious thermal sensitivity, and both mechanical and thermal pain-related hypersensitivity after nerve injury.SIGNIFICANCE STATEMENT In recent decades, the expansion of technologies to experimentally manipulate the rodent genome has contributed significantly to the field of neuroscience. While introduction of enzymatic or fluorescent reporter proteins to label neuronal populations is now commonplace, often potential toxicity effects are not fully considered. We show a role of Kv1.6 in acute and neuropathic pain states through analysis of two mouse models lacking Kv1.6 potassium channels: one with additional expression of LacZ and one without. We show that LacZ reporter enzymes induce unintended defects in sensory neurons, with an impact on behavioral data outcomes. To summarize we highlight the importance of Kv1.6 in recovery of normal sensory function following nerve injury, and careful interpretation of data from LacZ reporter models.


Assuntos
Técnicas de Inativação de Genes/efeitos adversos , Genes Reporter , Canal de Potássio Kv1.6/genética , Óperon Lac , Neuralgia/metabolismo , Nociceptores/metabolismo , Animais , Sistemas CRISPR-Cas , Feminino , Técnicas de Inativação de Genes/métodos , Integrases/metabolismo , Canal de Potássio Kv1.6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nociceptores/patologia , Sinapses/metabolismo , Sinapses/patologia
2.
Mar Drugs ; 18(8)2020 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-32823677

RESUMO

Recently, Conorfamide-Sr3 (CNF-Sr3) was isolated from the venom of Conus spurius and was demonstrated to have an inhibitory concentration-dependent effect on the Shaker K+ channel. The voltage-gated potassium channels play critical functions on cellular signaling, from the regeneration of action potentials in neurons to the regulation of insulin secretion in pancreatic cells, among others. In mammals, there are at least 40 genes encoding voltage-gated K+ channels and the process of expression of some of them may include alternative splicing. Given the enormous variety of these channels and the proven use of conotoxins as tools to distinguish different ligand- and voltage-gated ion channels, in this work, we explored the possible effect of CNF-Sr3 on four human voltage-gated K+ channel subtypes homologous to the Shaker channel. CNF-Sr3 showed a 10 times higher affinity for the Kv1.6 subtype with respect to Kv1.3 (IC50 = 2.7 and 24 µM, respectively) and no significant effect on Kv1.4 and Kv1.5 at 10 µM. Thus, CNF-Sr3 might become a novel molecular probe to study diverse aspects of human Kv1.3 and Kv1.6 channels.


Assuntos
Venenos de Moluscos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Animais , Caramujo Conus , Ativação do Canal Iônico , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.4/antagonistas & inibidores , Canal de Potássio Kv1.4/genética , Canal de Potássio Kv1.4/metabolismo , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/metabolismo , Canal de Potássio Kv1.6/antagonistas & inibidores , Canal de Potássio Kv1.6/genética , Canal de Potássio Kv1.6/metabolismo , Potenciais da Membrana , Oócitos , Superfamília Shaker de Canais de Potássio/genética , Superfamília Shaker de Canais de Potássio/metabolismo , Xenopus laevis
3.
Sci Rep ; 6: 20322, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26831940

RESUMO

Cholinergic regulation of arterial luminal diameter involves intricate network of intercellular communication between the endothelial and smooth muscle cells that is highly dependent on the molecular mediators released by the endothelium. Albeit the well-recognized contribution of nitric oxide (NO) towards vasodilation, the identity of compensatory mechanisms that maintain vasomotor tone when NO synthesis is deranged remain largely unknown in the ophthalmic artery. This is the first study to identify the vasodilatory signalling mechanisms of the ophthalmic artery employing wild type mice. Acetylcholine (ACh)-induced vasodilation was only partially attenuated when NO synthesis was inhibited. Intriguingly, the combined blocking of cytochrome P450 oxygenase (CYP450) and lipoxygenase (LOX), as well as CYP450 and gap junctions, abolished vasodilation; demonstrating that the key compensatory mechanisms comprise arachidonic acid metabolites which, work in concert with gap junctions for downstream signal transmission. Furthermore, the voltage-gated potassium ion channel, Kv1.6, was functionally relevant in mediating vasodilation. Its localization was found exclusively in the smooth muscle. In conclusion, ACh-induced vasodilation of mouse ophthalmic artery is mediated in part by NO and predominantly via arachidonic acid metabolites, with active involvement of gap junctions. Particularly, the Kv1.6 channel represents an attractive therapeutic target in ophthalmopathologies when NO synthesis is compromised.


Assuntos
Artéria Oftálmica/fisiologia , Acetilcolina/farmacologia , Animais , Fatores Biológicos/metabolismo , Colinérgicos/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Epoprostenol/farmacologia , Canal de Potássio Kv1.6/metabolismo , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Artéria Oftálmica/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Vasodilatação/efeitos dos fármacos
4.
Biochem Pharmacol ; 98(4): 614-28, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26549367

RESUMO

Clinical use of non-steroidal anti-inflammatory drugs (NSAIDs) is well known to cause gastrointestinal ulcer formation via several mechanisms that include inhibiting epithelial cell migration and mucosal restitution. The drug-affected signaling pathways that contribute to inhibition of migration by NSAIDs are poorly understood, though previous studies have shown that NSAIDs depolarize membrane potential and suppress expression of calpain proteases and voltage-gated potassium (Kv) channel subunits. Kv channels play significant roles in cell migration and are targets of NSAID activity in white blood cells, but the specific functional effects of NSAID-induced changes in Kv channel expression, particularly on cell migration, are unknown in intestinal epithelial cells. Accordingly, we investigated the effects of NSAIDs on expression of Kv1.3, 1.4, and 1.6 in vitro and/or in vivo and evaluated the functional significance of loss of Kv subunit expression. Indomethacin or NS-398 reduced total and plasma membrane protein expression of Kv1.3 in cultured intestinal epithelial cells (IEC-6). Additionally, depolarization of membrane potential with margatoxin (MgTx), 40mM K(+), or silencing of Kv channel expression with siRNA significantly reduced IEC-6 cell migration and disrupted calpain activity. Furthermore, in rat small intestinal epithelia, indomethacin and NS-398 had significant, yet distinct, effects on gene and protein expression of Kv1.3, 1.4, or 1.6, suggesting that these may be clinically relevant targets. Our results show that inhibition of epithelial cell migration by NSAIDs is associated with decreased expression of Kv channel subunits, and provide a mechanism through which NSAIDs inhibit cell migration and may contribute to NSAID-induced gastrointestinal (GI) toxicity.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Calpaína/antagonistas & inibidores , Movimento Celular/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Animais , Calpaína/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.4/antagonistas & inibidores , Canal de Potássio Kv1.4/metabolismo , Canal de Potássio Kv1.6/antagonistas & inibidores , Canal de Potássio Kv1.6/metabolismo , Potenciais da Membrana/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
FEBS J ; 281(15): 3433-45, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24924920

RESUMO

The functions of voltage-gated potassium (Kv) channels in neurons have been well defined, whereas their roles in glial cells are not fully understood. Kv1.1, Kv1.3 and Kv1.6 are endogenously expressed in C6 astrocytoma cells, but their trafficking and subcellular localization have not been well studied. In C6 cells, Kv1.1 was localized to the cell surface, Kv1.3 was predominantly localized in the cis-Golgi, and Kv1.6 was enriched in the endoplasmic reticulum. Disruption of the Golgi stacks with brefeldin A treatment redirected Kv1.3 to the endoplasmic reticulum, further confirming that Kv1.3 was localized in the Golgi. Denaturing and reducing immunoblot analysis identified an expected Kv1.3 monomer and an unexpected Kv1.3 dimer/aggregate. These two forms had different protein half-lives: that of the monomer form T1/2 was 5.1 h, whereas the dimer/aggregate form was stable over the 8-h measurement period. The Kv1.3 dimer/aggregate form on immunoblots appeared to be correlated with its Golgi retention, based on examination with several cell types that expressed Kv1.3. Glycosidase treatment showed that Kv1.3 contained complex-type N-glycans terminated with sialic acids, suggesting that Kv1.3 had traveled to the trans-Golgi network for sialylation before it was recycled to the cis-Golgi for retention. Inhibition of N-glycosylation did not affect Kv1.3 localization, indicating that N-glycans did not play a role in its Golgi retention. Thus, Kv1.3 appears to be distributed to the cis-Golgi membrane of rat astrocytes in a similar way as a Golgi resident protein, and this unusual distribution appears to be correlated with its SDS/2-mercaptoethanol-resistant dimer/aggregate forms on immunoblots.


Assuntos
Astrócitos/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.6/metabolismo , Animais , Linhagem Celular Tumoral , Glicosilação , Humanos , Membranas Intracelulares/metabolismo , Células MCF-7 , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Transporte Proteico , Ratos
6.
BMC Genomics ; 13: 635, 2012 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-23157480

RESUMO

BACKGROUND: LEF1/TCF transcription factors and their activator ß-catenin are effectors of the canonical Wnt pathway. Although Wnt/ß-catenin signaling has been implicated in neurodegenerative and psychiatric disorders, its possible role in the adult brain remains enigmatic. To address this issue, we sought to identify the genetic program activated by ß-catenin in neurons. We recently showed that ß-catenin accumulates specifically in thalamic neurons where it activates Cacna1g gene expression. In the present study, we combined bioinformatics and experimental approaches to find new ß-catenin targets in the adult thalamus. RESULTS: We first selected the genes with at least two conserved LEF/TCF motifs within the regulatory elements. The resulting list of 428 putative LEF1/TCF targets was significantly enriched in known Wnt targets, validating our approach. Functional annotation of the presumed targets also revealed a group of 41 genes, heretofore not associated with Wnt pathway activity, that encode proteins involved in neuronal signal transmission. Using custom polymerase chain reaction arrays, we profiled the expression of these genes in the rat forebrain. We found that nine of the analyzed genes were highly expressed in the thalamus compared with the cortex and hippocampus. Removal of nuclear ß-catenin from thalamic neurons in vitro by introducing its negative regulator Axin2 reduced the expression of six of the nine genes. Immunoprecipitation of chromatin from the brain tissues confirmed the interaction between ß-catenin and some of the predicted LEF1/TCF motifs. The results of these experiments validated four genes as authentic and direct targets of ß-catenin: Gabra3 for the receptor of GABA neurotransmitter, Calb2 for the Ca(2+)-binding protein calretinin, and the Cacna1g and Kcna6 genes for voltage-gated ion channels. Two other genes from the latter cluster, Cacna2d2 and Kcnh8, appeared to be regulated by ß-catenin, although the binding of ß-catenin to the regulatory sequences of these genes could not be confirmed. CONCLUSIONS: In the thalamus, ß-catenin regulates the expression of a novel group of genes that encode proteins involved in neuronal excitation. This implies that the transcriptional activity of ß-catenin is necessary for the proper excitability of thalamic neurons, may influence activity in the thalamocortical circuit, and may contribute to thalamic pathologies.


Assuntos
Canais de Cálcio Tipo T/genética , Canal de Potássio Kv1.6/genética , Neurônios/metabolismo , Receptores de GABA-A/genética , Proteína G de Ligação ao Cálcio S100/genética , Tálamo/metabolismo , beta Catenina/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Sítios de Ligação , Calbindina 2 , Canais de Cálcio Tipo T/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Canal de Potássio Kv1.6/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Masculino , Neurônios/citologia , Neurotransmissores , Cultura Primária de Células , Regiões Promotoras Genéticas , Ligação Proteica , Ratos , Receptores de GABA-A/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Transdução de Sinais , Tálamo/citologia , Ativação Transcricional , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
7.
Biochem Pharmacol ; 83(9): 1307-17, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22305749

RESUMO

Given their medical importance, most attention has been paid toward the venom composition of scorpions of the Buthidae family. Nevertheless, research has shown that the venom of scorpions of other families is also a remarkable source of unique peptidyl toxins. The κ-KTx family of voltage-gated potassium channel (VGPC) scorpion toxins is hereof an example. From the telson of the scorpion Heterometrus laoticus (Scorpionidae), a peptide, HelaTx1, with unique primary sequence was purified through HPLC and sequenced by Edman degradation. Based on the amino acid sequence, the peptide could be cloned and the cDNA sequence revealed. HelaTx1 was chemically synthesized and functionally characterized on VGPCs of the Shaker-related, Shab-related, Shaw-related and Shal-related subfamilies. Furthermore, the toxin was also tested on small- and intermediate conductance Ca(2+)-activated K(+) channels. From the channels studied, K(v)1.1 and K(v)1.6 were found to be the most sensitive (K(v)1.1 EC(50)=9.9±1.6 µM). The toxin did not alter the activation of the channels. Competition experiments with TEA showed that the toxin is a pore blocker. Mutational studies showed that the residues E353 and Y379 in the pore of K(v)1.1 act as major interaction points for binding of the toxin. Given the amino acid sequence, the predicted secondary structure and the biological activity on VGPCs, HelaTx1 should be included in the κ-KTX family. Based on a phylogenetic study, we rearranged this family of VGPC toxins into five subfamilies and suggest that HelaTx1 is the first member of the new κ-KTx5 subfamily.


Assuntos
Peptídeos/genética , Peptídeos/isolamento & purificação , Peptídeos/farmacologia , Venenos de Aranha/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.6/metabolismo , Dados de Sequência Molecular , Mutação , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Técnicas de Patch-Clamp , Peptídeos/metabolismo , Filogenia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Estrutura Secundária de Proteína , Escorpiões/química , Homologia de Sequência de Aminoácidos , Proteínas de Xenopus/metabolismo , Xenopus laevis
8.
Biochem J ; 438(2): 389-96, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21352098

RESUMO

Assembly of distinct α subunits of Kv1 (voltage-gated K(+) channels) into tetramers underlies the diversity of their outward currents in neurons. Kv1.4-containing channels normally exhibit N-type rapid inactivation, mediated through an NIB (N-terminal inactivation ball); this can be over-ridden if associated with a Kv1.6 α subunit, via its NIP (N-type inactivation prevention) domain. Herein, NIP function was shown to require positioning of Kv1.6 adjacent to the Kv1.4 subunit. Using a recently devised gene concatenation, heterotetrameric Kv1 channels were expressed as single-chain proteins on the plasmalemma of HEK (human embryonic kidney)-293 cells, so their constituents could be arranged in different positions. Placing the Kv1.4 and 1.6 genes together, followed by two copies of Kv1.2, yielded a K(+) current devoid of fast inactivation. Mutation of critical glutamates within the NIP endowed rapid inactivation. Moreover, separating Kv1.4 and 1.6 with a copy of Kv1.2 gave a fast-inactivating K(+) current with steady-state inactivation shifted to more negative potentials and exhibiting slower recovery, correlating with similar inactivation kinetics seen for Kv1.4-(1.2)(3). Alternatively, separating Kv1.4 and 1.6 with two copies of Kv1.2 yielded slow-inactivating currents, because in this concatamer Kv1.4 and 1.6 should be together. These findings also confirm that the gene concatenation can generate K(+) channels with α subunits in pre-determined positions.


Assuntos
Ativação do Canal Iônico , Canal de Potássio Kv1.4/metabolismo , Canal de Potássio Kv1.6/metabolismo , Animais , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Canal de Potássio Kv1.6/química , Mutagênese/genética , Plasmídeos/genética , Estrutura Terciária de Proteína , Subunidades Proteicas/metabolismo , Ratos
9.
J Neurosci ; 30(22): 7554-62, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20519529

RESUMO

Slo2 Na(+)-activated potassium channels are widely expressed in neurons and other cells, such as kidney, heart, and skeletal muscle. Although their important physiological roles continue to be appreciated, molecular determinants responsible for sensing intracellular Na(+) remain unknown. Here we report identification of an Na(+) regulatory site, similar to an Na(+) coordination motif described in Kir channels, localized in the RCK2 domain of Slo2.2 channels. Molecular simulations of the homology-modeled Slo2.2 RCK2 domain provided structural insights into the organization of this Na(+) coordination site. Furthermore, free energy calculations reproduced the experimentally derived monovalent cation selectivity. Our results suggest that Slo2.2 and Kir channels share a similar mechanism to coordinate Na(+). The localization of an Na(+) sensor within the RCK2 domain of Slo2.2 further supports the role of RCK (regulators of conductance of K(+)) domains of Slo channels in coupling ion sensing to channel gating.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Canal de Potássio Kv1.6/metabolismo , Sódio/metabolismo , Sequência de Aminoácidos , Aminoácidos/genética , Animais , Fenômenos Biofísicos/efeitos dos fármacos , Fenômenos Biofísicos/genética , Fenômenos Biofísicos/fisiologia , Cloretos/farmacologia , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Canal de Potássio Kv1.6/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Microinjeções/métodos , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Oócitos , Técnicas de Patch-Clamp , Sódio/farmacologia , Xenopus laevis
10.
Brain Res ; 1255: 170-9, 2009 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-19109933

RESUMO

Cerebro Spinal Fluid (CSF) from patients with ALS has been documented to have a toxic effect on motor neurons both in vivo and in vitro. Here we show that the CSF from Amyotrophic Lateral Sclerosis (ALS) patients (ALS-CSF) has the potential to perturb ion channel expression, specifically the Na(v)1.6, and K(v)1.6 channels in newborn rat spinal motor neurons both in vivo and in vitro. ALS-CSF and CSF from nonALS patients (nonALS-CSF) were intrathecally injected into 3-day-old rat pups at the rate of 1 microl/2.5 min using a microinjector. In addition, embryonic rat spinal cord cultures were also exposed to 10% ALS or nonALS-CSF on the 9th day in vitro (9DIV) in serum free DMEM medium. After 48 h of CSF exposure, the cultures and the spinal cord sections were processed for immunostaining of the above mentioned ion channels. We observed a decrease in the expression of Na(v)1.6 and K(v)1.6 channels in motor neurons in ALS-CSF treated group, and the presence of trophic factors like Brain Derived Neurotrophic Factor (BDNF) and Ciliary Neurotrophic Factor CNTF partially reversed the effects produced by ALS-CSF. Altered expression of these voltage-gated channels may interfere with the electrical activity of motor neurons, and thereby lead to the degeneration of neurons.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Fatores Biológicos/toxicidade , Líquido Cefalorraquidiano , Canal de Potássio Kv1.6/metabolismo , Neurônios Motores/efeitos dos fármacos , Canais de Sódio/metabolismo , Animais , Animais Recém-Nascidos , Fatores Biológicos/líquido cefalorraquidiano , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Injeções Espinhais , Microinjeções , Neurônios Motores/metabolismo , Ratos , Ratos Wistar , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
11.
In Silico Biol ; 7(2): 175-86, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17688443

RESUMO

A novel conotoxin pl14a containing 25 amino acid residues with an amidated C-terminus from vermivorous cone snail, Conus planorbis belongs to J-conotoxin superfamily and this is the first conotoxin, which inhibits both nicotinic acetylcholine receptor subtypes and Kv1.6 channel. We have attempted through bioinformatics approaches to elucidate the extent of specificity of pl14a towards Kv1 channel subtypes (Kv1.1-Kv1.6). Our work provides rationale for the relatively high specificity and binding mode of pl14a to Kv1.6 channel. The pl14a peptide contains two types of structural elements, namely the putative dyad (Lys18 and Tyr19) and basic residue ring constituted of arginine residues. We have carried out in silico docking studies so as to assess the contribution of one or combination of both structural elements of pl14a in blocking of Kv1.6 channel. For this purpose, we have built by homology modelling, the theoretical 3D structure of Kv1.6 channel based on the available crystal structure of mammalian shaker Kv1.2 channel. Docking studies suggest that positively charged residues ring may be involved in the blocking mechanism of Kv1.6 channel. The models suggest that the peptide interacts with negatively charged extracellular loops and pore-mouth of the potassium channel and blocks the channel by covering the pore as a lid, akin to previously proposed blocking mechanism of kappaM-conotoxin RIIIK from Conus radiatus to Tsha1 potassium channel. The newly detected pharmacophore for pl14a interacting with Kv1.6 channel provides a pointer to experimental work to validate the observations made here. Based on differences in the number and distribution of the positively-charged residues in other conopeptides from the J-superfamily, we hypothesize different selectivity profiles against subtypes of the potassium channels for these conopeptides.


Assuntos
Conotoxinas/metabolismo , Caramujo Conus/química , Canal de Potássio Kv1.6/metabolismo , Neurotoxinas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Biologia Computacional , Canal de Potássio Kv1.6/antagonistas & inibidores , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Ligação Proteica , Conformação Proteica , Homologia de Sequência de Aminoácidos
12.
Br J Pharmacol ; 150(8): 1003-10, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17339841

RESUMO

BACKGROUND AND PURPOSE: Methylamine is an endogenous aliphatic amine exhibiting anorexigenic properties in mice. The aim of this work was to show whether methylamine also modifies feeding behaviour in rats and, if so, to identify the mediator(s) responsible for such effects. EXPERIMENTAL APPROACH: Microdialysis experiments with the probe inserted in the periventricular hypothalamic nucleus were carried out in 12 h starved, freely moving rats. Collected perfusate samples following methylamine injection (i.c.v.) were analysed for nitric oxide by chemiluminescence and for dopamine and 5-hydroxytryptamine content by HPLC. Kv1.6 potassium channel expression was reduced by antisense strategy and this decrease quantified by semi-quantitative RT-PCR analysis. KEY RESULTS: Methylamine showed biphasic dose-related effects on rat feeding. At doses of 15-30 microg per rat, it was hyperphagic whereas higher doses (60-80 microg) were hypophagic. Methylamine stimulated central nitric oxide (+115% vs. basal) following hyperphagic and dopamine release (60% over basal values) at hypophagic doses, respectively. Treatment with L-N(G)-nitro-L-arginine-methyl ester (i.c.v. 2 microg 10 microl(-1)) or with alpha-methyl-p-tyrosine (i.p. 100 mg kg(-1)) before methylamine injection, reduced nitric oxide output and hyperphagia, or dopamine release and hypophagia respectively. Moreover, hypophagia and hyperphagia, as well as nitric oxide and dopamine release were significantly reduced by down-regulating brain Kv1.6 potassium channel expression. CONCLUSIONS AND IMPLICATIONS: The effects of methylamine on feeding depend on the hypothalamic release of nitric oxide and dopamine as a result of interaction at the Kv1.6 channels. The study of methylamine levels in the CNS may provide new perspectives on the physiopathology of alimentary behaviour.


Assuntos
Depressores do Apetite/metabolismo , Regulação do Apetite , Dopamina/metabolismo , Jejum , Hipotálamo/metabolismo , Metilaminas/metabolismo , Óxido Nítrico/metabolismo , Animais , Depressores do Apetite/administração & dosagem , Regulação do Apetite/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Hiperfagia/metabolismo , Hiperfagia/fisiopatologia , Hipotálamo/efeitos dos fármacos , Injeções Intraventriculares , Canal de Potássio Kv1.6/efeitos dos fármacos , Canal de Potássio Kv1.6/genética , Canal de Potássio Kv1.6/metabolismo , Masculino , Metilaminas/administração & dosagem , Microdiálise , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Ratos , Ratos Wistar , Serotonina/metabolismo , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/antagonistas & inibidores , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Metiltirosina/farmacologia
13.
J Neurophysiol ; 96(4): 1887-901, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16624997

RESUMO

The axon initial segment plays important roles in spike initiation and invasion of axonal spikes into the soma. Among primary sensory neurons, those in the mesencephalic trigeminal nucleus (MTN) are exceptional in their ability to initiate soma spikes (S-spikes) in response to synaptic inputs, consequently displaying two kinds of S-spikes, one caused by invasion of an axonal spike arising from the sensory receptor and the other initiated by somatic inputs. We investigated where spikes are initiated in such MTN neurons and whether there are any differences between the two kinds of S-spikes. Simultaneous patch-clamp recordings from the soma and axon hillock revealed a spike-backpropagation from the spike-initiation site in the stem axon to the soma in response to 1-ms somatic current pulse, which disclosed the delayed emergence of S-spikes after the current-pulse offset. These initiated S-spikes were smaller in amplitude than S-spikes generated by stimulation of the stem axon; however, 4-AP (< or =0.5 mM) eliminated the amplitude difference. Furthermore, 4-AP dramatically shortened the delay in spike initiation without affecting the spike-backpropagation time in the stem axon, whereas it substantially prolonged the refractory period of S-spikes arising from axonal-spike invasion without significantly affecting that of presumed axonal spikes. These observations suggest that 4-AP-sensitive K(+) currents exert two opposing effects on S-spikes depending on their origins: suppression of spike initiation and facilitation of axonal-spike invasion at higher frequencies. Consistent with these findings, strong immunoreactivities for Kv1.1 and Kv1.6, among 4-AP-sensitive and low-voltage-activated Kv1 family examined, were detected in the soma but not in the stem axon of MTN neurons.


Assuntos
4-Aminopiridina/farmacologia , Potenciais de Ação/fisiologia , Neurônios Aferentes/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/fisiologia , Núcleos do Trigêmeo/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Axônios/fisiologia , Regulação da Expressão Gênica , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.6/genética , Canal de Potássio Kv1.6/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mesencéfalo/fisiologia , Neurônios Aferentes/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Wistar , Núcleos do Trigêmeo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...