Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurophysiol ; 125(5): 1954-1972, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33852808

RESUMO

Temporal coding precision of bushy cells in the ventral cochlear nucleus (VCN), critical for sound localization and communication, depends on the generation of rapid and temporally precise action potentials (APs). Voltage-gated potassium (Kv) channels are critically involved in this. The bushy cells in rat VCN express Kv1.1, 1.2, 1.3, 1.6, 3.1, 4.2, and 4.3 subunits. The Kv1.1 subunit contributes to the generation of a temporally precise single AP. However, the understanding of the functions of other Kv subunits expressed in the bushy cells is limited. Here, we investigated the functional diversity of Kv subunits concerning their contributions to temporal coding. We characterized the electrophysiological properties of the Kv channels with different subunits using whole cell patch-clamp recording and pharmacological methods. The neuronal firing pattern changed from single to multiple APs only when the Kv1.1 subunit was blocked. The Kv subunits, including the Kv1.1, 1.2, 1.6, or 3.1, were involved in enhancing temporal coding by lowering membrane excitability, shortening AP latencies, reducing jitter, and regulating AP kinetics. Meanwhile, all the Kv subunits contributed to rapid repolarization and sharpening peaks by narrowing half-width and accelerating fall rate, and the Kv1.1 subunit also affected the depolarization of AP. The Kv1.1, 1.2, and 1.6 subunits endowed bushy cells with a rapid time constant and a low input resistance of membrane for enhancing spike timing precision. The present results indicate that the Kv channels differentially affect intrinsic membrane properties to optimize the generation of rapid and reliable APs for temporal coding.NEW & NOTEWORTHY This study investigates the roles of Kv channels in effecting precision using electrophysiological and pharmacological methods in bushy cells. Different Kv channels have varying electrophysiological characteristics, which contribute to the interplay between changes in the membrane properties and regulation of neuronal excitability which then improve temporal coding. We conclude that the Kv channels are specialized to promote the precise and rapid coding of acoustic input by optimizing the generation of reliable APs.


Assuntos
Potenciais de Ação/fisiologia , Núcleo Coclear/fisiologia , Neurônios/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Feminino , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/fisiologia , Canal de Potássio Kv1.2/antagonistas & inibidores , Canal de Potássio Kv1.2/fisiologia , Canal de Potássio Kv1.6/antagonistas & inibidores , Canal de Potássio Kv1.6/fisiologia , Masculino , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
2.
Bioorg Chem ; 100: 103918, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32428746

RESUMO

Members of the voltage-gated K+ channel subfamily (Kv1), involved in regulating transmission between neurons or to muscles, are associated with human diseases and, thus, putative targets for neurotherapeutics. This applies especially to those containing Kv1.1 α subunits which become prevalent in murine demyelinated axons and appear abnormally at inter-nodes, underlying the perturbed propagation of nerve signals. To overcome this dysfunction, akin to the consequential debilitation in multiple sclerosis (MS), small inhibitors were sought that are selective for the culpable hyper-polarising K+ currents. Herein, we report a new semi-podand - compound 3 - that was designed based on the modelling of its interactions with the extracellular pore region in a deduced Kv1.1 channel structure. After synthesis, purification, and structural characterisation, compound 3 was found to potently (IC50 = 8 µM) and selectively block Kv1.1 and 1.6 channels. The tested compound showed no apparent effect on native Nav and Cav channels expressed in F-11 cells. Compound 3 also extensively and selectively inhibited MS-related Kv1.1 homomer but not the brain native Kv1.1- or 1.6-containing channels. These collective findings highlight the therapeutic potential of compound 3 to block currents mediated by Kv1.1 channels enriched in demyelinated central neurons.


Assuntos
Canal de Potássio Kv1.1/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Animais , Linhagem Celular , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/metabolismo , Desenho de Fármacos , Células HEK293 , Humanos , Canal de Potássio Kv1.1/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Neurônios/metabolismo , Bloqueadores dos Canais de Potássio/síntese química , Ratos
3.
Proc Natl Acad Sci U S A ; 117(10): 5494-5501, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32079727

RESUMO

Somatosensory neurons have historically been classified by a variety of approaches, including structural, anatomical, and genetic markers; electrophysiological properties; pharmacological sensitivities; and more recently, transcriptional profile differentiation. These methodologies, used separately, have yielded inconsistent classification schemes. Here, we describe phenotypic differences in response to pharmacological agents as measured by changes in cytosolic calcium concentration for the rapid classification of neurons in vitro; further analysis with genetic markers, whole-cell recordings, and single-cell transcriptomics validated these findings in a functional context. Using this general approach, which we refer to as tripartite constellation analysis (TCA), we focused on large-diameter dorsal-root ganglion (L-DRG) neurons with myelinated axons. Divergent responses to the K-channel antagonist, κM-conopeptide RIIIJ (RIIIJ), reliably identified six discrete functional cell classes. In two neuronal subclasses (L1 and L2), block with RIIIJ led to an increase in [Ca] i Simultaneous electrophysiology and calcium imaging showed that the RIIIJ-elicited increase in [Ca] i corresponded to different patterns of action potentials (APs), a train of APs in L1 neurons, and sporadic firing in L2 neurons. Genetically labeled mice established that L1 neurons are proprioceptors. The single-cell transcriptomes of L1 and L2 neurons showed that L2 neurons are Aδ-low-threshold mechanoreceptors. RIIIJ effects were replicated by application of the Kv1.1 selective antagonist, Dendrotoxin-K, in several L-DRG subclasses (L1, L2, L3, and L5), suggesting the presence of functional Kv1.1/Kv1.2 heteromeric channels. Using this approach on other neuronal subclasses should ultimately accelerate the comprehensive classification and characterization of individual somatosensory neuronal subclasses within a mixed population.


Assuntos
Gânglios Espinais/citologia , Células Receptoras Sensoriais/classificação , Células Receptoras Sensoriais/fisiologia , Animais , Cálcio/metabolismo , Conotoxinas/farmacologia , Citosol/metabolismo , Gânglios Espinais/efeitos dos fármacos , Canal de Potássio Kv1.1/antagonistas & inibidores , Camundongos , Camundongos Transgênicos , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Células Receptoras Sensoriais/efeitos dos fármacos , Análise de Célula Única , Transcriptoma
4.
Biochem Pharmacol ; 174: 113782, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31881193

RESUMO

Urotoxin (α-KTx 6), a peptide from venom of the Australian scorpion Urodacus yaschenkoi, is the most potent inhibitor of Kv1.2 described to date (IC50 = 160 pM). The native peptide also inhibits Kv1.1, Kv1.3 and KCa3.1 with nanomolar affinity but its low abundance in venom precluded further studies of its actions. Here we produced recombinant Urotoxin (rUro) and characterized the molecular determinants of Kv1 channel inhibition. The 3D structure of rUro determined using NMR spectroscopy revealed a canonical cysteine-stabilised α/ß (CSα/ß) fold. Functional assessment of rUro using patch-clamp electrophysiology revealed the importance of C-terminal amidation for potency against Kv1.1-1.3 and Kv1.5. Neutralization of the putative pore-blocking K25 residue in rUro by mutation to Ala resulted in a major decrease in rUro potency against all Kv channels tested, without perturbing the toxin's structure. Reciprocal mutations in the pore of Uro-sensitive Kv1.2 and Uro-resistant Kv1.5 channels revealed a direct interaction between Urotoxin and the Kv channel pore. Our experimental work supports postulating a mechanism of action in which occlusion of the permeation pathway by the K25 residue in Urotoxin is the basis of its Kv1 inhibitory activity. Docking analysis was consistent with occlusion of the pore by K25 and the requirement of a small, non-charged amino acid in the Kv1 channel vestibule to facilitate toxin-channel interactions. Finally, computational studies revealed key interactions between the amidated C-terminus of Urotoxin and a conserved Asp residue in the turret of Kv1 channels, offering a potential rationale for potency differences between native and recombinant Urotoxin.


Assuntos
Canal de Potássio Kv1.1/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/isolamento & purificação , Venenos de Escorpião/química , Animais , Cromatografia Líquida de Alta Pressão , Escherichia coli/genética , Humanos , Canal de Potássio Kv1.1/genética , Simulação de Acoplamento Molecular , Ressonância Magnética Nuclear Biomolecular , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Conformação Proteica , Escorpiões , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Linfócitos T/metabolismo
5.
Sci Rep ; 9(1): 19307, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848433

RESUMO

ShK is a 35-residue disulfide-linked polypeptide produced by the sea anemone Stichodactyla helianthus, which blocks the potassium channels Kv1.1 and Kv1.3 with pM affinity. An analogue of ShK has been developed that blocks Kv1.3 > 100 times more potently than Kv1.1, and has completed Phase 1b clinical trials for the treatment of autoimmune diseases such as psoriasis and rheumatoid arthritis. Previous studies have indicated that ShK undergoes a conformational exchange that is critical to its function, but this has proved difficult to characterise. Here, we have used high hydrostatic pressure as a tool to increase the population of the alternative state, which is likely to resemble the active form that binds to the Kv1.3 channel. By following changes in chemical shift with pressure, we have derived the chemical shift values of the low- and high-pressure states, and thus characterised the locations of structural changes. The main difference is in the conformation of the Cys17-Cys32 disulfide, which is likely to affect the positions of the critical Lys22-Tyr23 pair by twisting the 21-24 helix and increasing the solvent exposure of the Lys22 sidechain, as indicated by molecular dynamics simulations.


Assuntos
Venenos de Cnidários/química , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.3/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/química , Sequência de Aminoácidos/genética , Animais , Doenças Autoimunes/tratamento farmacológico , Venenos de Cnidários/genética , Venenos de Cnidários/farmacologia , Humanos , Canal de Potássio Kv1.1/química , Canal de Potássio Kv1.1/ultraestrutura , Canal de Potássio Kv1.3/química , Canal de Potássio Kv1.3/ultraestrutura , Conformação Molecular , Simulação de Dinâmica Molecular , Peptídeos/química , Peptídeos/genética , Bloqueadores dos Canais de Potássio/farmacologia , Anêmonas-do-Mar/química
6.
CNS Neurosci Ther ; 25(4): 442-451, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30242974

RESUMO

AIMS: Kv1.1 (KCNA1) channels contribute to the control of neuronal excitability and have been associated with epilepsy. Kv1.1 channels can associate with the cytoplasmic Kvß1 subunit resulting in rapid inactivating A-type currents. We hypothesized that removal of channel inactivation, by modulating Kv1.1/Kvß1 interaction with a small molecule, would lead to decreased neuronal excitability and anticonvulsant activity. METHODS: We applied high-throughput screening to identify ligands able to modulate the Kv1.1-T1 domain/Kvß1 protein complex. We then selected a compound that was characterized on recombinant Kv1.1/Kvß1 channels by electrophysiology and further evaluated on sustained neuronal firing and on in vitro epileptiform activity using a high K+ -low Ca2+ model in hippocampal slices. RESULTS: We identified a novel compound able to modulate the interaction of the Kv1.1/Kvß1 complex and that produced a functional inhibition of Kv1.1/Kvß1 channel inactivation. We demonstrated that this compound reduced the sustained repetitive firing in hippocampal neurons and was able to abolish the development of in vitro epileptiform activity. CONCLUSIONS: This study describes a rational drug discovery approach for the identification of novel ligands that inhibit Kv1.1 channel inactivation and provides pharmacological evidence that such a mechanism translates into physiological effects by reducing in vitro epileptiform activity.


Assuntos
Potenciais de Ação/fisiologia , Descoberta de Drogas/métodos , Hipocampo/fisiologia , Canal de Potássio Kv1.1/fisiologia , Neurônios/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Ensaios de Triagem em Larga Escala/métodos , Hipocampo/efeitos dos fármacos , Humanos , Canal de Potássio Kv1.1/agonistas , Canal de Potássio Kv1.1/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Bloqueadores dos Canais de Potássio/farmacologia , Estrutura Secundária de Proteína , Ratos , Xenopus laevis
7.
Am J Physiol Cell Physiol ; 316(2): C154-C161, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30427720

RESUMO

Voltage-gated Kv1.1 potassium channel α-subunits, encoded by the Kcna1 gene, have traditionally been regarded as neural-specific with no expression or function in the heart. However, recent data revealed that Kv1.1 subunits are expressed in atria where they may have an overlooked role in controlling repolarization and arrhythmia susceptibility independent of the nervous system. To explore this concept in more detail and to identify functional and molecular effects of Kv1.1 channel impairment in the heart, atrial cardiomyocyte patch-clamp electrophysiology and gene expression analyses were performed using Kcna1 knockout ( Kcna1-/-) mice. Specifically, we hypothesized that Kv1.1 subunits contribute to outward repolarizing K+ currents in mouse atria and that their absence prolongs cardiac action potentials. In voltage-clamp experiments, dendrotoxin-K (DTX-K), a Kv1.1-specific inhibitor, significantly reduced peak outward K+ currents in wild-type (WT) atrial cells but not Kcna1-/- cells, demonstrating an important contribution by Kv1.1-containing channels to mouse atrial repolarizing currents. In current-clamp recordings, Kcna1-/- atrial myocytes exhibited significant action potential prolongation which was exacerbated in right atria, effects that were partially recapitulated in WT cells by application of DTX-K. Quantitative RT-PCR measurements showed mRNA expression remodeling in Kcna1-/- atria for several ion channel genes that contribute to the atrial action potential including the Kcna5, Kcnh2, and Kcnj2 potassium channel genes and the Scn5a sodium channel gene. This study demonstrates a previously undescribed heart-intrinsic role for Kv1.1 subunits in mediating atrial repolarization, thereby adding a new member to the already diverse collection of known K+ channels in the heart.


Assuntos
Potenciais de Ação/fisiologia , Átrios do Coração/metabolismo , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/genética , Miócitos Cardíacos/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Feminino , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Canal de Potássio Kv1.1/deficiência , Masculino , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética
8.
J Med Chem ; 60(6): 2245-2256, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28225274

RESUMO

K+ channels containing Kv1.1 α subunits, which become prevalent at internodes in demyelinated axons, may underlie their dysfunctional conduction akin to muscle weakness in multiple sclerosis. Small inhibitors were sought with selectivity for the culpable hyper-polarizing K+ currents. Modeling of interactions with the extracellular pore in a Kv1.1-deduced structure identified diaryldi(2-pyrrolyl)methane as a suitable scaffold with optimized alkyl ammonium side chains. The resultant synthesized candidate [2,2'-((5,5'(di-p-topyldiaryldi(2-pyrrolyl)methane)bis(2,2'carbonyl)bis(azanediyl)) diethaneamine·2HCl] (8) selectively blocked Kv1.1 channels (IC50 ≈ 15 µM) recombinantly expressed in mammalian cells, induced a positive shift in the voltage dependency of K+ current activation, and slowed its kinetics. It preferentially inhibited channels containing two or more Kv1.1 subunits regardless of their positioning in concatenated tetramers. In slices of corpus callosum from mice subjected to a demyelination protocol, this novel inhibitor improved neuronal conduction, highlighting its potential for alleviating symptoms in multiple sclerosis.


Assuntos
Canal de Potássio Kv1.1/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Animais , Linhagem Celular , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Desenho de Fármacos , Humanos , Canal de Potássio Kv1.1/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Bloqueadores dos Canais de Potássio/uso terapêutico , Pirróis/química , Pirróis/farmacologia , Pirróis/uso terapêutico
9.
Hear Res ; 345: 57-68, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28065805

RESUMO

Low-voltage-activated K+ (gKL) and hyperpolarization-activated mixed cation conductances (gh) mediate currents, IKL and Ih, through channels of the Kv1 (KCNA) and HCN families respectively and give auditory neurons the temporal precision required for signaling information about the onset, fine structure, and time of arrival of sounds. Being partially activated at rest, gKL and gh contribute to the resting potential and shape responses to even small subthreshold synaptic currents. Resting gKL and gh also affect the coupling of somatic depolarization with the generation of action potentials. To learn how these important conductances are regulated we have investigated how genetic perturbations affect their expression in octopus cells of the ventral cochlear nucleus (VCN). We report five new findings: First, the magnitude of gh and gKL varied over more than two-fold between wild type strains of mice. Second, average resting potentials are not different in different strains of mice even in the face of large differences in average gKL and gh. Third, IKL has two components, one being α-dendrotoxin (α-DTX)-sensitive and partially inactivating and the other being α-DTX-insensitive, tetraethylammonium (TEA)-sensitive, and non-inactivating. Fourth, the loss of Kv1.1 results in diminution of the α-DTX-sensitive IKL, and compensatory increased expression of an α-DTX-insensitive, tetraethylammonium (TEA)-sensitive IKL. Fifth, Ih and IKL are balanced at the resting potential in all wild type and mutant octopus cells even when resting potentials vary in individual cells over nearly 10 mV, indicating that the resting potential influences the expression of gh and gKL. The independence of resting potentials on gKL and gh shows that gKL and gh do not, over days or weeks, determine the resting potential but rather that the resting potential plays a role in regulating the magnitude of either or both gKL and gh.


Assuntos
Vias Auditivas/metabolismo , Núcleo Coclear/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canal de Potássio Kv1.1/genética , Potenciais da Membrana , Canais de Potássio/genética , Animais , Vias Auditivas/citologia , Vias Auditivas/efeitos dos fármacos , Núcleo Coclear/citologia , Núcleo Coclear/efeitos dos fármacos , Regulação da Expressão Gênica , Genótipo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/antagonistas & inibidores , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/deficiência , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/deficiência , Potenciais da Membrana/efeitos dos fármacos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Plasticidade Neuronal , Técnicas de Patch-Clamp , Fenótipo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/deficiência , Fatores de Tempo
10.
Biochem Biophys Res Commun ; 482(4): 1135-1140, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27916464

RESUMO

The neurotoxic cone snail peptide µ-GIIIA specifically blocks skeletal muscle voltage-gated sodium (NaV1.4) channels. The related conopeptides µ-PIIIA and µ-SIIIA, however, exhibit a wider activity spectrum by also inhibiting the neuronal NaV channels NaV1.2 and NaV1.7. Here we demonstrate that those µ-conopeptides with a broader target range also antagonize select subtypes of voltage-gated potassium channels of the KV1 family: µ-PIIIA and µ-SIIIA inhibited KV1.1 and KV1.6 channels in the nanomolar range, while being inactive on subtypes KV1.2-1.5 and KV2.1. Construction and electrophysiological evaluation of chimeras between KV1.5 and KV1.6 revealed that these toxins block KV channels involving their pore regions; the subtype specificity is determined in part by the sequence close to the selectivity filter but predominantly by the so-called turret domain, i.e. the extracellular loop connecting the pore with transmembrane segment S5. Conopeptides µ-SIIIA and µ-PIIIA, thus, are not specific for NaV channels, and the known structure of some KV channel subtypes may provide access to structural insight into the molecular interaction between µ-conopeptides and their target channels.


Assuntos
Conotoxinas/química , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.2/antagonistas & inibidores , Canal de Potássio Kv1.4/antagonistas & inibidores , Canal de Potássio Kv1.6/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/química , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Eletrofisiologia , Células HEK293 , Humanos , Neurônios/metabolismo , Peptídeos/química , Domínios Proteicos
11.
Nat Commun ; 7: 12102, 2016 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-27381274

RESUMO

Brain development and interictal function are unaffected in many paroxysmal neurological channelopathies, possibly explained by homoeostatic plasticity of synaptic transmission. Episodic ataxia type 1 is caused by missense mutations of the potassium channel Kv1.1, which is abundantly expressed in the terminals of cerebellar basket cells. Presynaptic action potentials of small inhibitory terminals have not been characterized, and it is not known whether developmental plasticity compensates for the effects of Kv1.1 dysfunction. Here we use visually targeted patch-clamp recordings from basket cell terminals of mice harbouring an ataxia-associated mutation and their wild-type littermates. Presynaptic spikes are followed by a pronounced afterdepolarization, and are broadened by pharmacological blockade of Kv1.1 or by a dominant ataxia-associated mutation. Somatic recordings fail to detect such changes. Spike broadening leads to increased Ca(2+) influx and GABA release, and decreased spontaneous Purkinje cell firing. We find no evidence for developmental compensation for inherited Kv1.1 dysfunction.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Ataxia/fisiopatologia , Canalopatias/fisiopatologia , Canal de Potássio Kv1.1/metabolismo , Mioquimia/fisiopatologia , Células de Purkinje/metabolismo , Animais , Ataxia/genética , Ataxia/metabolismo , Cálcio/metabolismo , Canalopatias/genética , Canalopatias/metabolismo , Modelos Animais de Doenças , Venenos Elapídicos/farmacologia , Feminino , Expressão Gênica , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/genética , Camundongos , Camundongos Transgênicos , Microtomia , Mutação , Mioquimia/genética , Mioquimia/metabolismo , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/patologia , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/patologia , Transmissão Sináptica/efeitos dos fármacos , Técnicas de Cultura de Tecidos , Ácido gama-Aminobutírico/metabolismo
12.
J Biol Chem ; 291(13): 7097-106, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-26817841

RESUMO

The structural similarity between defensins and scorpion neurotoxins suggests that they might have evolved from a common ancestor. However, there is no direct experimental evidence demonstrating a functional link between scorpion neurotoxins and defensins. The scorpion defensin BmKDfsin4 from Mesobuthus martensiiKarsch contains 37 amino acid residues and a conserved cystine-stabilized α/ß structural fold. The recombinant BmKDfsin4, a classical defensin, has been found to have inhibitory activity against Gram-positive bacteria such as Staphylococcus aureus, Bacillus subtilis, and Micrococcus luteusas well as methicillin-resistant Staphylococcus aureus Interestingly, electrophysiological experiments showed that BmKDfsin4,like scorpion potassium channel neurotoxins, could effectively inhibit Kv1.1, Kv1.2, and Kv1.3 channel currents, and its IC50value for the Kv1.3 channel was 510.2 nm Similar to the structure-function relationships of classical scorpion potassium channel-blocking toxins, basic residues (Lys-13 and Arg-19) of BmKDfsin4 play critical roles in peptide-Kv1.3 channel interactions. Furthermore, mutagenesis and electrophysiological experiments demonstrated that the channel extracellular pore region is the binding site of BmKDfsin4, indicating that BmKDfsin4 adopts the same mechanism for blocking potassium channel currents as classical scorpion toxins. Taken together, our work identifies scorpion BmKDfsin4 as the first invertebrate defensin to block potassium channels. These findings not only demonstrate that defensins from invertebrate animals are a novel type of potassium channel blockers but also provide evidence of a functional link between defensins and neurotoxins.


Assuntos
Antibacterianos/química , Defensinas/química , Neurotoxinas/química , Bloqueadores dos Canais de Potássio/química , Venenos de Escorpião/química , Sequência de Aminoácidos , Animais , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/crescimento & desenvolvimento , Defensinas/genética , Defensinas/metabolismo , Defensinas/farmacologia , Expressão Gênica , Humanos , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.2/antagonistas & inibidores , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/metabolismo , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Camundongos , Micrococcus luteus/efeitos dos fármacos , Micrococcus luteus/crescimento & desenvolvimento , Modelos Moleculares , Dados de Sequência Molecular , Neurotoxinas/genética , Neurotoxinas/metabolismo , Neurotoxinas/farmacologia , Bloqueadores dos Canais de Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Venenos de Escorpião/biossíntese , Escorpiões/química , Escorpiões/fisiologia , Alinhamento de Sequência , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Homologia Estrutural de Proteína , Relação Estrutura-Atividade
13.
J Cell Physiol ; 231(6): 1375-84, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26580832

RESUMO

Voltage-gated potassium Kv1.2 channels play pivotal role in maintaining of resting membrane potential and, consequently, regulation of cellular excitability of neurons. Endogenously generated electric field (EF) have been proven as an important regulator for cell migration and tissue repair. The mechanisms of ion channel involvement in EF-induced cell responses are extensively studied but largely are poorly understood. In this study we generated three COS-7 clones with different expression levels of Kv1.2 channel, and confirmed their functional variations with patch clamp analysis. Time-lapse imaging analysis showed that EF-induced cell migration response was Kv1.2 channel expression level depended. Inhibition of Kv1.2 channels with charybdotoxin (ChTX) constrained the sensitivity of COS-7 cells to EF stimulation more than their motility. Immunocytochemistry and pull-down analyses demonstrated association of Kv1.2 channels with actin-binding protein cortactin and its re-localization to the cathode-facing membrane at EF stimulation, which confirms the mechanism of EF-induced directional migration. This study displays that Kv1.2 channels represent an important physiological link in EF-induced cell migration. The described mechanism suggests a potential application of EF which may improve therapeutic performance in curing injuries of neuronal and/or cardiac tissue repair, post operational therapy, and various degenerative syndromes.


Assuntos
Movimento Celular , Canal de Potássio Kv1.1/metabolismo , Animais , Células COS , Movimento Celular/efeitos dos fármacos , Chlorocebus aethiops , Cortactina/metabolismo , Relação Dose-Resposta a Droga , Estimulação Elétrica , Imunoprecipitação , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/genética , Potenciais da Membrana , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Ligação Proteica , Transdução de Sinais , Fatores de Tempo , Imagem com Lapso de Tempo , Transfecção
14.
J Med Chem ; 58(17): 6784-802, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26288216

RESUMO

To realize the medicinal potential of peptide toxins, naturally occurring disulfide-rich peptides, as ion channel antagonists, more efficient pharmaceutical optimization technologies must be developed. Here, we show that the therapeutic properties of multiple cysteine toxin peptides can be rapidly and substantially improved by combining direct chemical strategies with high-throughput electrophysiology. We applied whole-molecule, brute-force, structure-activity analoging to ShK, a peptide toxin from the sea anemone Stichodactyla helianthus that inhibits the voltage-gated potassium ion channel Kv1.3, to effectively discover critical structural changes for 15× selectivity against the closely related neuronal ion channel Kv1.1. Subsequent site-specific polymer conjugation resulted in an exquisitely selective Kv1.3 antagonist (>1000× over Kv1.1) with picomolar functional activity in whole blood and a pharmacokinetic profile suitable for weekly administration in primates. The pharmacological potential of the optimized toxin peptide was demonstrated by potent and sustained inhibition of cytokine secretion from T cells, a therapeutic target for autoimmune diseases, in cynomolgus monkeys.


Assuntos
Venenos de Cnidários/química , Canal de Potássio Kv1.3/antagonistas & inibidores , Peptídeos/química , Polietilenoglicóis/química , Animais , Células CHO , Venenos de Cnidários/farmacocinética , Venenos de Cnidários/farmacologia , Cricetulus , Cristalografia por Raios X , Cães , Células HEK293 , Humanos , Interferon gama/sangue , Interferon gama/metabolismo , Interleucina-17/sangue , Interleucina-17/metabolismo , Interleucina-2/sangue , Interleucina-2/metabolismo , Canal de Potássio Kv1.1/antagonistas & inibidores , Macaca fascicularis , Masculino , Camundongos , Simulação de Acoplamento Molecular , Técnicas de Patch-Clamp , Peptídeos/farmacocinética , Peptídeos/farmacologia , Ratos Sprague-Dawley , Especificidade da Espécie , Estereoisomerismo , Relação Estrutura-Atividade , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
15.
J Biol Chem ; 290(19): 12195-209, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25792741

RESUMO

The lesser Asian scorpion Mesobuthus eupeus (Buthidae) is one of the most widely spread and dispersed species of the Mesobuthus genus, and its venom is actively studied. Nevertheless, a considerable amount of active compounds is still under-investigated due to the high complexity of this venom. Here, we report a comprehensive analysis of putative potassium channel toxins (KTxs) from the cDNA library of M. eupeus venom glands, and we compare the deduced KTx structures with peptides purified from the venom. For the transcriptome analysis, we used conventional tools as well as a search for structural motifs characteristic of scorpion venom components in the form of regular expressions. We found 59 candidate KTxs distributed in 30 subfamilies and presenting the cysteine-stabilized α/ß and inhibitor cystine knot types of fold. M. eupeus venom was then separated to individual components by multistage chromatography. A facile fluorescent system based on the expression of the KcsA-Kv1.1 hybrid channels in Escherichia coli and utilization of a labeled scorpion toxin was elaborated and applied to follow Kv1.1 pore binding activity during venom separation. As a result, eight high affinity Kv1.1 channel blockers were identified, including five novel peptides, which extend the panel of potential pharmacologically important Kv1 ligands. Activity of the new peptides against rat Kv1.1 channel was confirmed (IC50 in the range of 1-780 nm) by the two-electrode voltage clamp technique using a standard Xenopus oocyte system. Our integrated approach is of general utility and efficiency to mine natural venoms for KTxs.


Assuntos
Canal de Potássio Kv1.1/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/química , Venenos de Escorpião/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Cromatografia , Escherichia coli/metabolismo , Feminino , Corantes Fluorescentes/química , Biblioteca Gênica , Concentração Inibidora 50 , Ligantes , Espectrometria de Massas , Dados de Sequência Molecular , Oócitos , Filogenia , Proteoma , Ratos , Escorpiões , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Transcriptoma , Xenopus
16.
Mar Drugs ; 13(1): 529-42, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25603346

RESUMO

ShK, from the sea anemone Stichodactyla helianthus, is a 35-residue disulfide-rich peptide that blocks the voltage-gated potassium channel Kv1.3 at ca. 10 pM and the related channel Kv1.1 at ca. 16 pM. We developed an analog of this peptide, ShK-186, which is currently in Phase 1b-2a clinical trials for the treatment of autoimmune diseases such as multiple sclerosis and rheumatoid arthritis. While ShK-186 displays a >100-fold improvement in selectivity for Kv1.3 over Kv1.1 compared with ShK, there is considerable interest in developing peptides with an even greater selectivity ratio. In this report, we describe several variants of ShK that incorporate p-phophono-phenylalanine at the N-terminus coupled with internal substitutions at Gln16 and Met21. In addition, we also explored the combinatorial effects of these internal substitutions with an alanine extension at the C-terminus. Their selectivity was determined by patch-clamp electrophysiology on Kv1.3 and Kv1.1 channels stably expressed in mouse fibroblasts. The peptides with an alanine extension blocked Kv1.3 at low pM concentrations and exhibited up to 2250-fold selectivity for Kv1.3 over Kv1.1. Analogs that incorporates p-phosphono-phenylalanine at the N-terminus blocked Kv1.3 with IC50s in the low pM range and did not affect Kv1.1 at concentrations up to 100 nM, displaying a selectivity enhancement of >10,000-fold for Kv1.3 over Kv1.1. Other potentially important Kv channels such as Kv1.4 and Kv1.6 were only partially blocked at 100 nM concentrations of each of the ShK analogs.


Assuntos
Canal de Potássio Kv1.3/antagonistas & inibidores , Peptídeos/farmacologia , Anêmonas-do-Mar/química , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Cromatografia de Fase Reversa , Relação Dose-Resposta a Droga , Concentração Inibidora 50 , Canal de Potássio Kv1.1/antagonistas & inibidores , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Peptídeos/genética , Peptídeos/isolamento & purificação , Anêmonas-do-Mar/genética
17.
Eur J Neurosci ; 41(3): 293-304, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25394682

RESUMO

Synaptic transmission usually depends on action potentials (APs) in an all-or-none (digital) fashion. Recent studies indicate, however, that subthreshold presynaptic depolarization may facilitate spike-evoked transmission, thus creating an analog modulation of spike-evoked synaptic transmission, also called analog-digital (AD) synaptic facilitation. Yet, the underlying mechanisms behind this facilitation remain unclear. We show here that AD facilitation at rat CA3-CA3 synapses is time-dependent and requires long presynaptic depolarization (5-10 s) for its induction. This depolarization-induced AD facilitation (d-ADF) is blocked by the specific Kv1.1 channel blocker dendrotoxin-K. Using fast voltage-imaging of the axon, we show that somatic depolarization used for induction of d-ADF broadened the AP in the axon through inactivation of Kv1.1 channels. Somatic depolarization enhanced spike-evoked calcium signals in presynaptic terminals, but not basal calcium. In conclusion, axonal Kv1.1 channels determine glutamate release in CA3 neurons in a time-dependent manner through the control of the presynaptic spike waveform.


Assuntos
Potenciais de Ação/fisiologia , Região CA3 Hipocampal/fisiologia , Canal de Potássio Kv1.1/metabolismo , Transmissão Sináptica/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Região CA3 Hipocampal/efeitos dos fármacos , Cálcio/metabolismo , Quelantes de Cálcio/farmacologia , Ácido Egtázico/farmacologia , Ácido Glutâmico/metabolismo , Canal de Potássio Kv1.1/antagonistas & inibidores , Modelos Neurológicos , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Ratos Wistar , Sódio/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Tempo , Técnicas de Cultura de Tecidos
18.
Sci Rep ; 4: 4509, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24676092

RESUMO

HsTX1 toxin, from the scorpion Heterometrus spinnifer, is a 34-residue, C-terminally amidated peptide cross-linked by four disulfide bridges. Here we describe new HsTX1 analogues with an Ala, Phe, Val or Abu substitution at position 14. Complexes of HsTX1 with the voltage-gated potassium channels Kv1.3 and Kv1.1 were created using docking and molecular dynamics simulations, then umbrella sampling simulations were performed to construct the potential of mean force (PMF) of the ligand and calculate the corresponding binding free energy for the most stable configuration. The PMF method predicted that the R14A mutation in HsTX1 would yield a > 2 kcal/mol gain for the Kv1.3/Kv1.1 selectivity free energy relative to the wild-type peptide. Functional assays confirmed the predicted selectivity gain for HsTX1[R14A] and HsTX1[R14Abu], with an affinity for Kv1.3 in the low picomolar range and a selectivity of more than 2,000-fold for Kv1.3 over Kv1.1. This remarkable potency and selectivity for Kv1.3, which is significantly up-regulated in activated effector memory cells in humans, suggest that these analogues represent valuable leads in the development of therapeutics for autoimmune diseases.


Assuntos
Doenças Autoimunes/metabolismo , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Venenos de Escorpião/farmacologia , Sequência de Aminoácidos , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/imunologia , Linhagem Celular , Concentração Inibidora 50 , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/química , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.3/química , Ativação Linfocitária , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Bloqueadores dos Canais de Potássio/química , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Venenos de Escorpião/química
19.
Peptides ; 54: 9-18, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24418069

RESUMO

Spider venom contains a very valuable repertoire of natural resources to discover novel components for molecular diversity analyses and therapeutic applications. In this study, HWTX-XI toxins from the spider venom glands of Ornithoctonus huwena which are Kunitz-type toxins (KTTs) and were directly cloned, analyzed and functionally characterized. To date, the HWTX-XI superfamily consists of 38 members deduced from 121 high-quality expressed sequence tags, which is the largest spider KTT superfamily with significant molecular diversity mainly resulted from cDNA tandem repeats as well as focal hypermutation. Among them, HW11c40 and HW11c50 may be intermediate variants between native Kunitz toxins and sub-Kunitz toxins based on evolutionary analyses. In order to elucidate their biological activities, recombinant HW11c4, HW11c24, HW11c27 and HW11c39 were successfully expressed, further purified and functionally characterized. Both HW11c4 and HW11c27 display inhibitory activities against trypsin, chymotrypsin and kallikrein. Moreover, HW11c4 is also an inhibitor relatively specific for Kv1.1 channels. HW11c24 and HW11c39 are found to be inactive on chymotrysin, trypsin, kallikrein, thrombin and ion channels. These findings provide molecular evidence for toxin diversification of the HWTX-XI superfamily and useful molecular templates of serine protease inhibitors and ion channel blockers for the development of potentially clinical applications.


Assuntos
Inibidores de Serina Proteinase/farmacologia , Venenos de Aranha/metabolismo , Aranhas/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Evolução Molecular , Etiquetas de Sequências Expressas , Feminino , Canal de Potássio Kv1.1/antagonistas & inibidores , Dados de Sequência Molecular , Oócitos/efeitos dos fármacos , Filogenia , Isoformas de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Inibidores de Serina Proteinase/química , Venenos de Aranha/química , Venenos de Aranha/genética , Venenos de Aranha/isolamento & purificação , Venenos de Aranha/farmacologia , Aranhas/genética , Xenopus laevis
20.
J Neurophysiol ; 111(6): 1153-64, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24335214

RESUMO

Each neuron possesses a unique firing property, which is largely attributed to heterogeneity in the composition of voltage-gated ion channel complexes. Zebrafish Mauthner (M) cells, which are bilaterally paired giant reticulospinal neurons (RSNs) in the hindbrain and induce rapid escape behavior, generate only a single spike at the onset of depolarization. This single spiking is in contrast with the repetitive firing of the M cell's morphologically homologous RSNs, MiD2cm and MiD3cm, which are also involved in escapes. However, how the unique firing property of M cells is established and the underlying molecular mechanisms remain unclear. In the present study, we first demonstrated that the single-spiking property of M cells was acquired at 4 days postfertilization (dpf), accompanied by an increase in dendrotoxin I (DTX)-sensitive low-threshold K(+) currents, prior to which the M cell repetitively fires as its homologs. Second, in situ hybridization showed that among DTX-sensitive Kv1 channel α-subunits, zKv1.1a was unexpectedly expressed even in the homologs and the bursting M cells at 2 dpf. In contrast, zKvß2b, an auxiliary ß-subunit of Kv1 channels, was expressed only in the single-spiking M cells. Third, zKv1.1a expressed in Xenopus oocytes functioned as a low-threshold K(+) channel, and its currents were enhanced by coexpression of zKvß2b subunits. Finally, knockdown of zKvß2b expression in zebrafish larvae resulted in repetitive firing of M cells at 4 dpf. Taken together, these results suggest that associative expression of Kvß2 subunits with Kv1.1 channels is crucial for developmental acquisition of the unique firing properties of the M cells among homologous neurons.


Assuntos
Potenciais de Ação , Canal de Potássio Kv1.1/metabolismo , Neurônios/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Venenos Elapídicos/farmacologia , Canal de Potássio Kv1.1/antagonistas & inibidores , Canal de Potássio Kv1.1/genética , Neurônios/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Multimerização Proteica , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Rombencéfalo/citologia , Rombencéfalo/crescimento & desenvolvimento , Rombencéfalo/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...