Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 288(18): 12544-53, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23511633

RESUMO

Potassium channel functions are often deciphered by using selective and potent scorpion toxins. Among these toxins, only a limited subset is capable of selectively blocking small conductance Ca(2+)-activated K(+) (SK) channels. The structural bases of this selective SK channel recognition remain unclear. In this work, we demonstrate the key role of the electric charges of two conserved arginine residues (Arg-485 and Arg-489) from the SK3 channel outer vestibule in the selective recognition by the SK3-blocking BmP05 toxin. Indeed, individually substituting these residues with histidyl or lysyl (maintaining the positive electric charge partially or fully), although decreasing BmP05 affinity, still preserved the toxin sensitivity profile of the SK3 channel (as evidenced by the lack of recognition by many other types of potassium channel-sensitive charybdotoxin). In contrast, when Arg-485 or Arg-489 of the SK3 channel was mutated to an acidic (Glu) or alcoholic (Ser) amino acid residue, the channel lost its sensitivity to BmP05 and became susceptible to the "new" blocking activity by charybdotoxin. In addition to these SK3 channel basic residues important for sensitivity, two acidic residues, Asp-492 and Asp-518, also located in the SK3 channel outer vestibule, were identified as being critical for toxin affinity. Furthermore, molecular modeling data indicate the existence of a compact SK3 channel turret conformation (like a peptide screener), where the basic rings of Arg-485 and Arg-489 are stabilized by strong ionic interactions with Asp-492 and Asp-518. In conclusion, the unique properties of Arg-485 and Arg-489 (spatial orientations and molecular interactions) in the SK3 channel account for its toxin sensitivity profile.


Assuntos
Arginina/metabolismo , Modelos Moleculares , Venenos de Escorpião/química , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Arginina/genética , Charibdotoxina/química , Charibdotoxina/metabolismo , Células HEK293 , Humanos , Venenos de Escorpião/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/química , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética
2.
Biophys J ; 105(8): 1829-37, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24138859

RESUMO

The Ca(2+)-activated channel of intermediate-conductance (KCa3.1) is a target for antisickling and immunosuppressant agents. Many small peptides isolated from animal venoms inhibit KCa3.1 with nanomolar affinities and are promising drug scaffolds. Although the inhibitory effect of peptide toxins on KCa3.1 has been examined extensively, the structural basis of toxin-channel recognition has not been understood in detail. Here, the binding modes of two selected scorpion toxins, charybdotoxin (ChTx) and OSK1, to human KCa3.1 are examined in atomic detail using molecular dynamics (MD) simulations. Employing a homology model of KCa3.1, we first determine conduction properties of the channel using Brownian dynamics and ascertain that the simulated results are in accord with experiment. The model structures of ChTx-KCa3.1 and OSK1-KCa3.1 complexes are then constructed using MD simulations biased with distance restraints. The ChTx-KCa3.1 complex predicted from biased MD is consistent with the crystal structure of ChTx bound to a voltage-gated K(+) channel. The dissociation constants (Kd) for the binding of both ChTx and OSK1 to KCa3.1 determined experimentally are reproduced within fivefold using potential of mean force calculations. Making use of the knowledge we gained by studying the ChTx-KCa3.1 complex, we attempt to enhance the binding affinity of the toxin by carrying out a theoretical mutagenesis. A mutant toxin, in which the positions of two amino acid residues are interchanged, exhibits a 35-fold lower Kd value for KCa3.1 than that of the wild-type. This study provides insight into the key molecular determinants for the high-affinity binding of peptide toxins to KCa3.1, and demonstrates the power of computational methods in the design of novel toxins.


Assuntos
Charibdotoxina/química , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Simulação de Dinâmica Molecular , Sequência de Aminoácidos , Sítios de Ligação , Charibdotoxina/metabolismo , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/química , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ligação Proteica , Venenos de Escorpião/química , Venenos de Escorpião/metabolismo
3.
Biophys J ; 100(10): 2466-74, 2011 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-21575581

RESUMO

Free energy calculations for protein-ligand dissociation have been tested and validated for small ligands (50 atoms or less), but there has been a paucity of studies for larger, peptide-size ligands due to computational limitations. Previously we have studied the energetics of dissociation in a potassium channel-charybdotoxin complex by using umbrella sampling molecular-dynamics simulations, and established the need for carefully chosen coordinates and restraints to maintain the physiological ligand conformation. Here we address the ligand integrity problem further by constructing additional potential of mean forces for dissociation of charybdotoxin using restraints. We show that the large discrepancies in binding free energy arising from simulation artifacts can be avoided by using appropriate restraints on the ligand, which enables determination of the binding free energy within the chemical accuracy. We make several suggestions for optimal choices of harmonic potential parameters and restraints to be used in binding studies of large ligands.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Charibdotoxina/química , Charibdotoxina/metabolismo , Canais de Potássio/química , Canais de Potássio/metabolismo , Amidas/química , Fenômenos Biomecânicos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Propriedades de Superfície , Termodinâmica
4.
Biophys J ; 101(11): 2652-60, 2011 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22261053

RESUMO

The conduction properties of the voltage-gated potassium channel Kv1.3 and its modes of interaction with several polypeptide venoms are examined using Brownian dynamics simulations and molecular dynamics calculations. Employing an open-state homology model of Kv1.3, we first determine current-voltage and current-concentration curves and ascertain that simulated results accord with experimental measurements. We then investigate, using a molecular docking method and molecular dynamics simulations, the complexes formed between the Kv1.3 channel and several Kv-specific polypeptide toxins that are known to interfere with the conducting mechanisms of several classes of voltage-gated K(+) channels. The depths of potential of mean force encountered by charybdotoxin, α-KTx3.7 (also known as OSK1) and ShK are, respectively, -19, -27, and -25 kT. The dissociation constants calculated from the profiles of potential of mean force correspond closely to the experimentally determined values. We pinpoint the residues in the toxins and the channel that are critical for the formation of the stable venom-channel complexes.


Assuntos
Canal de Potássio Kv1.3/metabolismo , Modelos Moleculares , Toxinas Biológicas/metabolismo , Animais , Charibdotoxina/química , Charibdotoxina/metabolismo , Venenos de Cnidários/química , Venenos de Cnidários/metabolismo , Ligação de Hidrogênio , Ativação do Canal Iônico , Íons , Canal de Potássio Kv1.3/química , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Ratos , Venenos de Escorpião/química , Venenos de Escorpião/metabolismo , Eletricidade Estática , Termodinâmica , Toxinas Biológicas/química
5.
Biophys J ; 101(11): 2671-8, 2011 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22261055

RESUMO

Using both Brownian and molecular dynamics, we replicate many of the salient features of Kv1.2, including the current-voltage-concentration profiles and the binding affinity and binding mechanisms of charybdotoxin, a scorpion venom. We also elucidate how structural differences in the inner vestibule can give rise to significant differences in its permeation characteristics. Current-voltage-concentration profiles are constructed using Brownian dynamics simulations, based on the crystal structure 2A79. The results are compatible with experimental data, showing similar conductance, rectification, and saturation with current. Unlike KcsA, for example, the inner pore of Kv1.2 is mainly hydrophobic and neutral, and to explore the consequences of this, we investigate the effect of mutating neutral proline residues at the mouth of the inner vestibule to charged aspartate residues. We find an increased conductance, less inward rectification, and quicker saturation of the current-voltage profile. Our simulations use modifications to our Brownian dynamics program that extend the range of channels that can be usefully modeled. Using molecular dynamics, we investigate the binding of the charybdotoxin scorpion venom to the outer vestibule of the channel. A potential of mean force is derived using umbrella sampling, giving a dissociation constant within a factor of ∼2 to experimentally derived constants. The residues involved in the toxin binding are in agreement with experimental mutagenesis studies. We thus show that the experimental observations on the voltage-gated channel, including the toxin-channel interaction, can reliably be replicated by using the two widely used computational tools.


Assuntos
Permeabilidade da Membrana Celular , Canal de Potássio Kv1.2/antagonistas & inibidores , Canal de Potássio Kv1.2/metabolismo , Modelos Biológicos , Simulação de Dinâmica Molecular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Charibdotoxina/química , Charibdotoxina/metabolismo , Charibdotoxina/farmacologia , Eletricidade , Ativação do Canal Iônico/efeitos dos fármacos , Porosidade/efeitos dos fármacos , Termodinâmica
6.
Biophys J ; 96(7): 2577-88, 2009 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-19348743

RESUMO

Ion channel-toxin complexes are ideal systems for computational studies of protein-ligand interactions, because, in most cases, the channel axis provides a natural reaction coordinate for unbinding of a ligand and a wealth of physiological data is available to check the computational results. We use a recently determined structure of a potassium channel-charybdotoxin complex in molecular dynamics simulations to investigate the mechanism and energetics of unbinding. Pairs of residues on the channel protein and charybdotoxin that are involved in the binding are identified, and their behavior is traced during umbrella-sampling simulations as charybdotoxin is moved away from the binding site. The potential of mean force for the unbinding of charybdotoxin is constructed from the umbrella sampling simulations using the weighted histogram analysis method, and barriers observed are correlated with specific breaking of interactions and influx of water molecules into the binding site. Charybdotoxin is found to undergo conformational changes as a result of the reaction coordinate choice--a nontrivial decision for larger ligands--which we explore in detail, and for which we propose solutions. Agreement between the calculated and the experimental binding energies is obtained once the energetic consequences of these conformational changes are included in the calculations.


Assuntos
Charibdotoxina/metabolismo , Modelos Moleculares , Canais de Potássio/metabolismo , Charibdotoxina/química , Ligantes , Espectroscopia de Ressonância Magnética , Canais de Potássio/química , Ligação Proteica , Conformação Proteica , Termodinâmica , Água/metabolismo
7.
Science ; 280(5360): 106-9, 1998 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-9525854

RESUMO

Toxins from scorpion venom interact with potassium channels. Resin-attached, mutant K+ channels from Streptomyces lividans were used to screen venom from Leiurus quinquestriatus hebraeus, and the toxins that interacted with the channel were rapidly identified by mass spectrometry. One of the toxins, agitoxin2, was further studied by mutagenesis and radioligand binding. The results show that a prokaryotic K+ channel has the same pore structure as eukaryotic K+ channels. This structural conservation, through application of techniques presented here, offers a new approach for K+ channel pharmacology.


Assuntos
Proteínas de Bactérias , Canais de Potássio/química , Canais de Potássio/metabolismo , Conformação Proteica , Venenos de Escorpião/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Charibdotoxina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação Puntual , Bloqueadores dos Canais de Potássio , Canais de Potássio/genética , Ensaio Radioligante , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Superfamília Shaker de Canais de Potássio , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Streptomyces/química
8.
Elife ; 82019 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-31271355

RESUMO

In silico and in vitro studies have made progress in understanding protein-protein complex formation; however, the molecular mechanisms for their dissociation are unclear. Protein-protein complexes, lasting from microseconds to years, often involve induced-fit, challenging computational or kinetic analysis. Charybdotoxin (CTX), a peptide from the Leiurus scorpion venom, blocks voltage-gated K+-channels in a unique example of binding/unbinding simplicity. CTX plugs the external mouth of K+-channels pore, stopping K+-ion conduction, without inducing conformational changes. Conflicting with a tight binding, we show that external permeant ions enhance CTX-dissociation, implying a path connecting the pore, in the toxin-bound channel, with the external solution. This sensitivity is explained if CTX wobbles between several bound conformations, producing transient events that restore the electrical and ionic trans-pore gradients. Wobbling may originate from a network of contacts in the interaction interface that are in dynamic stochastic equilibria. These partially-bound intermediates could lead to distinct, and potentially manipulable, dissociation pathways.


Assuntos
Charibdotoxina/metabolismo , Íons/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Potássio/metabolismo , Animais , Aracnídeos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Ligação Proteica , Conformação Proteica
9.
Neuron ; 40(1): 15-23, 2003 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-14527430

RESUMO

I(Ks) voltage-gated K(+) channels contain four pore-forming KCNQ1 subunits and MinK accessory subunits in a number that has been controversial. Here, I(Ks) channels assembled naturally by monomer subunits are compared to those with linked subunits that force defined stoichiometries. Two strategies that exploit charybdotoxin (CTX)-sensitive subunit variants are applied. First, CTX on rate, off rate, and equilibrium affinity are found to be the same for channels of monomers and those with a fixed 2:4 MinK:KCNQ1 valence. Second, 3H-CTX and an antibody are used to directly quantify channels and MinK subunits, respectively, showing 1.97 +/- 0.07 MinK per I(Ks) channel. Additional MinK subunits do not enter channels of monomeric subunits or those with fixed 2:4 valence. We conclude that two MinK subunits are necessary, sufficient, and the norm in I(Ks) channels. This stoichiometry is expected for other K(+) channels that contain MinK or MinK-related peptides (MiRPs).


Assuntos
Charibdotoxina/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Canais de Potássio/química , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Feminino , Humanos , Dados de Sequência Molecular , Mutação , Canais de Potássio/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Ligação Proteica/fisiologia , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Homologia de Sequência de Aminoácidos , Xenopus
10.
Neuron ; 16(1): 123-30, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8562075

RESUMO

Charybdotoxin, a peptide neurotoxin of known molecular structure, blocks Shaker K+ channels by binding to a receptor at the outer opening of the ion conduction pathway. Analysis of variants of CTX at position 29 and of Shaker at position 449 shows that these two residues interact closely in the channel-toxin complex. The CTX mutation M29I leads to a slight strengthening of block when tested on Shaker-449T; the same CTX mutation weakens block 1700-fold when tested on Shaker-449F. The known position of CTX-29 on the toxin's interaction surface thus locates Shaker-449 within 5 A of the pore axis of the closed channel. All four subunits must carry the 449F mutation to produce a highly toxin-insensitive channel.


Assuntos
Charibdotoxina/química , Canais de Potássio/química , Conformação Proteica , Sequência de Aminoácidos , Animais , Sítios de Ligação , Charibdotoxina/genética , Charibdotoxina/metabolismo , Charibdotoxina/farmacologia , Proteínas de Drosophila , Drosophila melanogaster/genética , Ativação do Canal Iônico/efeitos dos fármacos , Lisina/química , Dados de Sequência Molecular , Oócitos , Fenilalanina/química , Potássio/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , Canais de Potássio/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Escorpiões/genética , Superfamília Shaker de Canais de Potássio , Treonina/química , Transfecção , Xenopus laevis
11.
Sci Rep ; 8(1): 4571, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29545539

RESUMO

Large-conductance Ca2+- and voltage-dependent K+ (BK) channels display diverse biological functions while their pore-forming α subunit is coded by a single Slo1 gene. The variety of BK channels is correlated with the effects of BKα coexpression with auxiliary ß (ß1-ß4) subunits, as well as newly defined γ subunits. Charybdotoxin (ChTX) blocks BK channel through physically occluding the K+-conduction pore. Human brain enriched ß4 subunit (hß4) alters the conductance-voltage curve, slows activation and deactivation time courses of BK channels. Its extracellular loop (hß4-loop) specifically impedes ChTX to bind BK channel pore. However, the structure of ß4 subunit's extracellular loop and the molecular mechanism for gating kinetics, toxin sensitivity of BK channels regulated by ß4 are still unclear. To address them, here, we first identified four disulfide bonds in hß4-loop by mass spectroscopy and NMR techniques. Then we determined its three-dimensional solution structure, performed NMR titration and electrophysiological analysis, and found that residue Asn123 of ß4 subunit regulated the gating and pharmacological characteristics of BK channel. Finally, by constructing structure models of BKα/ß4 and thermodynamic double-mutant cycle analysis, we proposed that BKα subunit might interact with ß4 subunit through the conserved residue Glu264(BKα) coupling with residue Asn123(ß4).


Assuntos
Charibdotoxina/química , Canais de Potássio Ativados por Cálcio de Condutância Alta/química , Charibdotoxina/metabolismo , Microscopia Crioeletrônica , Dissulfetos/química , Humanos , Cinética , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Espectrometria de Massas , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação
12.
Biomed Res Int ; 2016: 9497041, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28018918

RESUMO

Stretch-induced relaxation has not been clearly identified in gastrointestinal tract. The present study is to explore the role of large conductance calcium-activated potassium channels (BKCa) in stretch-induced relaxation of colon. The expression and currents of BKCa were detected and the basal muscle tone and contraction amplitude of colonic smooth muscle strips were measured. The expression of BKCa in colon is higher than other GI segments (P < 0.05). The density of BKCa currents was very high in colonic smooth muscle cells (SMCs). BKCa in rat colonic SMCs were sensitive to stretch. The relaxation response of colonic SM strips to stretch was attenuated by charybdotoxin (ChTX), a nonspecific BKCa blocker (P < 0.05). After blocking enteric nervous activities by tetrodotoxin (TTX), the stretch-induced relaxation did not change (P > 0.05). Still, ChTX and iberiotoxin (IbTX, a specific BKCa blocker) attenuated the relaxation of the colonic muscle strips enduring stretch (P < 0.05). These results suggest stretch-activation of BKCa in SMCs was involved in the stretch-induced relaxation of colon. Our study highlights the role of mechanosensitive ion channels in SMCs in colon motility regulation and their physiological and pathophysiological significance is worth further study.


Assuntos
Colo/fisiologia , Fenômenos Mecânicos , Relaxamento Muscular/fisiologia , Músculo Liso/fisiologia , Animais , Cálcio/metabolismo , Charibdotoxina/metabolismo , Colo/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Peptídeos/administração & dosagem , Ratos , Tetrodotoxina/administração & dosagem
13.
Toxicon ; 101: 70-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25953725

RESUMO

Animal venoms, such as those from scorpions, are a potent source for new pharmacological substances. In this study we have determined the structure of the α-KTx3.8 (named as Bs6) scorpion toxin by multidimensional (1)H homonuclear NMR spectroscopy and investigated its function by molecular dynamics (MD) simulations and electrophysiological measurements. Bs6 is a potent inhibitor of the Kv1.3 channel which plays an important role during the activation and proliferation of memory T-cells (TEM), which play an important role in autoimmune diseases. Therefore, it could be an interesting target for treatment of autoimmune diseases. In this study, Bs6 was synthesised by solid phase synthesis and its three-dimensional (3D) structure has been determined. To gain a deeper insight into the interaction of Bs6 with different potassium channels like hKv1.1 and hKv1.3, the protein-protein complex was modelled based on known toxin-channel structures and tested for stability in MD simulations using GROMACS. The toxin-channel interaction was further analysed by electrophysiological measurements of different potassium channels like hKv1.3 and hKv7.1. As potassium channel inhibitors could play an important role to overcome autoimmune diseases like multiple sclerosis and type-1 diabetes mellitus, our data contributes to the understanding of the molecular mechanism of action and will ultimately help to develop new potent inhibitors in future.


Assuntos
Fenômenos Eletrofisiológicos , Espectroscopia de Ressonância Magnética/métodos , Simulação de Acoplamento Molecular , Venenos de Escorpião/química , Superfamília Shaker de Canais de Potássio/química , Técnicas de Síntese em Fase Sólida/métodos , Animais , Doenças Autoimunes/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Charibdotoxina/metabolismo , Humanos , Concentração Inibidora 50 , Canal de Potássio KCNQ1/química , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/química , Conformação Molecular , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Conformação Proteica , Venenos de Escorpião/farmacologia , Escorpiões/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
14.
Protein Sci ; 12(2): 266-77, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12538890

RESUMO

Animal toxins are small proteins built on the basis of a few disulfide bonded frameworks. Because of their high variability in sequence and biologic function, these proteins are now used as templates for protein engineering. Here we report the extensive characterization of the structure and dynamics of two toxin folds, the "three-finger" fold and the short alpha/beta scorpion fold found in snake and scorpion venoms, respectively. These two folds have a very different architecture; the short alpha/beta scorpion fold is highly compact, whereas the "three-finger" fold is a beta structure presenting large flexible loops. First, the crystal structure of the snake toxin alpha was solved at 1.8-A resolution. Then, long molecular dynamics simulations (10 ns) in water boxes of the snake toxin alpha and the scorpion charybdotoxin were performed, starting either from the crystal or the solution structure. For both proteins, the crystal structure is stabilized by more hydrogen bonds than the solution structure, and the trajectory starting from the X-ray structure is more stable than the trajectory started from the NMR structure. The trajectories started from the X-ray structure are in agreement with the experimental NMR and X-ray data about the protein dynamics. Both proteins exhibit fast motions with an amplitude correlated to their secondary structure. In contrast, slower motions are essentially only observed in toxin alpha. The regions submitted to rare motions during the simulations are those that exhibit millisecond time-scale motions. Lastly, the structural variations within each fold family are described. The localization and the amplitude of these variations suggest that the regions presenting large-scale motions should be those tolerant to large insertions or deletions.


Assuntos
Charibdotoxina/química , Proteínas Neurotóxicas de Elapídeos/química , Engenharia de Proteínas , Sequência de Aminoácidos , Animais , Charibdotoxina/metabolismo , Proteínas Neurotóxicas de Elapídeos/metabolismo , Simulação por Computador , Cristalografia por Raios X , Ligação de Hidrogênio , Modelos Moleculares , Dados de Sequência Molecular , Movimento (Física) , Ressonância Magnética Nuclear Biomolecular , Dobramento de Proteína , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Temperatura
15.
Proteins ; 52(2): 146-54, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12833539

RESUMO

To gain insight into the molecular determinants that define the specificity of interaction of pore-blocking peptides, such as agitoxin 2 (AgTX2), charybdotoxin (ChTX), and iberiotoxin (IbTX) with the Shaker-type voltage-gated potassium channel Kv1.3, or the large-conductance Ca(2+)-activated K(+) (Maxi-K) channel, homology models of these channels were generated based on the crystal structure of the bacterial, KcsA, potassium channel. Peptide-channel complexes were analyzed to evaluate the predicted interaction interfaces between the peptides and the channels' outer vestibules. The docking model, for either AgTX2 or ChTX with the Kv1.3 channel, predicts a novel hydrogen bonding interaction between the Asn30 side-chain of the peptide and the Asp381 side-chain of the channel. This interaction is consistent with the >500-fold decreased potency of both AgTX2 and ChTX mutants at position 30 for the Shaker channel [(Ranganathan et al., Neuron 1996;16:131-139); (Goldstein et al., Neuron 1994;12:1377-1388)]. This hydrogen bonding interaction also suggests that Gly30 in IbTX may be the critical determinant for its lack of activity against Shaker Kv channels. The model of the Maxi-K channel reveals a narrower and more structurally restrained outer vestibule in which the aromatic residues Phe266 and Tyr294 may stabilize binding of IbTX and ChTX by pi-pi stacking with the aromatic residues Trp14 and Tyr36 of the peptides. This study also suggests that the extra net negative charge of IbTX is not related to the selectivity of this peptide for the Maxi-K channel.


Assuntos
Bloqueadores dos Canais de Potássio/química , Canais de Potássio Cálcio-Ativados/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio/química , Venenos de Escorpião/química , Toxinas Biológicas/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Charibdotoxina/química , Charibdotoxina/metabolismo , Ligação de Hidrogênio , Canal de Potássio Kv1.3 , Canais de Potássio Ativados por Cálcio de Condutância Alta , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/metabolismo , Canais de Potássio/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ligação Proteica , Subunidades Proteicas , Venenos de Escorpião/metabolismo , Alinhamento de Sequência
16.
Neuropharmacology ; 35(7): 915-21, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8938722

RESUMO

Charybdotoxin is a small peptide blocker of K+ channels, rigidly held in active conformation by three disulfide bonds. The toxin blocks K+ channels by binding to a receptor site located at the external "vestibule", and thus physically occluding the outer opening of the K+ conduction pore. In the blocked complex, K27, a residue on the toxin's molecular surface, projects its epsilon-amino group into the K(+)-selective pore. The results here show that CTX, produced by heterologous expression in E. coli, may be manipulated to place unnatural positively charged residues at position 27. The toxin folds faithfully to its native conformation when the crucial lysine at position 27 is replaced by a cysteine residue, a maneuver that allows specific chemical modification of this side-chain. Replacements of K27 by side-chains slightly shorter or slightly longer than lysine yield active toxins. The toxin variant with ornithine at this position interacts much less strongly with K+ ions in the pore of slowpoke-type Ca2(+)-activated K+ channels than does wild-type toxin. This result argues that the epsilon-amino group of K27 in bound toxin lies only a few ångstroms away from a K+ ion occupying the blocked pore. The peptide folds with high efficiency to form the correct disulfides even in the presence of strong denaturants.


Assuntos
Charibdotoxina/metabolismo , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Charibdotoxina/química , Cisteína/química , Escherichia coli/genética , Escherichia coli/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta , Mutação , Ornitina/química , Ligação Proteica , Conformação Proteica , Dobramento de Proteína
17.
Br J Pharmacol ; 126(8): 1707-16, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10372812

RESUMO

1. UK-78,282, a novel piperidine blocker of the T lymphocyte voltage-gated K+ channel, Kv1.3, was discovered by screening a large compound file using a high-throughput 86Rb efflux assay. This compound blocks Kv1.3 with a IC50 of approximately 200 nM and 1:1 stoichiometry. A closely related compound, CP-190,325, containing a benzyl moiety in place of the benzhydryl in UK-78,282, is significantly less potent. 2 Three lines of evidence indicate that UK-78,282 inhibits Kv1.3 in a use-dependent manner by preferentially blocking and binding to the C-type inactivated state of the channel. Increasing the fraction of inactivated channels by holding the membrane potential at - 50 mV enhances the channel's sensitivity to UK-78,282. Decreasing the number of inactivated channels by exposure to approximately 160 mM external K+ decreases the sensitivity to UK-78,282. Mutations that alter the rate of C-type inactivation also change the channel's sensitivity to UK-78,282 and there is a direct correlation between tau(h) and IC50 values. 3. Competition experiments suggest that UK-78,282 binds to residues at the inner surface of the channel overlapping the site of action of verapamil. Internal tetraethylammonium and external charybdotoxin do not compete UK-78,282's action on the channel. 4. UK-78,282 displays marked selectivity for Kv1.3 over several other closely related K+ channels, the only exception being the rapidly inactivating voltage-gated K+ channel, Kv1.4. 5. UK-78,282 effectively suppresses human T-lymphocyte activation.


Assuntos
Compostos Benzidrílicos/farmacologia , Imunossupressores/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Piperidinas/farmacologia , Bloqueadores dos Canais de Potássio , Linfócitos T/efeitos dos fármacos , Animais , Ligação Competitiva , Células COS , Bovinos , Charibdotoxina/metabolismo , Charibdotoxina/farmacologia , Células HeLa , Humanos , Radioisótopos do Iodo , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Canais de Potássio/metabolismo , Canais de Potássio/fisiologia , Ratos , Ratos Endogâmicos Lew , Radioisótopos de Rubídio , Linfócitos T/imunologia , Tetraetilamônio/metabolismo , Tetraetilamônio/farmacologia
18.
Br J Pharmacol ; 121(1): 141-9, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9146898

RESUMO

1. In the rat hepatic artery, the acetylcholine-induced relaxation mediated by endothelium-derived hyperpolarizing factor (EDHF) is abolished by a combination of apamin and charybdotoxin, inhibitors of small (SKCa) and large (BKCa) conductance calcium-sensitive potassium (K)-channels, respectively, but not by each toxin alone. The selective BKCa inhibitor iberiotoxin cannot replace charybdotoxin in this combination. Since delayed rectifier K-channels (KV) represent another target for charybdotoxin, we explored the possible involvement of KV in EDHF-mediated relaxation in this artery. 2. The KV inhibitors, agitoxin-2 (0.3 microM), kaliotoxin (0.3 microM), beta-dendrotoxin (0.3 microM), dofetilide (1 microM) and terikalant (10 microM), each in combination with apamin (0.3 microM) had no effect on the EDHF-mediated relaxation induced by acetylcholine in the presence of N omega-nitro-L-arginine (0.3 mM) and indomethacin (10 microM), inhibitors of nitric oxide (NO) synthase and cyclo-oxygenase, respectively (n = 2-3). Although the KV inhibitor margatoxin (0.3 microM) was also without effect (n = 5), the combination of margatoxin and apamin produced a small inhibition of the response (pEC50 and Emax values were 7.5 +/- 0.0 and 95 +/- 1% in the absence and 7.0 +/- 0.1 and 81 +/- 6% in the presence of margatoxin plus apamin, respectively; n = 6; P < 0.05). 3. Ciclazindol (10 microM) partially inhibited the EDHF-mediated relaxation by shifting the acetylcholine-concentration-response curve 12 fold to the right (n = 6; P < 0.05) and abolished the response when combined with apamin (0.3 microM; n = 6). This combination did not inhibit acetylcholine-induced relaxations mediated by endothelium-derived NO (n = 5). 4. A 4-aminopyridine-sensitive delayed rectifier current (IK(V)) was identified in freshly-isolated single smooth muscle cells from rat hepatic artery. None of the cells displayed a rapidly-activating and -inactivating A-type current. Neither charybdotoxin (0.3 microM; n = 3) nor ciclazindol (10 microM; n = 5), alone or in combination with apamin (0.3 microM; n = 4-5), had an effect on IK(V). A tenfold higher concentration of ciclazindol (0.1 mM, n = 4) markedly inhibited IK(V), but this effect was not increased in the additional presence of apamin (0.3 microM; n = 2). 5. By use of membranes prepared from rat brain cortex. [125I]-charybdotoxin binding was consistent with an interaction at a single site with a KD of approximately 25 pM. [125I]-charybdotoxin binding was unaffected by iberiotoxin (0.1 microM, n = 6), but was increased by apamin in a concentration-dependent manner (Emax 43 +/- 10%, P < 0.05 and pEC50 7.1 +/- 0.2; n = 7-8). Agitoxin-2 (10 nM) displaced [125I]-charybdotoxin binding by 91 +/- 3% (n = 6) and prevented the effect of apamin (1 microM; n = 6). 6. It is concluded that the EDHF-mediated relaxation in the rat hepatic artery is not mediated by the opening of either KV or BKCa. Instead, the target K-channels for EDHF seem to be structurally related to both KV and BKCa. The possibility that a subtype of SKCa may be the target for EDHF is discussed.


Assuntos
Fatores Biológicos/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , Artéria Hepática/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico Sintase/antagonistas & inibidores , Canais de Potássio/fisiologia , 4-Aminopiridina/farmacologia , Acetilcolina/farmacologia , Animais , Antiarrítmicos/metabolismo , Antiarrítmicos/farmacologia , Apamina/metabolismo , Apamina/farmacologia , Ligação Competitiva , Fatores Biológicos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Charibdotoxina/metabolismo , Charibdotoxina/farmacologia , Cromanos/metabolismo , Cromanos/farmacologia , Interações Medicamentosas , Feminino , Artéria Hepática/metabolismo , Indóis/metabolismo , Indóis/farmacologia , Indometacina/farmacologia , Relaxamento Muscular/efeitos dos fármacos , Relaxamento Muscular/fisiologia , Músculo Liso Vascular/fisiologia , Nitroarginina/farmacologia , Técnicas de Patch-Clamp , Fenetilaminas/metabolismo , Fenetilaminas/farmacologia , Piperidinas/metabolismo , Piperidinas/farmacologia , Bloqueadores dos Canais de Potássio , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sulfonamidas/metabolismo , Sulfonamidas/farmacologia
19.
J Neurotrauma ; 18(7): 691-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11497095

RESUMO

Evidence in the literature suggests that endothelium-derived hyperpolarizing factor (EDHF) may act in a compensatory manner such that during conditions of compromised nitric oxide (NO), EDHF serves as a back-up mechanism. Given that constitutive NO synthase is chronically downregulated after head trauma, we tested the hypothesis that EDHF is potentiated following injury. Male adult rats were subjected to either sham injury (n = 27) or mild controlled cortical impact (CCI) injury (n = 26). Branches of the middle cerebral artery (MCA) directly within the contusion site were harvested either 1 or 24 h later, pressurized to 60 mm Hg in a vessel chamber and allowed to develop spontaneous tone. Relaxation to luminal application of adenosine triphosphate (ATP) was similar in all groups. Relaxation to ATP in the presence of L-NAME (N(G)-nitro-L-arginine methyl ester) and indomethacin was similar in all groups except for vessels isolated at 24 h following mild CCI injury. In this case, L-NAME and indomethacin had no effect on the ATP-mediated dilation. The ATP-mediated dilation in L-NAME and indomethacin-treated MCA branches was inhibited by charybdotoxin, an inhibitor of large conductance Ca2+-sensitive K+ channels. These findings suggest that there is a significant potentiation of the EDHF-mediated dilation to ATP in cerebral arteries isolated at 24 h following mild CCI injury.


Assuntos
Fatores Biológicos/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Endotélio Vascular/metabolismo , Artéria Cerebral Média/metabolismo , Trifosfato de Adenosina/metabolismo , Análise de Variância , Animais , Charibdotoxina/metabolismo , Dilatação Patológica/metabolismo , Dilatação Patológica/patologia , Modelos Animais de Doenças , Indometacina/metabolismo , Masculino , Artéria Cerebral Média/patologia , NG-Nitroarginina Metil Éster/metabolismo , Ratos , Ratos Long-Evans , Fatores de Tempo
20.
Exp Biol Med (Maywood) ; 228(5): 474-80, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12709572

RESUMO

The activation of big-conductance K(Ca) channels in vascular smooth muscle cells by carbon monoxide (CO) has been demonstrated previously. One specific target of CO on K(Ca) channel proteins is the histidine residue. The roles of other amino acid residues on the functionality of K(Ca) channels, as well as their reactions to CO, have been unclear. In the present study, the cell-free single channel recording technique was used to investigate the chemical modification of K(Ca) channels by CO and other chemical agents. The modification of negatively charged carboxyl groups and the epsilon -amino group of lysine did not affect the open probability, but decreased single-channel conductance of K(Ca) channels. When sulfhydryl groups of cysteine were modified with N-ethylmaleimide, the open probability of K(Ca) channels was decreased, but single-channel conductance was not affected. None of the above chemical modifications affected the CO-induced increase in the open probability of K(Ca) channels. However, N-ethylmaleimide treatment reduced the stimulatory effect of nitric oxide (NO) on K(Ca) channels. Finally, pretreatment of smooth muscle cells with NO abolished the effects of subsequently applied CO on K(Ca) channel proteins. Our study demonstrates that CO and NO acted on different amino acid residues of K(Ca) channel proteins. The interaction of CO and NO determines the functional status of K(Ca) channels in vascular smooth muscle cells


Assuntos
Monóxido de Carbono/metabolismo , Miócitos de Músculo Liso/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Animais , Células Cultivadas , Charibdotoxina/metabolismo , Etilmaleimida/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Óxido Nítrico/metabolismo , Oniocompostos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio Cálcio-Ativados/genética , Ratos , Reagentes de Sulfidrila/metabolismo , Ácido Trinitrobenzenossulfônico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA