Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 289(42): 29261-72, 2014 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-25190810

RESUMO

Tolerance is a well described component of alcohol abuse and addiction. The large conductance voltage- and Ca(2+)-gated potassium channel (BK) has been very useful for studying molecular tolerance. The influence of association with the ß4 subunit can be observed at the level of individual channels, action potentials in brain slices, and finally, drinking behavior in the mouse. Previously, we showed that 50 mm alcohol increases both α and αß4 BK channel open probability, but only α BK develops acute tolerance to this effect. Currently, we explore the possibility that the influence of the ß4 subunit on tolerance may result from a striking effect of ß4 on kinase modulation of the BK channel. We examine the influence of the ß4 subunit on PKA, CaMKII, and phosphatase modulation of channel activity, and on molecular tolerance to alcohol. We record from human BK channels heterologously expressed in HEK 293 cells composed of its core subunit, α alone (Insertless), or co-expressed with the ß4 BK auxiliary subunit, as well as, acutely dissociated nucleus accumbens neurons using the cell-attached patch clamp configuration. Our results indicate that BK channels are strongly modulated by activation of specific kinases (PKA and CaMKII) and phosphatases. The presence of the ß4 subunit greatly influences this modulation, allowing a variety of outcomes for BK channel activity in response to acute alcohol.


Assuntos
Etanol/química , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Charibdotoxina/química , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Eletrofisiologia , Células HEK293 , Humanos , Neurônios/metabolismo , Núcleo Accumbens/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Potássio/metabolismo , Fatores de Tempo
2.
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
3.
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
4.
Bioorg Med Chem Lett ; 23(24): 6743-6, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24252544

RESUMO

The botulinum neurotoxins, characterized by their neuromuscular paralytic effects, are the most toxic proteins known to man. Due to their extreme potency, ease of production, and duration of activity, the BoNT proteins have been classified by the Centers for Disease Control as high threat agents for bioterrorism. In an attempt to discover effective BoNT therapeutics, we have pursued a strategy in which we leverage the blockade of K(+) channels that ultimately results in the reversal of neuromuscular paralysis. Towards this end, we utilized peptides derived from scorpion venom that are highly potent K(+) channel blockers. Herein, we report the synthesis of charybdotoxin, a 37 amino acid peptide, and detail its activity, along with iberiotoxin and margatoxin, in a mouse phrenic nerve hemidiaphragm assay in the absence and the presence of BoNT/A.


Assuntos
Toxinas Botulínicas , Charibdotoxina/química , Paralisia/induzido quimicamente , Paralisia/tratamento farmacológico , Nervo Frênico/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/química , Venenos de Escorpião , Sequência de Aminoácidos , Animais , Charibdotoxina/farmacologia , Charibdotoxina/uso terapêutico , Camundongos , Dados de Sequência Molecular , Contração Muscular/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Potássio/uso terapêutico , Resinas Sintéticas/química , Venenos de Escorpião/química , Venenos de Escorpião/farmacologia , Venenos de Escorpião/uso terapêutico
5.
Biochim Biophys Acta ; 1808(1): 454-60, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20974108

RESUMO

Recent studies have indicated a calcium-activated large conductance potassium channel in rat brain mitochondrial inner membrane (mitoBK channel). Accordingly, we have characterized the functional and pharmacological profile of a BK channel from rat brain mitochondria in the present study. Brain mitochondrial inner membrane preparations were subjected to SDS-PAGE analysis and channel protein reconstitution into planar lipid bilayers. Western blotting and antibodies directed against various cellular proteins revealed that mitochondrial inner membrane fractions did not contain specific proteins of the other subcellular compartments except a very small fraction of endoplasmic reticulum. Channel incorporation into planar lipid bilayers revealed a voltage dependent 211 pS potassium channel with a voltage for half activation (V(1/2)) of 11.4±1.1mV and an effective gating charge z(d) of 4.7±0.9. Gating and conducting behaviors of this channel were unaffected by the addition of 2.5mM ATP, and 500 nM charybdotoxin (ChTx), but the channel appeared sensitive to 100 nM iberiotoxin (IbTx). Adding 10mM TEA at positive potentials and 10mM 4-AP at negative or positive voltages inhibited the channel activities. These results demonstrate that the mitoBK channel, present in brain mitochondrial inner membrane, displays different pharmacological properties than those classically described for plasma membrane, especially in regard to its sensitivity to iberiotoxin and charybdotoxin sensitivity.


Assuntos
Eletrofisiologia/métodos , Canal de Potássio Kv1.1/efeitos dos fármacos , Canal de Potássio Kv1.1/metabolismo , Mitocôndrias/metabolismo , Animais , Biofísica/métodos , Encéfalo/metabolismo , Membrana Celular/metabolismo , Charibdotoxina/química , Peptídeos/química , Fosfatidilcolinas/química , Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Wistar , Frações Subcelulares/metabolismo
6.
Protein Sci ; 31(12): e4506, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36369672

RESUMO

Epilepsy is the results from the imbalance between inhibition and excitation in neural circuits, which is mainly treated by some chemical drugs with side effects. Gain-of-function of BK channels or knockout of its ß4 subunit associates with spontaneous epilepsy. Currently, few reports were published about the efficacy of BK(α + ß4) channel modulators in epilepsy prevention. Charybdotoxin is a non-specific inhibitor of BK and other K+ channels. Here, by nuclear magnetic resonance (NMR) and other biochemical techniques, we found that charybdotoxin might interact with the extracellular loop of human ß4 subunit (i.e., hß4-loop) of BK(α + ß4) channel at a molar ratio 4:1 (hß4-loop vs. charybdotoxin). Charybdotoxin enhanced its ability to prevent K+ current of BK(α + ß4 H101Y) channel. The charybdotoxin Q18F variant selectively reduced the neuronal spiking frequency and increased interspike intervals of BK(α + ß4) channel by π-π stacking interactions between its residue Phe18 and residue His101 of hß4-loop. Moreover, intrahippocampal infusion of charybdotoxin Q18F variant significantly increased latency time of seizure, reduced seizure duration and seizure numbers on pentylenetetrazole-induced pre-sensitized rats, inhibited hippocampal hyperexcitability and c-Fos expression, and displayed neuroprotective effects on hippocampal neurons. These results implied that charybdotoxin Q18F variant could be potentially used for intractable epilepsy treatment by therapeutically targeting BK(α + ß4) channel.


Assuntos
Charibdotoxina , Epilepsia , Canais de Potássio Ativados por Cálcio de Condutância Alta , Animais , Humanos , Ratos , Charibdotoxina/química , Charibdotoxina/farmacologia , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Neurônios/metabolismo , Peptídeos/metabolismo , Convulsões/tratamento farmacológico , Convulsões/metabolismo
7.
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
8.
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
9.
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
10.
Proc Natl Acad Sci U S A ; 105(5): 1478-82, 2008 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-18223154

RESUMO

Ion channels are multisubunit proteins responsible for the generation and propagation of action potentials in nerve, skeletal muscle, and heart as well as maintaining salt and water homeostasis in epithelium. The subunit composition and stoichiometry of these membrane protein complexes underlies their physiological function, as different cells pair ion-conducting alpha-subunits with specific regulatory beta-subunits to produce complexes with diverse ion-conducting and gating properties. However, determining the number of alpha- and beta-subunits in functioning ion channel complexes is challenging and often fraught with contradictory results. Here we describe the synthesis of a chemically releasable, irreversible K(+) channel inhibitor and its iterative application to tally the number of beta-subunits in a KCNQ1/KCNE1 K(+) channel complex. Using this inhibitor in electrical recordings, we definitively show that there are two KCNE subunits in a functioning tetrameric K(+) channel, breaking the apparent fourfold arrangement of the ion-conducting subunits. This digital determination rules out any measurable contribution from supra, sub, and multiple stoichiometries, providing a uniform structural picture to interpret KCNE beta-subunit modulation of voltage-gated K(+) channels and the inherited mutations that cause dysfunction. Moreover, the architectural asymmetry of the K(+) channel complex affords a unique opportunity to therapeutically target ion channels that coassemble with KCNE beta-subunits.


Assuntos
Membrana Celular/química , Charibdotoxina/análogos & derivados , Dissulfetos/farmacologia , Canal de Potássio KCNQ1/análise , Canais de Potássio de Abertura Dependente da Tensão da Membrana/análise , Subunidades Proteicas/análise , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Charibdotoxina/síntese química , Charibdotoxina/química , Charibdotoxina/farmacologia , Dissulfetos/síntese química , Dissulfetos/química , Humanos , Canal de Potássio KCNQ1/antagonistas & inibidores , Canal de Potássio KCNQ1/metabolismo , Oócitos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/metabolismo , Xenopus
11.
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
12.
Cancer Med ; 8(4): 1679-1693, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30806044

RESUMO

Breast cancer is one of the most common malignant tumors among women worldwide. About 70-75% of primary breast cancers belong to estrogen receptor (ER)-positive breast cancer. In the development of ER-positive breast cancer, abnormal activation of the ERα pathway plays an important role and is also a key point leading to the failure of clinical endocrine therapy. In this study, we found that the small molecule peptide chlorotoxin (CTX) can significantly inhibit the proliferation, migration and invasion of breast cancer cells. In in vitro study, CTX inhibits the expression of ERα in breast cancer cells. Further studies showed that CTX can directly bind to ERα and change the protein secondary structure of its LBD domain, thereby inhibiting the ERα signaling pathway. In addition, we also found that vasodilator stimulated phosphoprotein (VASP) is a target gene of ERα signaling pathway, and CTX can inhibit breast cancer cell proliferation, migration, and invasion through ERα/VASP signaling pathway. In in vivo study, CTX significantly inhibits growth of ER overexpressing breast tumor and, more importantly, based on the mechanism of CTX interacting with ERα, we found that CTX can target ER overexpressing breast tumors in vivo. Our study reveals a new mechanism of CTX anti-ER-positive breast cancer, which also provides an important reference for the study of CTX anti-ER-related tumors.


Assuntos
Moléculas de Adesão Celular/metabolismo , Receptor alfa de Estrogênio/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Venenos de Escorpião/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Charibdotoxina/química , Charibdotoxina/isolamento & purificação , Charibdotoxina/farmacologia , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Ligação Proteica , Venenos de Escorpião/química , Venenos de Escorpião/isolamento & purificação
13.
Neuron ; 15(5): 1169-81, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7576659

RESUMO

The architecture of the pore-region of a voltage-gated K+ channel, Kv1.3, was probed using four high affinity scorpion toxins as molecular calipers. We established the structural relatedness of these toxins by solving the structures of kaliotoxin and margatoxin and comparing them with the published structure of charybdotoxin; a homology model of noxiustoxin was then developed. Complementary mutagenesis of Kv1.3 and these toxins, combined with electrostatic compliance and thermodynamic mutant cycle analyses, allowed us to identify multiple toxin-channel interactions. Our analyses reveal the existence of a shallow vestibule at the external entrance to the pore. This vestibule is approximately 28-32 A wide at its outer margin, approximately 28-34 A wide at its base, and approximately 4-8 A deep. The pore is 9-14 A wide at its external entrance and tapers to a width of 4-5 A at a depth of approximately 5-7 A from the vestibule. This structural information should directly aid in developing topological models of the pores of related ion channels and facilitate therapeutic drug design.


Assuntos
Espectroscopia de Ressonância Magnética , Canais de Potássio/química , Venenos de Escorpião/química , Sequência de Aminoácidos , Sítios de Ligação , Charibdotoxina/química , Condutividade Elétrica , Eletroquímica , Ativação do Canal Iônico , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Neurotoxinas/química , Canais de Potássio/fisiologia , Estrutura Terciária de Proteína , Soluções , Termodinâmica
14.
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
15.
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
16.
Sci Rep ; 6: 23904, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27044983

RESUMO

Due to their central role in essential physiological processes, potassium channels are common targets for animal toxins. These toxins in turn are of great value as tools for studying channel function and as lead compounds for drug development. Here, we used a direct toxin pull-down assay with immobilised KcsA potassium channel to isolate a novel KcsA-binding toxin (called Tx7335) from eastern green mamba snake (Dendroaspis angusticeps) venom. Sequencing of the toxin by Edman degradation and mass spectrometry revealed a 63 amino acid residue peptide with 4 disulphide bonds that belongs to the three-finger toxin family, but with a unique modification of its disulphide-bridge scaffold. The toxin induces a dose-dependent increase in both open probabilities and mean open times on KcsA in artificial bilayers. Thus, it unexpectedly behaves as a channel activator rather than an inhibitor. A charybdotoxin-sensitive mutant of KcsA exhibits similar susceptibility to Tx7335 as wild-type, indicating that the binding site for Tx7335 is distinct from that of canonical pore-blocker toxins. Based on the extracellular location of the toxin binding site (far away from the intracellular pH gate), we propose that Tx7335 increases potassium flow through KcsA by allosterically reducing inactivation of the channel.


Assuntos
Venenos Elapídicos/química , Elapidae , Canais de Potássio/metabolismo , Proteínas de Répteis/química , Venenos de Serpentes/química , Toxinas Biológicas/química , Regulação Alostérica , Aminoácidos/química , Animais , Sítios de Ligação , Charibdotoxina/química , Cromatografia Líquida de Alta Pressão , Dissulfetos/química , Eletrofisiologia , Concentração de Íons de Hidrogênio , Espectrometria de Massas , Membranas Artificiais , Mutação , Peptídeos/química , Probabilidade , Ligação Proteica , Estrutura Secundária de Proteína , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
17.
Biochim Biophys Acta ; 1292(1): 31-8, 1996 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-8547346

RESUMO

Charybdotoxin is a 37-residue polypeptide toxin from scorpion venom, which acts by blocking voltage-gated and Ca(2+)-activated K+ channels. We have synthesized charybdotoxin and three mono-substituted analogues using an Fmoc-tBu protocol. The Phe-2 --> Tyr analogues was chosen to introduce a site for Tyr iodination which was distinct from the K+ channel binding surface, while the Glu-12 --> Gln and Arg-19 --> His analogues were studied to probe the roles of charged residues at these positions in the structure and activity of the toxin. The synthetic native molecule was equipped with natural toxin in inhibiting the human erythrocyte Ca(2+)-dependent K+ channel. The affinities of all three analogues for the erythrocyte K+ channel were slightly reduced, with the Arg-19 --> His analogue showing the greatest increase in IC50 (2.30-fold). Two-dimensional 1H-NMR studies of these analogues showed that the Glu-12 to Gln substitution, which appeared to destabilise the N-terminal half of the alpha-helix, possibly due to the weakening of an N-terminal helix capping interaction which is apparent from our NMR data. His-21 has a pKa more than one unit below the value for a non-interacting histidine. Possible reasons for this are that the imidazolium side chain is partly buried and is located near positively charged moieties. Thus, His-21 would be neutral at physiological pH, where charybdotoxin binds to the potassium channel.


Assuntos
Charibdotoxina/análogos & derivados , Charibdotoxina/química , Bloqueadores dos Canais de Potássio , Sequência de Aminoácidos , Aminoácidos/análise , Cálcio/farmacologia , Charibdotoxina/síntese química , Charibdotoxina/farmacologia , Cromatografia Líquida de Alta Pressão , Eritrócitos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/química , Peptídeos/farmacologia , Conformação Proteica , Estrutura Secundária de Proteína
18.
Indian J Pharmacol ; 47(3): 280-4, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26069365

RESUMO

OBJECTIVE: Charybdotoxin-C (ChTx-C), from the scorpion Leiurus, quinquestriatus hebraeus blocks the calcium-activated potassium channels and causes hyper excitability of the nervous system. Detailed understanding the structure of ChTx-C, conformational stability, and intermolecular interactions are required to select the potential inhibitors of the toxin. MATERIALS AND METHODS: The structure of ChTx-C was modeled using Modeller 9v7. The amino acid residues lining the binding site were predicted and used for toxin-ligand docking studies, further, selected toxin-inhibitor complexes were studied using molecular dynamics (MD) simulations. RESULTS: The predicted structure has 91.7% of amino acids in the core and allowed regions of Ramachandran plot. A total of 133 analog compounds of existing drugs for scorpion bites were used for docking. As a result of docking, a list of compounds was shown good inhibiting properties with target protein. By analyzing the interactions, Ser 15, Lys 32 had significant interactions with selected ligand molecules and Val5, which may have hydrophobic interaction with the cyclic group of the ligand. MD simulation studies revealed that the conformation and intermolecular interactions of all selected toxin-inhibitor complexes were stable. CONCLUSION: The interactions of the ligand and active site amino acids were found out for the best-docked poses in turn helpful in designing potential antitoxins which may further be exploited in toxin based therapies.


Assuntos
Antitoxinas/química , Antitoxinas/farmacologia , Charibdotoxina/antagonistas & inibidores , Charibdotoxina/química , Desenho de Fármacos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Bloqueadores dos Canais de Potássio/antagonistas & inibidores , Animais , Domínio Catalítico , Simulação por Computador , Ligantes , Bloqueadores dos Canais de Potássio/química , Conformação Proteica , Escorpiões/química
19.
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
20.
Protein Sci ; 9(11): 2059-67, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11152117

RESUMO

HpTX2 is a toxin from the venom of Heteropoda venatoria spider that has been demonstrated to bind on Kv4.2 potassium channel. We have determined the solution structure of recombinant HpTX2 by use of conventional two-dimensional NMR techniques followed by distance-geometry and molecular dynamics. The calculated structure belongs to the Inhibitory Cystin Knot structural family that consists in a compact disulfide-bonded core, from which four loops emerge. A poorly defined two-stranded antiparallel beta-sheet (residues 20-23 and 25-28) is detected. Analysis of the electrostatic charge anisotropy allows us to propose a functional map of HpTX2 different from the one described for kappa-conotoxin PVIIA, but strongly related to the one of charybdotoxin. The orientation of the dipole moment of HpTX2 emerges through K27 which could therefore be the critical lysine residue. Close to this lysine are a second basic residue, R23, an aromatic cluster (F7, W25, W30) and an hydrophobic side chain (L24). The high density in aromatic side chains of the putative functional surface as well as the lack of an asparagine is proposed to be the structural basis of the specificity of HpTX2 toward Kv4.2 channel.


Assuntos
Neuropeptídeos/química , Neurotoxinas/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/química , Venenos de Aranha/química , Sequência de Aminoácidos , Animais , Anisotropia , Células CHO , Charibdotoxina/química , Conotoxinas/química , Cricetinae , Dissulfetos , Eletrofisiologia , Escherichia coli/metabolismo , Lisina/química , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Canais de Potássio Shal , Fatores de Tempo , ômega-Conotoxinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA