Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Pharmacol Rev ; 69(1): 1-11, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28267675

RESUMO

A subset of potassium channels is regulated primarily by changes in the cytoplasmic concentration of ions, including calcium, sodium, chloride, and protons. The eight members of this subfamily were originally all designated as calcium-activated channels. More recent studies have clarified the gating mechanisms for these channels and have documented that not all members are sensitive to calcium. This article describes the molecular relationships between these channels and provides an introduction to their functional properties. It also introduces a new nomenclature that differentiates between calcium- and sodium-activated potassium channels.


Assuntos
Cálcio/metabolismo , Cloretos/metabolismo , Ativação do Canal Iônico , Canais de Potássio Cálcio-Ativados/classificação , Canais de Potássio Cálcio-Ativados/metabolismo , Canais de Potássio/classificação , Canais de Potássio/metabolismo , Sódio/metabolismo , Terminologia como Assunto , Animais , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/classificação , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/classificação , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Masculino , Espermatozoides/metabolismo
2.
Cell Physiol Biochem ; 46(3): 1112-1121, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29669325

RESUMO

BACKGROUND/AIMS: The replacement of the amino acid valine at position 388 (Shaker position 438) in hKv1.3 channels or at the homologue position 370 in hKv1.2 channels resulted in a channel with two different ion conducting pathways: One pathway was the central, potassium-selective α-pore, that was sensitive to block by peptide toxins (CTX or KTX in the hKv1.3_V388C channel and CTX or MTX in the hKv1.2_V370C channel). The other pathway (σ-pore) was behind the central α-pore creating an inward current at potentials more negative than -100 mV, a potential range where the central α-pore was closed. In addition, current through the σ-pore could not be reduced by CTX, KTX or MTX in the hKv1.3_V388C or the hKv1.2_V370C channel, respectively. METHODS: For a more detailed characterization of the σ-pore, we created a trimer consisting of three hKv1.3_V388C α-subunits linked together and characterized current through this trimeric hKv1.3_V388C channel. Additionally, we determined which amino acids line the σ-pore in the tetrameric hKv1.3_V388C channel by replacing single amino acids in the tetrameric hKv1.3_V388C mutant channel that could be involved in σ-pore formation. RESULTS: Overexpression of the trimeric hKv1.3_V388C channel in COS-7 cells yielded typical σ-pore currents at potentials more negative than -100 mV similar to what was observed for the tetrameric hKv1.3_V388C channel. Electrophysiological properties of the trimeric and tetrameric channel were similar: currents could be observed at potentials more negative than -100 mV, were not carried by protons or chloride ions, and could not be reduced by peptide toxins (CTX, MTX) or TEA. The σ-pore was mostly permeable to Na+ and Li+. In addition, in our site-directed mutagenesis experiments, we created a number of new double mutant channels in the tetrameric hKv1.3_V388C background channel. Two of these tetrameric double mutant channels (hKv1.3_V388C_T392Y and hKv1.3_V388C_Y395W) did not show currents through the σ-pore. CONCLUSIONS: From our experiments with the trimeric hKv1.3_V388C channel we conclude that the σ-pore exists in hKv1.3_V388C channels independently of the α-pore. From our site-directed mutagenesis experiments in the tetrameric hKv1.3_V388C channel we conclude that amino acid position 392 and 395 (Shaker position 442 and 445) line the σ-pore.


Assuntos
Canal de Potássio Kv1.3/metabolismo , Animais , Células COS , Charibdotoxina/toxicidade , Chlorocebus aethiops , Clonagem Molecular , Humanos , Canal de Potássio Kv1.3/genética , Potenciais da Membrana/efeitos dos fármacos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Estrutura Quaternária de Proteína
3.
Cell Physiol Biochem ; 44(1): 172-184, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29131061

RESUMO

BACKGROUND/AIMS: The human-voltage gated Kv1.3 channel (hKv1.3) is expressed in T- and B lymphocytes. Verapamil is able to block hKv1.3 channels. We characterized the effect of verapamil on currents through hKv1.3 channels paying special attention to the on-rate (kon) of verapamil. By comparing on-rates obtained in wild-type (wt) and mutant channels a binding pocket for verapamil and impacts of different amino acid residues should be investigated. METHODS: Using the whole-cell patch clamp technique the action of verapamil on currents through wild-type and six hKv1.3 mutant channels in the open state was investigated by measuring the time course of the open channel block in order to calculate kon of verapamil. RESULTS: The on-rate of verapamil to block current through hKv1.3_T419C mutant channels is similar to that obtained for hKv1.3_wt channels whereas the on-rate of verapamil to block currents through hKv1.3_L417C and hKv1.3_L418C mutant channels was ∼ 3 times slower compared to in wt channels. The on-rate of verapamil to block currents through hKv1.3_L346C and the double mutant hKv1.3_L346C_L418C channel was ∼ 2 times slower compared to that obtained in the wt channel. The hKv1.3_I420C mutant channel reduced the on-rate of verapamil to block currents ∼ 6 fold. CONCLUSIONS: We conclude that position 420 in hKv1.3 channels maximally interferes with verapamil reaching its binding site to block the channel. Positions 417 and 418 in hKv1.3 channels partially hinder verapamil reaching its binding site to block the channel whereas position 419 may not interfere with verapamil at all. Mutant hKv1.3_L346C and hKv1.3_L346C_L418C mutant channels might indirectly influence the ability of verapamil reaching its binding site to block current.


Assuntos
Canal de Potássio Kv1.3/metabolismo , Bloqueadores dos Canais de Potássio/metabolismo , Verapamil/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Chlorocebus aethiops , Humanos , Cinética , Canal de Potássio Kv1.3/genética , Potenciais da Membrana/efeitos dos fármacos , Simulação de Acoplamento Molecular , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Ligação Proteica , Estrutura Terciária de Proteína , Verapamil/química , Verapamil/farmacologia
4.
Cell Physiol Biochem ; 34(2): 474-90, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25096234

RESUMO

BACKGROUND/AIMS: Small-conductance calcium-activated (SK) channels play an important role by controlling the after-hyperpolarization of excitable cells. The level of expression and density of these channels is an essential factor for controlling different cellular functions. Several studies showed a co-localization of K(Ca)2.3 channels and Endophilin A3 in different tissues. Endophilin A3 belongs to a family of BAR- and SH3 domain containing proteins that bind to dynamin and are involved in the process of vesicle scission in clathrin-mediated endocytosis. METHODS: Using the yeast two-hybrid system and the GST pull down assay we demonstrated that Endophilin A3 interacts with the N-terminal part of K(Ca)2.3 channels. In addition, we studied the impact of this interaction on channel activity by patch clamp measurements in PC12 cells expressing endogenous K(Ca)2.3 channels. K(Ca)2.3 currents were activated by using pipette solutions containing 1 µM free Ca(2+). RESULTS: Whole-cell measurements of PC12 cells transfected with Endophilin A3 showed a reduction of KCa2.3 specific Cs(+) currents indicating that the interaction of Endophilin A3 with K(Ca)2.3 channels also occurs in mammalian cells and that this interaction has functional consequences for current flowing through K(Ca)2.3 channels. Since K(Ca)2.3 specific currents could be increased in PC12 cells transfected with Endophilin A3 with DC-EBIO (30 µM), a known SK-channel activator, these data also implicate that Endophilin A3 did not significantly remove K(Ca)2.3 channels from the membrane but changed the sensitivity of the channels to Ca(2+) which could be overcome by DC-EBIO. CONCLUSION: This interaction seems to be important for the function of K(Ca)2.3 channels and might therefore play a significant role in situations where channel activation is pivotal for cellular function.


Assuntos
Aciltransferases/metabolismo , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA , Dados de Sequência Molecular , Células PC12 , Reação em Cadeia da Polimerase , Canais de Potássio/química , Ratos , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido
5.
Br J Pharmacol ; 180 Suppl 2: S145-S222, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-38123150

RESUMO

The Concise Guide to PHARMACOLOGY 2023/24 is the sixth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of approximately 1800 drug targets, and over 6000 interactions with about 3900 ligands. There is an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (https://www.guidetopharmacology.org/), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes almost 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.16178. Ion channels are one of the six major pharmacological targets into which the Guide is divided, with the others being: G protein-coupled receptors, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2023, and supersedes data presented in the 2021/22, 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.


Assuntos
Bases de Dados de Produtos Farmacêuticos , Farmacologia , Humanos , Canais Iônicos/química , Ligantes , Receptores Acoplados a Proteínas G , Bases de Dados Factuais
6.
J Biol Chem ; 286(22): 20031-42, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21498510

RESUMO

Voltage-gated potassium channels are proteins composed of four subunits consisting of six membrane-spanning segments S1-S6, with S4 as the voltage sensor. The region between S5 and S6 forms the potassium-selective ion-conducting central α-pore. Recent studies showed that mutations in the voltage sensor of the Shaker channel could disclose another ion permeation pathway through the voltage-sensing domain (S1-S4) of the channel, the ω-pore. In our studies we used the voltage-gated hKv1.3 channel, and the insertion of a cysteine at position V388C (Shaker position 438) generated a current through the α-pore in high potassium outside and an inward current at hyperpolarizing potentials carried by different cations like Na(+), Li(+), Cs(+), and NH(4)(+). The observed inward current looked similar to the ω-current described for the R1C/S Shaker mutant channel and was not affected by some pore blockers like charybdotoxin and tetraethylammonium but was inhibited by a phenylalkylamine blocker (verapamil) that acts from the intracellular side. Therefore, we hypothesize that the hKv1.3_V388C mutation in the P-region generated a channel with two ion-conducting pathways. One, the α-pore allowing K(+) flux in the presence of K(+), and the second pathway, the σ-pore, functionally similar but physically distinct from the ω-pathway. The entry of this new pathway (σ-pore) is presumably located at the backside of Y395 (Shaker position 445), proceeds parallel to the α-pore in the S6-S6 interface gap, ending between S5 and S6 at the intracellular side of one α-subunit, and is blocked by verapamil.


Assuntos
Canal de Potássio Kv1.3/metabolismo , Substituição de Aminoácidos , Animais , Células COS , Bloqueadores dos Canais de Cálcio/farmacologia , Cátions Monovalentes , Chlorocebus aethiops , Humanos , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína , Verapamil/farmacologia
7.
Mol Pharmacol ; 79(4): 681-91, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21220411

RESUMO

hKv1.3 channels in lymphocytes are targets for the chemotherapy treatment of autoimmune diseases. Phenylalkylamines block Kv1.3 channels by poorly understood mechanisms. In the inactivation-reduced mutant H399T, the second mutation A413C in S6 substantially decreases the potency of phenylalkylamines with a para-methoxy group at the phenylethylamine end, whereas potency of phenylalkylamines lacking this group is less affected. Intriguingly, completely demethoxylated emopamil blocks mutant H399T/A413C with a 2:1 stoichiometry. Here, we generated a triple mutant, H399T/C412A/A413C, and found that its emopamil-binding properties are similar to those of the double mutant. These data rule out disulfide bonding Cys412-Cys413, which would substantially deform the inner helix, suggest a clash of Cys413 with the para-methoxy group, and provide a distance constraint to dock phenylalkylamines in a Kv1.2-based homology model. Monte Carlo minimizations predict that the verapamil ammonium group donates an H-bond to the backbone carbonyl of Thr391 at the P-loop turn, the pentanenitrilephenyl moiety occludes the pore, whereas the phenylethylamine meta- and para-methoxy substituents approach, respectively, the side chains of Met390 and Ala413. In the double-mutant model, the Cys413 side chains accept H-bonds from two emopamil molecules whose phenyl rings fit in the hydrophobic intersubunit interfaces, whereas the pentanenitrilephenyl moieties occlude the pore. Because these interfaces are unattractive for a methoxylated phenyl ring, the ammonium group of respective phenylalkylamines cannot approach the Cys413 side chain and binds at the focus of P-helices, whereas the para-methoxy group clashes with Cys413. Our study proposes an atomistic mechanism of Kv1.3 block by phenylalkylamines and highlights the intra- and intersubunit interfaces as ligand binding loci.


Assuntos
Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/química , Mutação , Bloqueadores dos Canais de Potássio/farmacologia , Verapamil/análogos & derivados , Verapamil/farmacologia , Alanina/genética , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Células COS , Chlorocebus aethiops , Cisteína/genética , Humanos , Ligação de Hidrogênio , Canal de Potássio Kv1.3/metabolismo , Dados de Sequência Molecular , Bloqueadores dos Canais de Potássio/metabolismo , Estrutura Secundária de Proteína , Estereoisomerismo , Verapamil/metabolismo
8.
Br J Pharmacol ; 178 Suppl 1: S157-S245, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34529831

RESUMO

The Concise Guide to PHARMACOLOGY 2021/22 is the fifth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of nearly 1900 human drug targets with an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes over 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/bph.15539. Ion channels are one of the six major pharmacological targets into which the Guide is divided, with the others being: G protein-coupled receptors, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2021, and supersedes data presented in the 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.


Assuntos
Bases de Dados de Produtos Farmacêuticos , Farmacologia , Humanos , Canais Iônicos , Bases de Conhecimento , Ligantes , Receptores Acoplados a Proteínas G
9.
Cell Physiol Biochem ; 24(1-2): 53-64, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19590193

RESUMO

Cell blebbing is a key feature in apoptosis. Because blebbing dynamically alters cell volume and regulatory volume changes have been linked to chloride (Cl) channels, we evaluated an association between blebbing and Cl channels activity. We used scanning electron microscopy, confocal laser microscopy, and cell sorting to quantify cell volume and blebbing and whole-cell recording to characterize Cl(-) currents. We found that blockade of Cl channel activity as well as inhibition of adenylyl cyclase or protein kinase A (PKA) activity suppressed ammonia-induced blebbing in the microglia cell line, BV-2. In further experiments, we elucidated the common mechanism of Cl channel activity and cyclic adenosine 3',5'-monophosphate (cAMP)-dependent pathway on cell blebbing. These experiments indicated that perfusion of cells with cAMP or the catalytic subunit of PKA activated a Cl(-) current under normotonic conditions. The pharmacological profile (sensitivity to 5-nitro-2-(3-phenylpropylamino)benzoic acid [NPPB], flufenamic acid, and [(dihydroindenyl)oxy]alkanoic acid [DIOA]), outward rectification, and kinetic of the current were identical to the swelling-activated Cl channel. Superfusion of cells with ammonia elicited an outwardly rectifying current sensitive to Cl channel blockers. We propose that ammonia induces a PKA-dependent phosphorylation of Cl channels. Localized influx of Cl(-) is followed by influx of water, required for bleb expansion.


Assuntos
Amônia/farmacologia , Apoptose , Canais de Cloreto/fisiologia , AMP Cíclico/metabolismo , Microglia/fisiologia , Adenina/análogos & derivados , Adenina/farmacologia , Adenilil Ciclases/metabolismo , Animais , Linhagem Celular , Tamanho Celular/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Condutividade Elétrica , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Microglia/citologia , Microglia/efeitos dos fármacos , Nitrobenzoatos/farmacologia , Fosforilação
10.
Bioorg Med Chem Lett ; 19(8): 2299-304, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19282171
11.
Protein Sci ; 17(1): 107-18, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18042681

RESUMO

Agitoxin 2 (AgTx2) is a 38-residue scorpion toxin, cross-linked by three disulfide bridges, which acts on voltage-gated K(+) (Kv) channels. Maurotoxin (MTX) is a 34-residue scorpion toxin with an uncommon four-disulfide bridge reticulation, acting on both Ca(2+)-activated and Kv channels. A 39-mer chimeric peptide, named AgTx2-MTX, was designed from the sequence of the two toxins and chemically synthesized. It encompasses residues 1-5 of AgTx2, followed by the complete sequence of MTX. As established by enzyme cleavage, the new AgTx2-MTX molecule displays half-cystine pairings of the type C1-C5, C2-C6, C3-C7, and C4-C8, which is different from that of MTX. The 3D structure of AgTx2-MTX solved by (1)H-NMR, revealed both alpha-helical and beta-sheet structures, consistent with a common alpha/beta scaffold of scorpion toxins. Pharmacological assays of AgTx2-MTX revealed that this new molecule is more potent than both original toxins in blocking rat Kv1.2 channel. Docking simulations, performed with the 3D structure of AgTx2-MTX, confirmed this result and demonstrated the participation of the N-terminal domain of AgTx2 in its increased affinity for Kv1.2 through additional molecular contacts. Altogether, the data indicated that replacement of the N-terminal domain of MTX by the one of AgTx2 in the AgTx2-MTX chimera results in a reorganization of the disulfide bridge arrangement and an increase of affinity to the Kv1.2 channel.


Assuntos
Venenos de Crotalídeos/química , Venenos de Crotalídeos/síntese química , Canal de Potássio Kv1.2/antagonistas & inibidores , Venenos de Escorpião/química , Venenos de Escorpião/síntese química , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Venenos de Crotalídeos/farmacologia , Humanos , Cinética , Espectroscopia de Ressonância Magnética/métodos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Proteínas Recombinantes/antagonistas & inibidores , Venenos de Escorpião/farmacologia , Transfecção
12.
J Neurochem ; 106(6): 2312-21, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18624921

RESUMO

Throughout the CNS, small conductance Ca(2+)-activated potassium (SK) channels modulate firing frequency and neuronal excitability. We have identified a novel, shorter isoform of standard SK2 (SK2-std) in mouse brain which we named SK2-sh. SK2-sh is alternatively spliced at exon 3 and therefore lacks 140 amino acids, which include transmembrane domains S3, S4 and S5, compared with SK2-std. Western blot analysis of mouse hippocampal tissue revealed a 47 kDa protein product as predicted for SK2-sh along with a 64 kDa band representing the standard SK2 isoform. Electrophysiological recordings from transiently expressed SK2-sh revealed no functional channel activity or interaction with SK2-std. With the help of real-time PCR, we found significantly higher expression levels of SK2-sh mRNA in cortical tissue from AD cases when compared with age-matched controls. A similar increase in SK2-sh expression was induced in cortical neurons from mice by cytokine exposure. Substantial clinical evidence suggests that excess cytokines are centrally involved in the pathogenesis of Alzheimer's disease. Thus, SK2-sh as a downstream target of cytokines, provide a promising target for additional investigation regarding potential therapeutic intervention.


Assuntos
Processamento Alternativo/genética , Encéfalo/metabolismo , Citocinas/metabolismo , Transdução de Sinais/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Células Cultivadas , Córtex Cerebral/metabolismo , Citocinas/farmacologia , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Peso Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa/isolamento & purificação
13.
Eur J Neurosci ; 28(11): 2173-82, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19046364

RESUMO

Proliferation of astrocytes plays an essential role during ontogeny and in the adult brain, where it occurs following trauma and in inflammation and neurodegenerative diseases as well as in normal, healthy mammals. The cellular mechanisms underlying glial proliferation remain poorly understood. As dopamine is known to modulate proliferation in different cell populations, we investigated the effects of dopamine on the proliferation of striatal astrocytes in vitro. We found that dopamine reduced proliferation. As proliferation involves, among other things, a change in cell volume, which normally comes with water movement across the membrane, water channels might represent a molecular target of the dopamine effect. Therefore we studied the effect of dopamine on aquaporin 4 (AQP4) expression, the main aquaporin subtype expressed in glial cells, and observed a down-regulation of the AQP4-M23 isoform. This down-regulation was the cause of the dopamine-induced decrease in proliferation as knockdown of AQP4 using siRNA techniques mimicked the effects of dopamine on proliferation. Furthermore, stimulation of glial proliferation by basic fibroblast growth factor was also abolished by knocking down AQP4. In addition, blocking of AQP4 with 10 mum tetraethylammonium inhibited osmotically induced cell swelling and stimulation of glial cell proliferation by basic fibroblast growth factor. These results demonstrate a clear-cut involvement of AQP4 in the regulation of proliferation and implicate that modulation of AQP4 could be used therapeutically in the treatment of neurodegenerative diseases as well as in the regulation of reactive astrogliosis by preventing or reducing the glia scar formation, thus improving regeneration following ischemia or other trauma.


Assuntos
Aquaporina 4/genética , Astrócitos/metabolismo , Proliferação de Células/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopamina/metabolismo , Gliose/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Células Cultivadas , Corpo Estriado/citologia , Corpo Estriado/crescimento & desenvolvimento , Dopamina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Gliose/tratamento farmacológico , Gliose/genética , Camundongos , Camundongos Endogâmicos BALB C , Bloqueadores dos Canais de Potássio/farmacologia , Interferência de RNA , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Tetraetilamônio/farmacologia , Equilíbrio Hidroeletrolítico/efeitos dos fármacos , Equilíbrio Hidroeletrolítico/fisiologia
14.
PLoS One ; 12(4): e0176078, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28426823

RESUMO

Current through the σ-pore was first detected in hKv1.3_V388C channels, where the V388C mutation in hKv1.3 channels opened a new pathway (σ-pore) behind the central α-pore. Typical for this mutant channel was inward current at potentials more negative than -100 mV when the central α-pore was closed. The α-pore blockers such as TEA+ and peptide toxins (CTX, MTX) could not reduce current through the σ-pore of hKv1.3_V388C channels. This new pathway would proceed in parallel to the α-pore in the S6-S6 interface gap. To see whether this phenomenon is restricted to hKv1.3 channels we mutated hKv1.2 at the homologue position (hKv1.2_V370C). By overexpression of hKv1.2_V370C mutant channels in COS-7 cells we could show typical σ-currents. The electrophysiological properties of the σ-pore in hKv1.3_V388C and hKv1.2_V370C mutant channels were similar. The σ-pore of hKv1.2_V370C channels was most permeable to Na+ and Li+ whereas Cl- and protons did not influence current through the σ-pore. Tetraethylammonium (TEA+), charybdotoxin (CTX) and maurotoxin (MTX), known α-pore blockers, could not reduce current through the σ-pore of hKv1.2_V370C channels. Taken together we conclude that the observation of σ-pore currents is not restricted to Kv1.3 potassium channels but can also be observed in a closely related potassium channel. This finding could have implications in the treatment of different ion channel diseases linked to mutations of the respective channels in regions close to homologue position investigated by us.


Assuntos
Canal de Potássio Kv1.2/fisiologia , Mutação , Substituição de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Cisteína/química , Canal de Potássio Kv1.2/química , Canal de Potássio Kv1.2/genética , Valina/química
15.
Neuropharmacology ; 50(4): 458-67, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16310228

RESUMO

Phenothiazines can be used as psychopharmaceutical agents and are known to cause many side effects during treatment since they interfere with many different cellular systems. Recently, phenothiazines were reported to block Ca(2+)-activated potassium channels of the SK type. Therefore we investigated their effect on the functionally related class of Ca(2+)-activated potassium channels of the IK type. The representative phenothiazine derivative promethazine (PTZ) blocked IK channels almost independently from the extracellular pH(o) with an IC(50) of 49 +/- 0.2 microM (pH(o) 7.4, n = 5) and 32 +/- 0.2 microM (pH(o) 6.2, n = 5) in whole cell experiments. The extracellularly applied membrane impermeable PTZ analogue methyl-promethazine (M-PTZ) had a strongly reduced blocking potency compared to PTZ. In contrast, intracellularly applied PTZ and M-PTZ had the same blocking potency on IK channels in excised inside out patch clamp experiments (K(d) = 9.3 +/- 0.5 microM for PTZ, n = 7 and 6.7 +/- 0.4 microM for M-PTZ, n = 5). The voltage dependency of the PTZ and M-PTZ block was investigated in excised inside out patch clamp experiments at a concentration of 100 microM. For both compounds the block was more pronounced at positive membrane potentials. The steepness of the voltage dependency was found to be 70 +/- 10 mV (for PTZ) and 61 +/- 6 mV (for M-PTZ) indicating that both compounds sensed approximately 40% of the entire membrane spanning electrical field from the inside. We conclude that PTZ and M-PTZ bind to a side in IK channels, which is located within the electrical field and is accessible from the intracellular side.


Assuntos
Cálcio/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Prometazina/farmacologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Antagonistas dos Receptores Histamínicos H1/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Rim , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp
16.
Biochem J ; 385(Pt 1): 95-104, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15588251

RESUMO

OSK1 (alpha-KTx3.7) is a 38-residue toxin cross-linked by three disulphide bridges that was initially isolated from the venom of the Asian scorpion Orthochirus scrobiculosus. OSK1 and several structural analogues were produced by solid-phase chemical synthesis, and were tested for lethality in mice and for their efficacy in blocking a series of 14 voltage-gated and Ca2+-activated K+ channels in vitro. In the present paper, we report that OSK1 is lethal in mice by intracerebroventricular injection, with a LD50 (50% lethal dose) value of 2 microg/kg. OSK1 blocks K(v)1.1, K(v)1.2, K(v)1.3 channels potently and K(Ca)3.1 channel moderately, with IC50 values of 0.6, 5.4, 0.014 and 225 nM respectively. Structural analogues of OSK1, in which we mutated positions 16 (Glu16-->Lys) and/or 20 (Lys20-->Asp) to amino acid residues that are conserved in all other members of the alpha-KTx3 toxin family except OSK1, were also produced and tested. Among the OSK1 analogues, [K16,D20]-OSK1 (OSK1 with Glu16-->Lys and Lys20-->Asp mutations) shows an increased potency on K(v)1.3 channel, with an IC50 value of 0.003 nM, without loss of activity on K(Ca)3.1 channel. These data suggest that OSK1 or [K16,D20]-OSK1 could serve as leads for the design and production of new immunosuppressive drugs.


Assuntos
Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Venenos de Escorpião/síntese química , Venenos de Escorpião/farmacologia , Escorpiões/química , Toxinas Biológicas/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Humanos , Concentração Inibidora 50 , Injeções Intraventriculares , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/toxicidade , Venenos de Escorpião/química , Venenos de Escorpião/toxicidade , Toxinas Biológicas/síntese química , Toxinas Biológicas/química , Toxinas Biológicas/toxicidade
17.
Proteins ; 60(3): 401-11, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15971207

RESUMO

Scorpion toxins interact with their target ion channels through multiple molecular contacts. Because a "gain of function" approach has never been described to evaluate the importance of the molecular contacts in defining toxin affinity, we experimentally examined whether increasing the molecular contacts between a toxin and an ion channel directly impacts toxin affinity. For this purpose, we focused on two scorpion peptides, the well-characterized maurotoxin with its variant Pi1-like disulfide bridging (MTX(Pi1)), used as a molecular template, and butantoxin (BuTX), used as an N-terminal domain provider. BuTX is found to be 60-fold less potent than MTX(Pi1) in blocking Kv1.2 (IC(50) values of 165 nM for BuTX versus 2.8 nM for MTX(Pi1)). Removal of its N-terminal domain (nine residues) further decreases BuTX affinity for Kv1.2 by 5.6-fold, which is in agreement with docking simulation data showing the importance of this domain in BuTX-Kv1.2 interaction. Transfer of the BuTX N-terminal domain to MTX(Pi1) results in a chimera with five disulfide bridges (BuTX-MTX(Pi1)) that exhibits 22-fold greater affinity for Kv1.2 than MTX(Pi1) itself, in spite of the lower affinity of BuTX as compared to MTX(Pi1). Docking experiments performed with the 3-D structure of BuTX-MTX(Pi1) in solution, as solved by (1)H-NMR, reveal that the N-terminal domain of BuTX participates in the increased affinity for Kv1.2 through additional molecular contacts. Altogether, the data indicate that acting on molecular contacts between a toxin and a channel is an efficient strategy to modulate toxin affinity.


Assuntos
Biologia Computacional/métodos , Canal de Potássio Kv1.2/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Proteômica/métodos , Venenos de Escorpião/química , Sequência de Aminoácidos , Animais , Dicroísmo Circular , Cisteína/química , Dissulfetos/química , Eletrofisiologia , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Escorpiões , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Ácido Trifluoracético/química
18.
Biochem J ; 377(Pt 1): 25-36, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12962541

RESUMO

Pi1 is a 35-residue scorpion toxin cross-linked by four disulphide bridges that acts potently on both small-conductance Ca2+-activated (SK) and voltage-gated (Kv) K+ channel subtypes. Two approaches were used to investigate the relative contribution of the Pi1 functional dyad (Tyr-33 and Lys-24) to the toxin action: (i) the chemical synthesis of a [A24,A33]-Pi1 analogue, lacking the functional dyad, and (ii) the production of a Pi1 analogue that is phosphorylated on Tyr-33 (P-Pi1). According to molecular modelling, this phosphorylation is expected to selectively impact the two amino acid residues belonging to the functional dyad without altering the nature and three-dimensional positioning of other residues. P-Pi1 was directly produced by peptide synthesis to rule out any possibility of trace contamination by the unphosphorylated product. Both Pi1 analogues were compared with synthetic Pi1 for bioactivity. In vivo, [A24,A33]-Pi1 and P-Pi1 are lethal by intracerebroventricular injection in mice (LD50 values of 100 and 40 microg/mouse, respectively). In vitro, [A24,A33]-Pi1 and P-Pi1 compete with 125I-apamin for binding to SK channels of rat brain synaptosomes (IC50 values of 30 and 10 nM, respectively) and block rat voltage-gated Kv1.2 channels expressed in Xenopus laevis oocytes (IC50 values of 22 microM and 75 nM, respectively), whereas they are inactive on Kv1.1 or Kv1.3 channels at micromolar concentrations. Therefore, although both analogues are less active than Pi1 both in vivo and in vitro, the integrity of the Pi1 functional dyad does not appear to be a prerequisite for the recognition and binding of the toxin to the Kv1.2 channels, thereby highlighting the crucial role of other toxin residues with regard to Pi1 action on these channels. The computed simulations detailing the docking of Pi1 peptides on to the Kv1.2 channels support an unexpected key role of specific basic amino acid residues, which form a basic ring (Arg-5, Arg-12, Arg-28 and Lys-31 residues), in toxin binding.


Assuntos
Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Venenos de Escorpião/química , Venenos de Escorpião/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Cultivadas , Canal de Potássio Kv1.2 , Lisina/fisiologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/metabolismo , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/química , Estrutura Terciária de Proteína , Ratos , Venenos de Escorpião/farmacologia , Alinhamento de Sequência , Tirosina/fisiologia , Xenopus laevis
19.
Biochem J ; 377(Pt 1): 37-49, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14498829

RESUMO

CoTX1 (cobatoxin 1) is a 32-residue toxin with three disulphide bridges that has been isolated from the venom of the Mexican scorpion Centruroides noxius Hoffmann. Here we report the chemical synthesis, disulphide bridge organization, 3-D (three-dimensional) solution structure determination, pharmacology on K+ channel subtypes (voltage-gated and Ca2+-activated) and docking-simulation experiments. An enzyme-based cleavage of the synthetic folded/oxidized CoTX1 indicated half-cystine pairs between Cys3-Cys22, Cys8-Cys27 and Cys12-Cys29. The 3-D structure of CoTX1 (solved by 1H-NMR) showed that it folds according to the common alpha/beta scaffold of scorpion toxins. In vivo, CoTX1 was lethal after intracerebroventricular injection to mice (LD50 value of 0.5 microg/mouse). In vitro, CoTX1 tested on cells expressing various voltage-gated or Ca2+-activated (IKCa1) K+ channels showed potent inhibition of currents from rat K(v)1.2 ( K(d) value of 27 nM). CoTX1 also weakly competed with 125I-labelled apamin for binding to SKCa channels (small-conductance Ca2+-activated K+ channels) on rat brain synaptosomes (IC50 value of 7.2 microM). The 3-D structure of CoTX1 was used in docking experiments which suggests a key role of Arg6 or Lys10, Arg14, Arg18, Lys21 (dyad), Ile23, Asn24, Lys28 and Tyr30 (dyad) residues of CoTX1 in its interaction with the rat K(v)1.2 channel. In addition, a [Pro7,Gln9]-CoTX1 analogue (ACoTX1) was synthesized. The two residue replacements were selected aiming to restore the RPCQ motif in order to increase peptide affinity towards SKCa channels, and to alter the CoTX1 dipole moment such that it is expected to decrease peptide activity on K(v) channels. Unexpectedly, ACoTX1 exhibited an activity similar to that of CoTX1 towards SKCa channels, while it was markedly more potent on IKCa1 and several voltage-gated K+ channels.


Assuntos
Bloqueadores dos Canais de Potássio , Canais de Potássio/metabolismo , Venenos de Escorpião , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Dicroísmo Circular , Simulação por Computador , Dissulfetos/química , Humanos , Canal de Potássio Kv1.2 , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/química , Canais de Potássio Cálcio-Ativados/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ratos , Venenos de Escorpião/síntese química , Venenos de Escorpião/química , Venenos de Escorpião/metabolismo , Venenos de Escorpião/farmacologia , Homologia de Sequência de Aminoácidos
20.
Brain Res ; 927(1): 55-68, 2002 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-11814432

RESUMO

In Xenopus laevis, several distinct K(+)-channels (xKv1.1, xKv1.2, xKv2,1, xKv2.2, xKv3.1) have been cloned, sequenced, and electrophysiologically characterized. K(+)-channels significantly shape neuronal excitability by setting the membrane potential, and latency and duration of action potentials. We identified a further Shaker homologue, xKv1.4, in X. laevis. The open reading frame encodes a K(+)-channel that shares 72% of its 698 amino acids with the human Shaker homologue, hKv1.4. Northern blot analysis revealed xKv1.4 in the brain, muscle, and spleen but not in the ovary, intestine, heart, liver, kidney, lung, and skin. Whole-cell patch clamp recording from rat basophilic leukaemia (RBL) cells transfected with xKv1.4 revealed a voltage-gated, outward rectifying, transient A-type, K(+) selective current. xKv1.4 was strongly dependent on extracellular K(+). Exposure of cells to K(+) free bath solution almost completely abolished the current, whereas in the presence of high K(+), inactivation in response to a maintained depolarizing step and the frequency-dependent cumulative inactivation decreased. Ion channels encoded by xKv1.4 are sensitive to 4-aminopyridine and quinidine but insensitive to tetraethylammonium and the peptide toxins, charybdotoxin, margatoxin, and dendrotoxin. In conclusion, our results indicate that the biophysical and pharmacological signature of xKv1.4 closely resemble those of the A-current described in Xenopus embryonic neurons and is similar to the human Shaker homologue, hKv1.4.


Assuntos
Neurônios/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Animais , Sequência de Bases , Química Encefálica/fisiologia , Células Cultivadas , Feminino , Expressão Gênica/fisiologia , Ativação do Canal Iônico/fisiologia , Canal de Potássio Kv1.4 , Leucemia Basofílica Aguda , Potenciais da Membrana/fisiologia , Dados de Sequência Molecular , Neurônios/citologia , Técnicas de Patch-Clamp , Canais de Potássio/química , Canais de Potássio/metabolismo , Ratos , Homologia de Sequência de Aminoácidos , Superfamília Shaker de Canais de Potássio , Transfecção , Células Tumorais Cultivadas , Proteínas de Xenopus , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA