Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurophysiol ; 127(1): 116-129, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34817286

RESUMO

Diverse physiological phenotypes in a neuronal population can broaden the range of computational capabilities within a brain region. The avian cochlear nucleus angularis (NA) contains a heterogeneous population of neurons whose variation in intrinsic properties results in electrophysiological phenotypes with a range of sensitivities to temporally modulated input. The low-threshold potassium conductance (GKLT) is a key feature of neurons involved in fine temporal structure coding for sound localization, but a role for these channels in intensity or spectrotemporal coding has not been established. To determine whether GKLT affects the phenotypical variation and temporal properties of NA neurons, we applied dendrotoxin-I (DTX), a potent antagonist of Kv1-type potassium channels, to chick brain stem slices in vitro during whole cell patch-clamp recordings. We found a cell-type specific subset of NA neurons that was sensitive to DTX: single-spiking NA neurons were most profoundly affected, as well as a subset of tonic-firing neurons. Both tonic I (phasic onset bursting) and tonic II (delayed firing) neurons showed DTX sensitivity in their firing rate and phenotypical firing pattern. Tonic III neurons were unaffected. Spike time reliability and fluctuation sensitivity measured in DTX-sensitive NA neurons was also reduced with DTX. Finally, DTX reduced spike threshold adaptation in these neurons, suggesting that GKLT contributes to the temporal properties that allow coding of rapid changes in the inputs to NA neurons. These results suggest that variation in Kv1 channel expression may be a key factor in functional diversity in the avian cochlear nucleus.NEW & NOTEWORTHY The dendrotoxin-sensitive voltage-gated potassium conductance typically associated with neuronal coincidence detection in the timing pathway for sound localization is demonstrated to affect spiking patterns and temporal input sensitivity in the intensity pathway in the avian auditory brain stem. The Kv1-family channels appear to be present in a subset of cochlear nucleus angularis neurons, regulate spike threshold dynamics underlying high-pass membrane filtering, and contribute to intrinsic firing diversity.


Assuntos
Potenciais de Ação/fisiologia , Núcleo Coclear/fisiologia , Neurônios/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Superfamília Shaker de Canais de Potássio/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Galinhas , Núcleo Coclear/efeitos dos fármacos , Núcleo Coclear/metabolismo , Venenos Elapídicos/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos
2.
J Gen Physiol ; 150(8): 1215-1230, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-30002162

RESUMO

Voltage-gated ion channels are key molecules for the generation of cellular electrical excitability. Many pharmaceutical drugs target these channels by blocking their ion-conducting pore, but in many cases, channel-opening compounds would be more beneficial. Here, to search for new channel-opening compounds, we screen 18,000 compounds with high-throughput patch-clamp technology and find several potassium-channel openers that share a distinct biaryl-sulfonamide motif. Our data suggest that the negatively charged variants of these compounds bind to the top of the voltage-sensor domain, between transmembrane segments 3 and 4, to open the channel. Although we show here that biaryl-sulfonamide compounds open a potassium channel, they have also been reported to block sodium and calcium channels. However, because they inactivate voltage-gated sodium channels by promoting activation of one voltage sensor, we suggest that, despite different effects on the channel gates, the biaryl-sulfonamide motif is a general ion-channel activator motif. Because these compounds block action potential-generating sodium and calcium channels and open an action potential-dampening potassium channel, they should have a high propensity to reduce excitability. This opens up the possibility to build new excitability-reducing pharmaceutical drugs from the biaryl-sulfonamide scaffold.


Assuntos
Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Sulfonamidas/farmacologia , Animais , Células CHO , Cricetulus , Ensaios de Triagem em Larga Escala , Cinética , Bibliotecas de Moléculas Pequenas
3.
J Neurosci ; 33(41): 16310-22, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-24107962

RESUMO

The molecular targets and neural circuits that underlie general anesthesia are not fully elucidated. Here, we directly demonstrate that Kv1-family (Shaker-related) delayed rectifier K(+) channels in the central medial thalamic nucleus (CMT) are important targets for volatile anesthetics. The modulation of Kv1 channels by volatiles is network specific as microinfusion of ShK, a potent inhibitor of Kv1.1, Kv1.3, and Kv1.6 channels, into the CMT awakened sevoflurane-anesthetized rodents. In heterologous expression systems, sevoflurane, isoflurane, and desflurane at subsurgical concentrations potentiated delayed rectifier Kv1 channels at low depolarizing potentials. In mouse thalamic brain slices, sevoflurane inhibited firing frequency and delayed the onset of action potentials in CMT neurons, and ShK-186, a Kv1.3-selective inhibitor, prevented these effects. Our findings demonstrate the exquisite sensitivity of delayed rectifier Kv1 channels to modulation by volatile anesthetics and highlight an arousal suppressing role of Kv1 channels in CMT neurons during the process of anesthesia.


Assuntos
Anestésicos Gerais/farmacologia , Nível de Alerta/efeitos dos fármacos , Núcleos Intralaminares do Tálamo/efeitos dos fármacos , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Desflurano , Núcleos Intralaminares do Tálamo/metabolismo , Isoflurano/análogos & derivados , Isoflurano/farmacologia , Espectroscopia de Ressonância Magnética , Masculino , Éteres Metílicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Sevoflurano , Superfamília Shaker de Canais de Potássio/metabolismo , Compostos Orgânicos Voláteis/farmacologia
4.
J Gen Physiol ; 141(2): 203-16, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23359283

RESUMO

Voltage-activated ion channels open and close in response to changes in membrane voltage, a property that is fundamental to the roles of these channels in electrical signaling. Protein toxins from venomous organisms commonly target the S1-S4 voltage-sensing domains in these channels and modify their gating properties. Studies on the interaction of hanatoxin with the Kv2.1 channel show that this tarantula toxin interacts with the S1-S4 domain and inhibits opening by stabilizing a closed state. Here we investigated the interaction of hanatoxin with the Shaker Kv channel, a voltage-activated channel that has been extensively studied with biophysical approaches. In contrast to what is observed in the Kv2.1 channel, we find that hanatoxin shifts the conductance-voltage relation to negative voltages, making it easier to open the channel with membrane depolarization. Although these actions of the toxin are subtle in the wild-type channel, strengthening the toxin-channel interaction with mutations in the S3b helix of the S1-S4 domain enhances toxin affinity and causes large shifts in the conductance-voltage relationship. Using a range of previously characterized mutants of the Shaker Kv channel, we find that hanatoxin stabilizes an activated conformation of the voltage sensors, in addition to promoting opening through an effect on the final opening transition. Chimeras in which S3b-S4 paddle motifs are transferred between Kv2.1 and Shaker Kv channels, as well as experiments with the related tarantula toxin GxTx-1E, lead us to conclude that the actions of tarantula toxins are not simply a product of where they bind to the channel, but that fine structural details of the toxin-channel interface determine whether a toxin is an inhibitor or opener.


Assuntos
Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Oócitos/fisiologia , Peptídeos/farmacologia , Superfamília Shaker de Canais de Potássio/fisiologia , Animais , Células Cultivadas , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Xenopus laevis
6.
J Biol Chem ; 284(23): 15659-67, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19269964

RESUMO

Hyperpolarization-activated cyclic nucleotide-modulated (HCN) channels resemble Shaker K+ channels in structure and function. In both, changes in membrane voltage produce directionally similar movement of positively charged residues in the voltage sensor to alter the pore structure at the intracellular side and gate ion flow. However, HCNs open when hyperpolarized, whereas Shaker opens when depolarized. Thus, electromechanical coupling between the voltage sensor and gate is opposite. A key determinant of this coupling is the intrinsic stability of the pore. In Shaker, an alanine/valine scan of residues across the pore, by single point mutation, showed that most mutations made the channel easier to open and steepened the response of the channel to changes in voltage. Because most mutations likely destabilize protein packing, the Shaker pore is most stable when closed, and the voltage sensor works to open it. In HCN channels, the pore energetics and vector of work by the voltage sensor are unknown. Accordingly, we performed a 22-residue alanine/valine scan of the distal pore of the HCN2 isoform and show that the effects of mutations on channel opening and on the steepness of the response of the channel to voltage are mixed and smaller than those in Shaker. These data imply that the stabilities of the open and closed pore are similar, the voltage sensor must apply force to close the pore, and the interactions between the pore and voltage sensor are weak. Moreover, cAMP binding to the channel heightens the effects of the mutations, indicating stronger interactions between the pore and voltage sensor, and tips the energetic balance toward a more stable open state.


Assuntos
AMP Cíclico/fisiologia , Canais Iônicos/genética , Canais Iônicos/fisiologia , Alanina , Substituição de Aminoácidos , Animais , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Cricetinae , Cricetulus , AMP Cíclico/farmacologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/química , Canais Iônicos/efeitos dos fármacos , Camundongos , Técnicas de Patch-Clamp , Canais de Potássio , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Superfamília Shaker de Canais de Potássio/fisiologia , Termodinâmica , Valina
7.
J Neurosci ; 28(51): 13716-26, 2008 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-19091962

RESUMO

Separation of the cortical sheet into functionally distinct regions is a hallmark of neocortical organization. Cortical circuit function emerges from afferent and efferent connectivity, local connectivity within the cortical microcircuit, and the intrinsic membrane properties of neurons that comprise the circuit. While localization of functions to particular cortical areas can be partially accounted for by regional differences in both long range and local connectivity, it is unknown whether the intrinsic membrane properties of cortical cell types differ between cortical regions. Here we report the first example of a region-specific firing type in layer 5 pyramidal neurons, and show that the intrinsic membrane and integrative properties of a discrete subtype of layer 5 pyramidal neurons differ between primary motor and somatosensory cortices due to region- and cell-type-specific Kv1 subunit expression.


Assuntos
Potenciais de Ação/fisiologia , Córtex Motor/fisiologia , Células Piramidais/fisiologia , Tratos Piramidais/fisiologia , Superfamília Shaker de Canais de Potássio/metabolismo , Córtex Somatossensorial/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Venenos Elapídicos/farmacologia , Estimulação Elétrica , Genes Reporter , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Transgênicos , Córtex Motor/citologia , Córtex Motor/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Células Piramidais/efeitos dos fármacos , Tratos Piramidais/citologia , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Córtex Somatossensorial/citologia , Córtex Somatossensorial/efeitos dos fármacos , Fatores de Tempo
9.
Nat Chem Biol ; 4(11): 708-14, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18806782

RESUMO

The Shaker family voltage-dependent potassium channels (Kv1) are expressed in a wide variety of cells and are essential for cellular excitability. In humans, loss-of-function mutations of Kv1 channels lead to hyperexcitability and are directly linked to episodic ataxia and atrial fibrillation. All Kv1 channels assemble with beta subunits (Kv betas), and certain Kv betas, for example Kv beta 1, have an N-terminal segment that closes the channel by the N-type inactivation mechanism. In principle, dissociation of Kv beta 1, although never reported, should eliminate inactivation and thus potentiate Kv1 current. We found that cortisone increases rat Kv1 channel activity by binding to Kv beta 1. A crystal structure of the Kv beta-cortisone complex was solved to 1.82-A resolution and revealed novel cortisone binding sites. Further studies demonstrated that cortisone promotes dissociation of Kv beta. The new mode of channel modulation may be explored by native or synthetic ligands to fine-tune cellular excitability.


Assuntos
Cortisona/farmacologia , Canal de Potássio Kv1.2/metabolismo , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Ligação Competitiva/efeitos dos fármacos , Cortisona/química , Cristalografia por Raios X , Humanos , Canal de Potássio Kv1.2/química , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Ligação Proteica/efeitos dos fármacos , Ratos , Superfamília Shaker de Canais de Potássio/metabolismo , Xenopus
10.
Eur J Pharmacol ; 581(1-2): 138-47, 2008 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-18155692

RESUMO

The aim of this study was to assess the role of K(+) and Ca(2+) fluxes in the cerebroarterial vasoactive effects of the phosphodiesterase-5 inhibitor sildenafil. We used isolated rabbit basilar arteries to assess the effects of extracellular K(+) raising on sildenafil-induced vasodilatation, and studied the pharmacological interaction of sildenafil with selective modulators of membrane K(+) and Ca(2+) channels. Expression of Kv1 subunits of K(+) channels was assessed at messenger and protein levels. Parallel experiments were carried out with zaprinast for comparison. Sildenafil (10 nM-0.1 mM) induced concentration-dependent relaxation of endothelin-1 (10 nM)-precontracted arteries, which was partially inhibited by depolarization with KCl (50 mM), 3 mM tetraethylammonium (non-selective K(+) channel blocker) or 1 mM aminopyridine (inhibitor of K(v) channels), but not by 1 microM glibenclamide (inhibitor of K(ATP) channels) or 50 nM iberiotoxin (inhibitor of K(Ca) channels). Arterial smooth muscle expressed messengers for Kv1.2, Kv1.3, Kv1.4, Kv1.5 and Kv1.6, and proteins of Kv1.1, Kv1.2 and Kv1.4. CaCl(2) (10 microM- 10 mM) induced concentration-dependent contraction in Ca(2+)-free, depolarizing (50 mM KCl) medium. Sildenafil (0.1-100 microM) produced reversible concentration-dependent inhibition of the response to CaCl(2), which was completely abolished by the highest sildenafil concentration. By contrast, only 100 microM zaprinast inhibited the response to CaCl(2). The L-type Ca(2+) channel activator Bay K 8644 (0.1 nM-1 microM) induced concentration-dependent potentiation of the response to CaCl(2) inhibited by 100 microM sildenafil. Moreover, Bay K 8644 (0.1 nM-1 microM) induced concentration-dependent contraction in slightly depolarizing (15 mM) medium, which was inhibited to the same extent and in a concentration-dependent way by sildenafil (0.1-100 microM) and zaprinast (1 or 100 microM). These results show that sildenafil relaxes the rabbit basilar artery by increasing K(+) efflux through K(v) channels, which in turn may affect Ca(2+) signalling. Expression of Kv1 subunits involved in this pharmacological effect occurs at the messenger and, in some cases, at the protein level. In addition to this phosphodiesterase-5-related effect, sildenafil and zaprinast inhibit cerebroarterial vasoconstriction at least in part by directly blocking L-type Ca(2+) channels, although a decrease in the sensitivity of the contractile apparatus to Ca(2+) can not be discarded.


Assuntos
Artéria Basilar/efeitos dos fármacos , Cálcio/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Piperazinas/farmacologia , Potássio/metabolismo , Sulfonas/farmacologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Artéria Basilar/fisiologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Relação Dose-Resposta a Droga , Técnicas In Vitro , Masculino , Purinas/farmacologia , RNA Mensageiro/análise , Coelhos , Superfamília Shaker de Canais de Potássio/análise , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Superfamília Shaker de Canais de Potássio/genética , Citrato de Sildenafila
11.
Pflugers Arch ; 456(2): 393-405, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18043943

RESUMO

N-linked glycans, including sialic acids, are integral components of ion channel complexes. To determine if N-linked sugars can modulate a rapidly inactivating K+ channel, the glycosylated Drosophila melanogaster Shaker K+ channel (ShB) and the N-glycosylation-deficient mutant (ShNQ), were studied under conditions of full and reduced sialylation. Through an apparent electrostatic mechanism, full sialylation induced uniform and significant hyperpolarizing shifts in all measured voltage-dependent ShB gating parameters compared to those measured under conditions of reduced sialylation. Steady-state gating of ShNQ was unaffected by changes in sialylation and was nearly identical to that observed for ShB under conditions of reduced sialylation, indicating that N-linked sialic acids were wholly responsible for the observed effects of sialic acid on ShB gating. Interestingly, the rates of transition among channel states and the voltage-independent rates of activation and inactivation were significantly slower for ShNQ compared to ShB. Both effects were independent of sialylation, indicating that N-linked sugars other than sialic acids alter ShB gating kinetics but have little to no effect on the steady-state distribution of channels among states. The effect of sialic acids on channel gating, particularly inactivation gating, and the impact of other N-linked sugars on channel gating kinetics are unique to the ShB isoform. Thus, ShB gating is modulated by two complementary but distinct sugar-dependent mechanisms, (1) an N-linked sialic acid-dependent surface charge effect and (2) a sialic acid-independent effect that is consistent with N-linked sugars affecting the stability of ShB among its functional states.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Ácido N-Acetilneuramínico/farmacologia , Superfamília Shaker de Canais de Potássio/fisiologia , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Drosophila melanogaster , Eletrofisiologia , Feminino , Glicosilação , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Superfamília Shaker de Canais de Potássio/genética , Eletricidade Estática , Transfecção
12.
Biochem Pharmacol ; 74(1): 74-85, 2007 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-17499219

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) contribute to gastrointestinal ulcer formation by inhibiting epithelial cell migration and mucosal restitution; however, the drug-affected signaling pathways are poorly defined. We investigated whether NSAID inhibition of intestinal epithelial migration is associated with depletion of intracellular polyamines, depolarization of membrane potential (E(m)) and altered surface expression of K(+) channels. Epithelial cell migration in response to the wounding of confluent IEC-6 and IEC-Cdx2 monolayers was reduced by indomethacin (100 microM), phenylbutazone (100 microM) and NS-398 (100 microM) but not by SC-560 (1 microM). NSAID-inhibition of intestinal cell migration was not associated with depletion of intracellular polyamines. Treatment of IEC-6 and IEC-Cdx2 cells with indomethacin, phenylbutazone and NS-398 induced significant depolarization of E(m), whereas treatment with SC-560 had no effect on E(m). The E(m) of IEC-Cdx2 cells was: -38.5+/-1.8 mV under control conditions; -35.9+/-1.6 mV after treatment with SC-560; -18.8+/-1.2 mV after treatment with indomethacin; and -23.7+/-1.4 mV after treatment with NS-398. Whereas SC-560 had no significant effects on the total cellular expression of K(v)1.4 channel protein, indomethacin and NS-398 decreased not only the total cellular expression of K(v)1.4, but also the cell surface expression of both K(v)1.4 and K(v)1.6 channel subunits in IEC-Cdx2. Both K(v)1.4 and K(v)1.6 channel proteins were immunoprecipitated by K(v)1.4 antibody from IEC-Cdx2 lysates, indicating that these subunits co-assemble to form heteromeric K(v) channels. These results suggest that NSAID inhibition of epithelial cell migration is independent of polyamine-depletion, and is associated with depolarization of E(m) and decreased surface expression of heteromeric K(v)1 channels.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Mucosa Intestinal/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Cromatografia Líquida de Alta Pressão , Humanos , Indometacina/toxicidade , Mucosa Intestinal/metabolismo , Nitrobenzenos/toxicidade , Técnicas de Patch-Clamp , Fenilbutazona/toxicidade , Poliaminas/análise , Poliaminas/metabolismo , Superfamília Shaker de Canais de Potássio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Espectrometria de Massas por Ionização por Electrospray , Sulfonamidas/toxicidade , Cicatrização/fisiologia
13.
FEBS Lett ; 581(13): 2478-84, 2007 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-17490656

RESUMO

We have purified BoiTx1, the first toxin from the venom of the Israeli scorpion, Buthus occitanus israelis, and studied its activity and genomic organization. BoiTx1 is a 37 amino acid-long peptide contained six conserved cysteines, and is classified as an alpha-KTx3.10 toxin. The pharmacological effects of BoiTx1 were studied on various cloned K(+) channels expressed in Xenopus laevis oocytes. BoiTx1 inhibited currents through Drosophila Shaker channels with an IC(50) value of 3.5+/-0.5nM, yet had much lesser effect on its mammalian orthologs. Thus, BoiTx1 is the first member of the alpha-KTx3 family that preferentially affects insect potassium channels.


Assuntos
Venenos de Escorpião/química , Venenos de Escorpião/farmacologia , Superfamília Shaker de Canais de Potássio/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sequência Conservada , DNA/genética , DNA/isolamento & purificação , Eletrofisiologia , Dados de Sequência Molecular , Venenos de Escorpião/genética , Venenos de Escorpião/isolamento & purificação , Escorpiões/genética , Homologia de Sequência de Aminoácidos , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
Artigo em Inglês | MEDLINE | ID: mdl-17270501

RESUMO

The venom from the Brazilian scorpion Tityus stigmurus was fractionated by high performance liquid chromatography (HPLC) and the corresponding components were used for molecular mass determination using electrospray ion trap mass spectrometry. One hundred distinct components were clearly assigned showing molecular masses from 216.5 to 44,800.0 Da. Fifteen new components were isolated and sequenced, four of them to completion: Tst-3 (similar to Na(+) channel specific scorpion toxins), Tst-17 (a K(+) channel blocking peptide similar to Tc1), Tst beta KTx (a peptide with identical sequence as that of TsTX-K beta toxin earlier described to exist in T. serrulatus venom) and finally a novel proline-rich peptide of unknown function. Among the eleven components partially sequenced were two enzymes: hyaluronidase and lysozyme. The first enzyme has a molecular mass of 44,800.0 Da. This enzyme showed high activity against the substrate hyaluronan in vitro. Amino acid sequence of the second enzyme showed that it is similar to other known lysozymes, with similar molecular mass and sequence to that of bona fide lysozymes reported in public protein data banks. Finally, this communication reports a correlation among HPLC retention times and molecular masses of folded scorpion toxins as well as a comparative structural and physiological analysis of components from the venom of several species of the genus Tityus.


Assuntos
Proteínas de Insetos/química , Bloqueadores dos Canais de Potássio/química , Proteômica , Venenos de Escorpião/química , Escorpiões , Sequência de Aminoácidos , Animais , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Eletrofisiologia , Hialuronoglucosaminidase/análise , Proteínas de Insetos/farmacologia , Dados de Sequência Molecular , Peso Molecular , Muramidase/análise , Técnicas de Patch-Clamp , Mapeamento de Peptídeos , Bloqueadores dos Canais de Potássio/farmacologia , Venenos de Escorpião/farmacologia , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Superfamília Shaker de Canais de Potássio/metabolismo , Especificidade da Espécie , Espectrometria de Massas por Ionização por Electrospray , Spodoptera/citologia , Spodoptera/efeitos dos fármacos
15.
J Physiol ; 581(Pt 1): 175-87, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17317746

RESUMO

In CA1 pyramidal neurons, burst firing is correlated with hippocampally dependent behaviours and modulation of synaptic strength. One of the mechanisms underlying burst firing in these cells is the afterdepolarization (ADP) that follows each action potential. Previous work has shown that the ADP results from the interaction of several depolarizing and hyperpolarizing conductances located in the soma and the dendrites. By using patch-clamp recordings from acute rat hippocampal slices we show that D-type potassium current modulates the size of the ADP and the bursting of CA1 pyramidal neurons. Sensitivity to alpha-dendrotoxin suggests that Kv1-containing potassium channels mediate this current. Dual somato-dendritic recording, outside-out dendritic recordings, and focal application of dendrotoxin together indicate that the channels mediating this current are located in the apical dendrites. Thus, our data present evidence for a dendritic segregation of Kv1-like channels in CA1 pyramidal neurons and identify a novel action for these channels, showing that they inhibit action potential bursting by restricting the size of the ADP.


Assuntos
Potenciais de Ação/fisiologia , Células Dendríticas/fisiologia , Hipocampo/fisiologia , Células Piramidais/fisiologia , Superfamília Shaker de Canais de Potássio/fisiologia , Animais , Células Dendríticas/citologia , Venenos Elapídicos/farmacologia , Potenciais Evocados/fisiologia , Hipocampo/citologia , Masculino , Técnicas de Patch-Clamp , Células Piramidais/citologia , Ratos , Ratos Wistar , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos
16.
Physiol Res ; 56(6): 807-813, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17087603

RESUMO

To understand the contribution of potassium (K+) channels, particularly alpha-dendrotoxin (D-type)-sensitive K+ channels (Kv.1, Kv1.2 or Kv1.6 subunits), to the generation of neuronal spike output we must have detailed information of the functional role of these channels in the neuronal membrane. Conventional intracellular recording methods in current clamp mode were used to identify the role of alpha-dendrotoxin (alpha-DTX)-sensitive K+ channel currents in shaping the spike output and modulation of neuronal properties of cerebellar Purkinje neurons (PCs) in slices. Addition of alpha-DTX revealed that D-type K+ channels play an important role in the shaping of Purkinje neuronal firing behavior. Repetitive firing capability of PCs was increased following exposure to artificial cerebrospinal fluid (aCSF) containing alpha-DTX, so that in response to the injection of 0.6 nA depolarizing current pulse of 600 ms, the number of action potentials insignificantly increased from 15 in the presence of 4-AP to 29 action potentials per second after application of DTX following pretreatment with 4-AP. These results indicate that D-type K+ channels (Kv.1, Kv1.2 or Kv1.6 subunits) may contribute to the spike frequency adaptation in PCs. Our findings suggest that the activation of voltage-dependent K+ channels (D and A types) markedly affect the firing pattern of PCs.


Assuntos
Venenos Elapídicos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Células de Purkinje/fisiologia , Superfamília Shaker de Canais de Potássio/fisiologia , 4-Aminopiridina/farmacologia , Potenciais de Ação/fisiologia , Animais , Eletrofisiologia , Canal de Potássio Kv1.2/efeitos dos fármacos , Canal de Potássio Kv1.2/fisiologia , Canal de Potássio Kv1.6/efeitos dos fármacos , Canal de Potássio Kv1.6/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Ratos , Ratos Sprague-Dawley , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos
17.
J Pharmacol Exp Ther ; 320(1): 162-72, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17015639

RESUMO

The effect of 4-aminopyridine (4-AP) on Kv channel activation has been extensively investigated, but its interaction with inactivation is less well understood. Voltage-clamp fluorimetry was used to directly monitor the action of 4-AP on conformational changes associated with slow inactivation of Shaker channels. Tetramethylrhodamine-5-maleimide was used to fluorescently label substituted cysteine residues in the S3-S4 linker (A359C) and pore (S424C). Activation- and inactivation-induced changes in fluorophore microenvironment produced fast and slow phases of fluorescence that were modified by 4-AP. In Shaker A359C, 4-AP block reduced the slow-phase contribution from 61 +/- 3 to 28 +/- 5%, suggesting that binding inhibits the conformational changes associated with slow inactivation and increased the fast phase that reports channel activation from 39 +/- 3 to 72 +/- 5%. In addition, 4-AP enhanced both fast and slow phases of fluorescence return upon repolarization (tau reduced from 87 +/- 15 to 40 +/- 1 ms and from 739 +/- 83 to 291 +/- 21 ms, respectively), suggesting that deactivation and recovery from inactivation were enhanced. In addition, the effect of 4-AP on the slow phase of fluorescence was dramatically reduced in channels with either reduced (T449V) or permanent P-type (W434F) inactivation. Interestingly, the slow phase of fluorescence return of W434F channels was enhanced by 4-AP, suggesting that 4-AP prevents the transition to C-type inactivation in these channels. These data directly demonstrate that 4-AP prevents slow inactivation of Kv channels and that 4-AP can bind to P-type-inactivated channels and selectively inhibit the onset of C-type inactivation.


Assuntos
4-Aminopiridina/farmacologia , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Animais , Feminino , Fluorescência , Rodaminas , Superfamília Shaker de Canais de Potássio/química , Superfamília Shaker de Canais de Potássio/classificação , Xenopus laevis
18.
J Gen Physiol ; 128(6): 687-99, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17101817

RESUMO

In nerve and muscle cells, the voltage-gated opening and closing of cation-selective ion channels is accompanied by the translocation of 12-14 elementary charges across the membrane's electric field. Although most of these charges are carried by residues in the S4 helix of the gating module of these channels, the precise nature of their physical movement is currently the topic of spirited debate. Broadly speaking, two classes of models have emerged: those that suggest that small-scale motions can account for the extensive charge displacement, and those that invoke a much larger physical movement. In the most recent incarnation of the latter type of model, which is based on structural and functional data from the archaebacterial K(+) channel KvAP, a "voltage-sensor paddle" comprising a helix-turn-helix of S3-S4 translocates approximately 20 A through the bilayer during the gating cycle (Jiang, Y., A. Lee, J. Chen, V. Ruta, M. Cadene, B.T. Chait, and R. MacKinnon. 2003. Nature. 423:33-41; Jiang, Y., V. Ruta, J. Chen, A. Lee, and R. MacKinnon. 2003. Nature. 423:42-48.; Ruta, V., J. Chen, and R. MacKinnon. 2005. Cell. 123:463-475). We used two methods to test for analogous motions in the Shaker K(+) channel, each examining the aqueous exposure of residues near S3. In the first, we employed a pore-blocking maleimide reagent (Blaustein, R.O., P.A. Cole, C. Williams, and C. Miller. 2000. Nat. Struct. Biol. 7:309-311) to probe for state-dependent changes in the chemical reactivity of substituted cysteines; in the second, we tested the state-dependent accessibility of a tethered biotin to external streptavidin (Qiu, X.Q., K.S. Jakes, A. Finkelstein, and S.L. Slatin. 1994. J. Biol. Chem. 269:7483-7488; Slatin, S.L., X.Q. Qiu, K.S. Jakes, and A. Finkelstein. 1994. Nature. 371:158-161). In both types of experiments, residues predicted to lie near the top of S3 did not exhibit any change in aqueous exposure during the gating cycle. This lack of state dependence argues against large-scale movements, either axially or radially, of Shaker's S3-S4 voltage-sensor paddle.


Assuntos
Ativação do Canal Iônico/fisiologia , Superfamília Shaker de Canais de Potássio/fisiologia , Sequência de Aminoácidos , Animais , Biotinilação , Cisteína/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Dados de Sequência Molecular , Movimento , Oócitos , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Estreptavidina/farmacologia , Xenopus
19.
J Gen Physiol ; 128(6): 649-57, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17130518

RESUMO

Open-channel blockers such as tetraethylammonium (TEA) have a long history as probes of the permeation pathway of ion channels. High affinity blockade by extracellular TEA requires the presence of an aromatic amino acid at a position that sits at the external entrance of the permeation pathway (residue 449 in the eukaryotic voltage-gated potassium channel Shaker). We investigated whether a cation-pi interaction between TEA and such an aromatic residue contributes to TEA block using the in vivo nonsense suppression method to incorporate a series of increasingly fluorinated Phe side chains at position 449. Fluorination, which is known to decrease the cation-pi binding ability of an aromatic ring, progressively increased the inhibitory constant K(i) for the TEA block of Shaker. A larger increase in K(i) was observed when the benzene ring of Phe449 was substituted by nonaromatic cyclohexane. These results support a strong cation-pi component to the TEA block. The data provide an empirical basis for choosing between Shaker models that are based on two classes of reported crystal structures for the bacterial channel KcsA, showing residue Tyr82 in orientations either compatible or incompatible with a cation-pi mechanism. We propose that the aromatic residue at this position in Shaker is favorably oriented for a cation-pi interaction with the permeation pathway. This choice is supported by high level ab initio calculations of the predicted effects of Phe modifications on TEA binding energy.


Assuntos
Bloqueadores dos Canais de Potássio/farmacologia , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Tetraetilamônio/farmacologia , Sequência de Aminoácidos , Proteínas de Bactérias/efeitos dos fármacos , Simulação por Computador , Eletrofisiologia , Fenilalanina/química , Canais de Potássio/efeitos dos fármacos , Conformação Proteica , Termodinâmica
20.
J Gen Physiol ; 127(5): 481-94, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16606688

RESUMO

Cyclic nucleotide-gated (CNG) channels play important roles in the transduction of visual and olfactory information by sensing changes in the intracellular concentration of cyclic nucleotides. We have investigated the interactions between intracellularly applied quaternary ammonium (QA) ions and the alpha subunit of rod cyclic nucleotide-gated channels. We have used a family of alkyl-triethylammonium derivatives in which the length of one chain is altered. These QA derivatives blocked the permeation pathway of CNG channels in a concentration- and voltage-dependent manner. For QA compounds with tails longer than six methylene groups, increasing the length of the chain resulted in higher apparent affinities of approximately 1.2 RT per methylene group added, which is consistent with the presence of a hydrophobic pocket within the intracellular mouth of the channel that serves as part of the receptor binding site. At the single channel level, decyltriethyl ammonium (C10-TEA) ions did not change the unitary conductance but they did reduce the apparent mean open time, suggesting that the blocker binds to open channels. We provide four lines of evidence suggesting that QA ions can also bind to closed channels: (1) the extent of C10-TEA blockade at subsaturating [cGMP] was larger than at saturating agonist concentration, (2) under saturating concentrations of cGMP, cIMP, or cAMP, blockade levels were inversely correlated with the maximal probability of opening achieved by each agonist, (3) in the closed state, MTS reagents of comparable sizes to QA ions were able to modify V391C in the inner vestibule of the channel, and (4) in the closed state, C10-TEA was able to slow the Cd2+ inhibition observed in V391C channels. These results are in stark contrast to the well-established QA blockade mechanism in Kv channels, where these compounds can only access the inner vestibule in the open state because the gate that opens and closes the channel is located cytoplasmically with respect to the binding site of QA ions. Therefore, in the context of Kv channels, our observations suggest that the regions involved in opening and closing the permeation pathways in these two types of channels are different.


Assuntos
Canais Iônicos/química , Canais Iônicos/efeitos dos fármacos , Compostos de Amônio Quaternário/farmacologia , Animais , Sítios de Ligação , GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Relação Dose-Resposta a Droga , Eletrofisiologia , Feminino , Canais Iônicos/metabolismo , Canais Iônicos/fisiologia , Potenciais da Membrana/fisiologia , Oócitos , Permeabilidade/efeitos dos fármacos , Compostos de Amônio Quaternário/metabolismo , Superfamília Shaker de Canais de Potássio/química , Superfamília Shaker de Canais de Potássio/efeitos dos fármacos , Superfamília Shaker de Canais de Potássio/metabolismo , Superfamília Shaker de Canais de Potássio/fisiologia , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...