Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PNAS Nexus ; 1(3): pgac083, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36741467

RESUMO

Kv3 ion-channels constitute a class of functionally distinct voltage-gated ion channels characterized by their ability to fire at a high frequency. Several disease relevant mutants, together with biological data, suggest the importance of this class of ion channels as drug targets for CNS disorders, and several drug discovery efforts have been reported. Despite the increasing interest for this class of ion channels, no structure of a Kv3 channel has been reported yet. We have determined the cryo-EM structure of Kv3.1 at 2.6 Å resolution using full-length wild type protein. When compared to known structures for potassium channels from other classes, a novel domain organization is observed with the cytoplasmic T1 domain, containing a well-resolved Zinc site and displaying a rotation by 35°. This suggests a distinct cytoplasmic regulation mechanism for the Kv3.1 channel. A high resolution structure was obtained for Kv3.1 in complex with a novel positive modulator Lu AG00563. The structure reveals a novel ligand binding site for the Kv class of ion channels located between the voltage sensory domain and the channel pore, a region which constitutes a hotspot for disease causing mutations. The discovery of a novel binding site for a positive modulator of a voltage-gated potassium channel could shed light on the mechanism of action for these small molecule potentiators. This finding could enable structure-based drug design on these targets with high therapeutic potential for the treatment of multiple CNS disorders.

2.
EJNMMI Res ; 10(1): 49, 2020 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-32409881

RESUMO

BACKGROUND: Accidental peripheral nerve injury during surgical intervention results in a broad spectrum of potentially debilitating side effects. Tissue distortion and poor visibility can significantly increase the risk of nerve injury with long-lasting consequences for the patient. We developed and characterized Hs1a-FL, a fluorescent near-infrared molecule for nerve visualization in the operating theater with the aim of helping physicians to visualize nerves during surgery. Hs1a was derived from the venom of the Chinese bird spider, Haplopelma schmidti, and conjugated to Cy7.5 dye. Hs1a-FL was injected intravenously in mice, and harvested nerves were imaged microscopically and with epifluorescence. RESULTS: Hs1a-FL showed specific and stable binding to the sodium channel NaV1.7, present on the surface of human and mouse nerves. Hs1a-FL allowed epifluorescence visualization of sciatic mouse nerves with favorable nerve-to-muscle contrast. CONCLUSIONS: Fluorescent NaV1.7-targeted tracers have the potential to be adopted clinically for the intraoperative visualization of peripheral nerves during surgery, providing guidance for the surgeon and potentially improving the standard of care.

3.
Mol Pharmacol ; 88(6): 1002-10, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26429937

RESUMO

Many spider-venom peptides are known to modulate the activity of the voltage-gated sodium (NaV) subtype 1.7 (NaV1.7) channel, which has emerged as a promising analgesic target. In particular, a class of spider-venom peptides (NaSpTx1) has been found to potently inhibit NaV1.7 (nanomolar IC50), and has been shown to produce analgesic effects in animals. However, one member of this family [µ-TRTX-Hhn2b (Hhn2b)] does not inhibit mammalian NaV channels expressed in dorsal root ganglia at concentrations up to 100 µM. This peptide is classified as a NaSpTx1 member by virtue of its cysteine spacing and sequence conservation over functionally important residues. Here, we have performed detailed structural and functional analyses of Hhn2b, leading us to identify two nonpharmacophore residues that contribute to human NaV1.7 (hNaV1.7) inhibition by nonoverlapping mechanisms. These findings allowed us to produce a double mutant of Hhn2b that shows nanomolar inhibition of hNaV1.7. Traditional structure/function analysis did not provide sufficient resolution to identify the mechanism underlying the observed gain of function. However, by solving the high-resolution structure of both the wild-type and mutant peptides using advanced multidimensional NMR experiments, we were able to uncover a previously unknown network of interactions that stabilize the pharmacophore region of this class of venom peptides. We further monitored the lipid binding properties of the peptides and identified that one of the key amino acid substitutions also selectively modulates the binding of the peptide to anionic lipids. These results will further aid the development of peptide-based analgesics for the treatment of chronic pain.


Assuntos
Analgésicos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Engenharia Genética/métodos , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Fragmentos de Peptídeos/metabolismo , Venenos de Aranha/metabolismo , Sequência de Aminoácidos , Analgésicos/química , Animais , Células CHO , Cricetinae , Cricetulus , Feminino , Genes de Troca/genética , Humanos , Canal de Sódio Disparado por Voltagem NAV1.7/química , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Venenos de Aranha/química , Venenos de Aranha/genética , Xenopus laevis
4.
Br J Pharmacol ; 172(10): 2445-58, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25754331

RESUMO

BACKGROUND AND PURPOSE: Chronic pain is a serious worldwide health issue, with current analgesics having limited efficacy and dose-limiting side effects. Humans with loss-of-function mutations in the voltage-gated sodium channel NaV 1.7 (hNaV 1.7) are indifferent to pain, making hNaV 1.7 a promising target for analgesic development. Since spider venoms are replete with NaV channel modulators, we examined their potential as a source of hNaV 1.7 inhibitors. EXPERIMENTAL APPROACH: We developed a high-throughput fluorescent-based assay to screen spider venoms against hNaV 1.7 and isolate 'hit' peptides. To examine the binding site of these peptides, we constructed a panel of chimeric channels in which the S3b-S4 paddle motif from each voltage sensor domain of hNaV 1.7 was transplanted into the homotetrameric KV 2.1 channel. KEY RESULTS: We screened 205 spider venoms and found that 40% contain at least one inhibitor of hNaV 1.7. By deconvoluting 'hit' venoms, we discovered seven novel members of the NaSpTx family 1. One of these peptides, Hd1a (peptide µ-TRTX-Hd1a from venom of the spider Haplopelma doriae), inhibited hNaV 1.7 with a high level of selectivity over all other subtypes, except hNaV 1.1. We showed that Hd1a is a gating modifier that inhibits hNaV 1.7 by interacting with the S3b-S4 paddle motif in channel domain II. The structure of Hd1a, determined using heteronuclear NMR, contains an inhibitor cystine knot motif that is likely to confer high levels of chemical, thermal and biological stability. CONCLUSION AND IMPLICATIONS: Our data indicate that spider venoms are a rich natural source of hNaV 1.7 inhibitors that might be useful leads for the development of novel analgesics.


Assuntos
Analgésicos/análise , Descoberta de Drogas/métodos , Ensaios de Triagem em Larga Escala/métodos , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Venenos de Aranha/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/análise , Sequência de Aminoácidos , Analgésicos/química , Analgésicos/farmacologia , Animais , Humanos , Dados de Sequência Molecular , Venenos de Aranha/análise , Venenos de Aranha/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia
5.
J Mol Biol ; 427(1): 158-175, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25088688

RESUMO

Ion channels are vital contributors to cellular communication in a wide range of organisms, a distinct feature that renders this ubiquitous family of membrane-spanning proteins a prime target for toxins found in animal venom. For many years, the unique properties of these naturally occurring molecules have enabled researchers to probe the structural and functional features of ion channels and to define their physiological roles in normal and diseased tissues. To illustrate their considerable impact on the ion channel field, this review will highlight fundamental insights into toxin-channel interactions and recently developed toxin screening methods and practical applications of engineered toxins.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Toxinas Biológicas/farmacologia , Animais , Humanos , Transdução de Sinais/efeitos dos fármacos
6.
Biochem Pharmacol ; 89(2): 276-86, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24561180

RESUMO

Spider venoms are replete with peptidic ion channel modulators, often with novel subtype selectivity, making them a rich source of pharmacological tools and drug leads. In a search for subtype-selective blockers of voltage-gated calcium (CaV) channels, we isolated and characterized a novel 39-residue peptide, ω-TRTX-Cc1a (Cc1a), from the venom of the tarantula Citharischius crawshayi (now Pelinobius muticus). Cc1a is 67% identical to the spider toxin ω-TRTX-Hg1a, an inhibitor of CaV2.3 channels. We assembled Cc1a using a combination of Boc solid-phase peptide synthesis and native chemical ligation. Oxidative folding yielded two stable, slowly interconverting isomers. Cc1a preferentially inhibited Ba(2+) currents (IBa) mediated by L-type (CaV1.2 and CaV1.3) CaV channels heterologously expressed in Xenopus oocytes, with half-maximal inhibitory concentration (IC50) values of 825nM and 2.24µM, respectively. In rat dorsal root ganglion neurons, Cc1a inhibited IBa mediated by high voltage-activated CaV channels but did not affect low voltage-activated T-type CaV channels. Cc1a exhibited weak activity at NaV1.5 and NaV1.7 voltage-gated sodium (NaV) channels stably expressed in mammalian HEK or CHO cells, respectively. Experiments with modified Cc1a peptides, truncated at the N-terminus (ΔG1-E5) or C-terminus (ΔW35-V39), demonstrated that the N- and C-termini are important for voltage-gated ion channel modulation. We conclude that Cc1a represents a novel pharmacological tool for probing the structure and function of L-type CaV channels.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Sistemas de Liberação de Medicamentos/métodos , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/isolamento & purificação , Venenos de Aranha/síntese química , Venenos de Aranha/isolamento & purificação , Sequência de Aminoácidos , Animais , Células CHO , Bloqueadores dos Canais de Cálcio/administração & dosagem , Bloqueadores dos Canais de Cálcio/síntese química , Bloqueadores dos Canais de Cálcio/isolamento & purificação , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/administração & dosagem , Ratos , Ratos Wistar , Venenos de Aranha/administração & dosagem , Aranhas , Xenopus laevis
7.
Proc Natl Acad Sci U S A ; 110(43): 17534-9, 2013 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-24082113

RESUMO

Loss-of-function mutations in the human voltage-gated sodium channel NaV1.7 result in a congenital indifference to pain. Selective inhibitors of NaV1.7 are therefore likely to be powerful analgesics for treating a broad range of pain conditions. Herein we describe the identification of µ-SLPTX-Ssm6a, a unique 46-residue peptide from centipede venom that potently inhibits NaV1.7 with an IC50 of ∼25 nM. µ-SLPTX-Ssm6a has more than 150-fold selectivity for NaV1.7 over all other human NaV subtypes, with the exception of NaV1.2, for which the selectivity is 32-fold. µ-SLPTX-Ssm6a contains three disulfide bonds with a unique connectivity pattern, and it has no significant sequence homology with any previously characterized peptide or protein. µ-SLPTX-Ssm6a proved to be a more potent analgesic than morphine in a rodent model of chemical-induced pain, and it was equipotent with morphine in rodent models of thermal and acid-induced pain. This study establishes µ-SPTX-Ssm6a as a promising lead molecule for the development of novel analgesics targeting NaV1.7, which might be suitable for treating a wide range of human pain pathologies.


Assuntos
Analgésicos/farmacologia , Venenos de Artrópodes/química , Morfina/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7/fisiologia , Dor/prevenção & controle , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Artrópodes/química , Sequência de Bases , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Humanos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oócitos/fisiologia , Dor/fisiopatologia , Técnicas de Patch-Clamp , Peptídeos/genética , Peptídeos/metabolismo , Ratos , Xenopus laevis
8.
PLoS One ; 8(5): e63865, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667680

RESUMO

Disulfide-rich peptides are the dominant component of most animal venoms. These peptides have received much attention as leads for the development of novel therapeutic agents and bioinsecticides because they target a wide range of neuronal receptors and ion channels with a high degree of potency and selectivity. In addition, their rigid disulfide framework makes them particularly well suited for addressing the crucial issue of in vivo stability. Structural and functional characterization of these peptides necessitates the development of a robust, reliable expression system that maintains their native disulfide framework. The bacterium Escherichia coli has long been used for economical production of recombinant proteins. However, the expression of functional disulfide-rich proteins in the reducing environment of the E. coli cytoplasm presents a significant challenge. Thus, we present here an optimised protocol for the expression of disulfide-rich venom peptides in the periplasm of E. coli, which is where the endogenous machinery for production of disulfide-bonds is located. The parameters that have been investigated include choice of media, induction conditions, lysis methods, methods of fusion protein and peptide purification, and sample preparation for NMR studies. After each section a recommendation is made for conditions to use. We demonstrate the use of this method for the production of venom peptides ranging in size from 2 to 8 kDa and containing 2-6 disulfide bonds.


Assuntos
Dissulfetos/metabolismo , Escherichia coli/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Periplasma/metabolismo , Proteínas Recombinantes/biossíntese , Animais , Soluções Tampão , Vetores Genéticos/genética , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Relação Estrutura-Atividade , Temperatura
9.
Toxicon ; 60(4): 478-91, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22543187

RESUMO

Voltage-gated sodium (Na(V)) channels play a central role in the propagation of action potentials in excitable cells in both humans and insects. Many venomous animals have therefore evolved toxins that modulate the activity of Na(V) channels in order to subdue their prey and deter predators. Spider venoms in particular are rich in Na(V) channel modulators, with one-third of all known ion channel toxins from spider venoms acting on Na(V) channels. Here we review the landscape of spider-venom peptides that have so far been described to target vertebrate or invertebrate Na(V) channels. These peptides fall into 12 distinct families based on their primary structure and cysteine scaffold. Some of these peptides have become useful pharmacological tools, while others have potential as therapeutic leads because they target specific Na(V) channel subtypes that are considered to be important analgesic targets. Spider venoms are conservatively predicted to contain more than 10 million bioactive peptides and so far only 0.01% of this diversity been characterised. Thus, it is likely that future research will reveal additional structural classes of spider-venom peptides that target Na(V) channels.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Peptídeos/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Venenos de Aranha/farmacologia , Sequência de Aminoácidos , Animais , Descoberta de Drogas , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/metabolismo , Venenos de Aranha/química , Venenos de Aranha/metabolismo , Aranhas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA