Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Psychiatry Res ; 303: 114038, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34174581

RESUMO

An extensive catalog of common and rare genetic variants contributes to overall risk for schizophrenia and related disorders. As a complement to population genetics efforts, here we present whole genome sequences of multiple affected probands within individual families to search for possible high penetrance driver variants. From a total of 15 families diagnostically evaluated by a single research psychiatrist, we performed whole genome sequencing of a total of 61 affected individuals, called SNPs, indels, and copy number variants, and compared to reference genomes. In fourteen out of fifteen families, the schizophrenia polygenic risk score for each proband was within the control range defined by the Thousand Genomes cohort. In six families, each affected member carried a very rare or private, predicted-damaging, variant in at least one gene. Among these genes, variants in LRP1 and TENM2 suggest these are candidate disease-related genes when taken into context with existing population genetic studies and biological information. Results add to the number of pedigree sequences reported, suggest pathways for the investigation of biological mechanisms, and are consistent with the overall accumulating evidence that very rare damaging variants contribute to the heritability of schizophrenia.


Assuntos
Esquizofrenia , Variações do Número de Cópias de DNA/genética , Predisposição Genética para Doença/genética , Células Germinativas , Humanos , Linhagem , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/genética
2.
Bioorg Med Chem Lett ; 27(15): 3477-3485, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28629594

RESUMO

The voltage-gated sodium channel NaV1.7 has received much attention from the scientific community due to compelling human genetic data linking gain- and loss-of-function mutations to pain phenotypes. Despite this genetic validation of NaV1.7 as a target for pain, high quality pharmacological tools facilitate further understanding of target biology, establishment of target coverage requirements and subsequent progression into the clinic. Within the sulfonamide class of inhibitors, reduced potency on rat NaV1.7 versus human NaV1.7 was observed, rendering in vivo rat pharmacology studies challenging. Herein, we report the discovery and optimization of novel benzoxazine sulfonamide inhibitors of human, rat and mouse NaV1.7 which enabled pharmacological assessment in traditional behavioral rodent models of pain and in turn, established a connection between formalin-induced pain and histamine-induced pruritus in mice. The latter represents a simple and efficient means of measuring target engagement.


Assuntos
Benzoxazinas/química , Benzoxazinas/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Sulfonamidas/química , Sulfonamidas/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Analgésicos/química , Analgésicos/farmacocinética , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Benzoxazinas/farmacocinética , Benzoxazinas/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Dor/tratamento farmacológico , Dor/metabolismo , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapêutico , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacocinética , Bloqueadores do Canal de Sódio Disparado por Voltagem/uso terapêutico
3.
Nat Struct Mol Biol ; 24(2): 108-113, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27991902

RESUMO

Current therapies to treat persistent pain and neuropathic pain are limited by poor efficacy, side effects and risk of addiction. Here, we present a novel class of potent selective, central nervous system (CNS)-penetrant potentiators of glycine receptors (GlyRs), ligand-gated ion channels expressed in the CNS. AM-1488 increased the response to exogenous glycine in mouse spinal cord and significantly reversed mechanical allodynia induced by nerve injury in a mouse model of neuropathic pain. We obtained an X-ray crystal structure of human homopentameric GlyRα3 in complex with AM-3607, a potentiator of the same class with increased potency, and the agonist glycine, at 2.6-Å resolution. AM-3607 binds a novel allosteric site between subunits, which is adjacent to the orthosteric site where glycine binds. Our results provide new insights into the potentiation of cysteine-loop receptors by positive allosteric modulators and hold promise in structure-based design of GlyR modulators for the treatment of neuropathic pain.


Assuntos
Receptores de Glicina/química , Regulação Alostérica , Sítios de Ligação , Ligação Competitiva , Cristalografia por Raios X , Glicina/química , Células HEK293 , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Domínios Proteicos , Subunidades Proteicas/química
4.
J Med Chem ; 59(6): 2704-17, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-26890998

RESUMO

There is interest in the identification and optimization of new molecular entities selectively targeting ion channels of therapeutic relevance. Peptide toxins represent a rich source of pharmacology for ion channels, and we recently reported GpTx-1 analogs that inhibit NaV1.7, a voltage-gated sodium ion channel that is a compelling target for improved treatment of pain. Here we utilize multi-attribute positional scan (MAPS) analoging, combining high-throughput synthesis and electrophysiology, to interrogate the interaction of GpTx-1 with NaV1.7 and related NaV subtypes. After one round of MAPS analoging, we found novel substitutions at multiple residue positions not previously identified, specifically glutamic acid at positions 10 or 11 or lysine at position 18, that produce peptides with single digit nanomolar potency on NaV1.7 and 500-fold selectivity against off-target sodium channels. Docking studies with a NaV1.7 homology model and peptide NMR structure generated a model consistent with the key potency and selectivity modifications mapped in this work.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Peptídeos/farmacologia , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/farmacologia , Venenos de Aranha/farmacologia , Sequência de Aminoácidos , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Especificidade por Substrato
5.
PLoS One ; 10(9): e0138140, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26379236

RESUMO

While genetic evidence shows that the Nav1.7 voltage-gated sodium ion channel is a key regulator of pain, it is unclear exactly how Nav1.7 governs neuronal firing and what biophysical, physiological, and distribution properties of a pharmacological Nav1.7 inhibitor are required to produce analgesia. Here we characterize a series of aminotriazine inhibitors of Nav1.7 in vitro and in rodent models of pain and test the effects of the previously reported "compound 52" aminotriazine inhibitor on the spiking properties of nociceptors in vivo. Multiple aminotriazines, including some with low terminal brain to plasma concentration ratios, showed analgesic efficacy in the formalin model of pain. Effective concentrations were consistent with the in vitro potency as measured on partially-inactivated Nav1.7 but were far below concentrations required to inhibit non-inactivated Nav1.7. Compound 52 also reversed thermal hyperalgesia in the complete Freund's adjuvant (CFA) model of pain. To study neuronal mechanisms, electrophysiological recordings were made in vivo from single nociceptive fibers from the rat tibial nerve one day after CFA injection. Compound 52 reduced the spontaneous firing of C-fiber nociceptors from approximately 0.7 Hz to 0.2 Hz and decreased the number of action potentials evoked by suprathreshold tactile and heat stimuli. It did not, however, appreciably alter the C-fiber thresholds for response to tactile or thermal stimuli. Surprisingly, compound 52 did not affect spontaneous activity or evoked responses of Aδ-fiber nociceptors. Results suggest that inhibition of inactivated states of TTX-S channels, mostly likely Nav1.7, in the peripheral nervous system produces analgesia by regulating the spontaneous discharge of C-fiber nociceptors.


Assuntos
Analgésicos/uso terapêutico , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Fibras Nervosas Amielínicas/metabolismo , Nociceptores/metabolismo , Dor/tratamento farmacológico , Tetrodotoxina/farmacologia , Potenciais de Ação/fisiologia , Analgesia/métodos , Animais , Formaldeído/farmacologia , Adjuvante de Freund/farmacologia , Masculino , Dor/induzido quimicamente , Manejo da Dor , Medição da Dor/métodos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/farmacologia
6.
Bioorg Med Chem Lett ; 25(21): 4866-4871, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26112439

RESUMO

Many efforts are underway to develop selective inhibitors of the voltage-gated sodium channel NaV1.7 as new analgesics. Thus far, however, in vitro selectivity has proved difficult for small molecules, and peptides generally lack appropriate pharmacokinetic properties. We previously identified the NaV1.7 inhibitory peptide GpTx-1 from tarantula venom and optimized its potency and selectivity via structure-guided analoging. To further understand GpTx-1 binding to NaV1.7, we have mapped the binding site to transmembrane segments 1-4 of the second pseudosubunit internal repeat (commonly referred to as Site 4) using NaV1.5/NaV1.7 chimeric protein constructs. We also report that select GpTx-1 amino acid residues apparently not contacting NaV1.7 can be derivatized with a hydrophilic polymer without adversely affecting peptide potency. Homodimerization of GpTx-1 with a bifunctional polyethylene glycol (PEG) linker resulted in a compound with increased potency and a significantly reduced off-rate, demonstrating the ability to modulate the function and properties of GpTx-1 by linking to additional molecules.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Engenharia de Proteínas , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Dimerização , Relação Dose-Resposta a Droga , Humanos , Conformação Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Bloqueadores do Canal de Sódio Disparado por Voltagem/química
7.
J Med Chem ; 58(5): 2299-314, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25658507

RESUMO

NaV1.7 is a voltage-gated sodium ion channel implicated by human genetic evidence as a therapeutic target for the treatment of pain. Screening fractionated venom from the tarantula Grammostola porteri led to the identification of a 34-residue peptide, termed GpTx-1, with potent activity on NaV1.7 (IC50 = 10 nM) and promising selectivity against key NaV subtypes (20× and 1000× over NaV1.4 and NaV1.5, respectively). NMR structural analysis of the chemically synthesized three disulfide peptide was consistent with an inhibitory cystine knot motif. Alanine scanning of GpTx-1 revealed that residues Trp(29), Lys(31), and Phe(34) near the C-terminus are critical for potent NaV1.7 antagonist activity. Substitution of Ala for Phe at position 5 conferred 300-fold selectivity against NaV1.4. A structure-guided campaign afforded additive improvements in potency and NaV subtype selectivity, culminating in the design of [Ala5,Phe6,Leu26,Arg28]GpTx-1 with a NaV1.7 IC50 value of 1.6 nM and >1000× selectivity against NaV1.4 and NaV1.5.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/química , Fragmentos de Peptídeos/farmacologia , Venenos de Aranha/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Eletrofisiologia , Feminino , Ensaios de Triagem em Larga Escala , Humanos , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.7/sangue , Fragmentos de Peptídeos/química , Conformação Proteica , Ratos , Espectrometria de Massas por Ionização por Electrospray , Venenos de Aranha/química , Aranhas , Relação Estrutura-Atividade , Bloqueadores do Canal de Sódio Disparado por Voltagem/química
8.
PLoS One ; 9(9): e105895, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25188265

RESUMO

Clinical genetic studies have shown that loss of Nav1.7 function leads to the complete loss of acute pain perception. The global deletion is reported lethal in mice, however, and studies of mice with promoter-specific deletions of Nav1.7 have suggested that the role of Nav1.7 in pain transduction depends on the precise form of pain. We developed genetic and animal husbandry strategies that overcame the neonatal-lethal phenotype and enabled construction of a global Nav1.7 knockout mouse. Knockouts were anatomically normal, reached adulthood, and had phenotype wholly analogous to human congenital indifference to pain (CIP): compared to littermates, knockouts showed no defects in mechanical sensitivity or overall movement yet were completely insensitive to painful tactile, thermal, and chemical stimuli and were anosmic. Knockouts also showed no painful behaviors resulting from peripheral injection of nonselective sodium channel activators, did not develop complete Freund's adjuvant-induced thermal hyperalgesia, and were insensitive to intra-dermal histamine injection. Tetrodotoxin-sensitive sodium current recorded from cell bodies of isolated sensory neurons and the mechanically-evoked spiking of C-fibers in a skin-nerve preparation each were reduced but not eliminated in tissue from knockouts compared to littermates. Results support a role for Nav1.7 that is conserved between rodents and humans and suggest several possibly translatable biomarkers for the study of Nav1.7-targeted therapeutics. Results further suggest that Nav1.7 may retain its key role in persistent as well as acute forms of pain.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/deficiência , Insensibilidade Congênita à Dor/etiologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.7/fisiologia , Fibras Nervosas Amielínicas/fisiologia , Sistema Nervoso/patologia , Sistema Nervoso/fisiopatologia , Transtornos do Olfato/genética , Transtornos do Olfato/fisiopatologia , Insensibilidade Congênita à Dor/genética , Insensibilidade Congênita à Dor/fisiopatologia , Limiar da Dor/fisiologia , Fenótipo , Células Receptoras Sensoriais/fisiologia
9.
Bioorg Med Chem Lett ; 22(5): 2033-42, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22306122

RESUMO

Herein the discovery of a novel class of aminoheterocyclic Na(v)1.7 antagonists is reported. Hit compound 1 was potent but suffered from poor pharmacokinetics and selectivity. The compact structure of 1 offered a modular synthetic strategy towards a broad structure-activity relationship analysis. This analysis led to the identification of aminopyrazine 41, which had vastly improved hERG selectivity and pharmacokinetic properties.


Assuntos
Pirazinas/química , Pirazinas/farmacologia , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Aminas/química , Aminas/metabolismo , Aminas/farmacocinética , Aminas/farmacologia , Animais , Descoberta de Drogas , Concentração Inibidora 50 , Masculino , Canal de Sódio Disparado por Voltagem NAV1.7 , Plasma/metabolismo , Pirazinas/metabolismo , Pirazinas/farmacocinética , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacocinética , Relação Estrutura-Atividade
10.
Bioorg Med Chem Lett ; 22(5): 2052-62, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22318156

RESUMO

Herein we describe the discovery, optimization, and structure-activity relationships of novel potent pyrrolopyrimidine Na(v)1.7 antagonists. Hit-to-lead SAR studies of the pyrrolopyrimidine core, head, and tail groups of the molecule led to the identification of pyrrolopyrimidine 48 as exceptionally potent Na(v)1.7 blocker with good selectivity over hERG and improved microsomal stability relative to our hit molecule and pyrazolopyrimidine 8 as a promising starting point for future optimization efforts.


Assuntos
Pirimidinas/química , Pirimidinas/farmacologia , Pirróis/química , Pirróis/farmacologia , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Descoberta de Drogas , Humanos , Microssomos Hepáticos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7 , Dor/tratamento farmacológico , Pirimidinas/metabolismo , Pirróis/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Relação Estrutura-Atividade
11.
Bioorg Med Chem Lett ; 22(2): 1055-60, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22209205

RESUMO

Clinical genetic data have shown that the product of the SCN9A gene, voltage-gated sodium ion channel Nav1.7, is a key control point for pain perception and a possible target for a next generation of analgesics. Sodium channels, however, historically have been difficult drug targets, and many of the existing structure-activity relationships (SAR) have been defined on pharmacologically modified channels with indirect reporter assays. Herein we describe the discovery, optimization, and SAR of potent aminopyrimidinone Nav1.7 antagonists using electrophysiology-based assays that measure the ligand-receptor interaction directly. Within this series, rapid functionalization at the polysubstituted aminopyrimidinone head group enabled exploration of SAR and of pharmacokinetic properties. Lead optimized N-Me-aminopyrimidinone 9 exhibited improved Nav1.7 potency, minimal off-target hERG liability, and improved rat PK properties.


Assuntos
Pirimidinonas/farmacologia , Canais de Sódio/metabolismo , Animais , Relação Dose-Resposta a Droga , Humanos , Ligantes , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Canal de Sódio Disparado por Voltagem NAV1.7 , Pirimidinonas/síntese química , Pirimidinonas/química , Ratos , Relação Estrutura-Atividade
12.
J Med Chem ; 54(13): 4427-45, 2011 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-21634377

RESUMO

Clinical human genetic studies have recently identified the tetrodotoxin (TTX) sensitive neuronal voltage gated sodium channel Nav1.7 (SCN9A) as a critical mediator of pain sensitization. Herein, we report structure-activity relationships for a novel series of 2,4-diaminotriazines that inhibit hNav1.7. Optimization efforts culminated in compound 52, which demonstrated pharmacokinetic properties appropriate for in vivo testing in rats. The binding site of compound 52 on Nav1.7 was determined to be distinct from that of local anesthetics. Compound 52 inhibited tetrodotoxin-sensitive sodium channels recorded from rat sensory neurons and exhibited modest selectivity against the hERG potassium channel and against cloned and native tetrodotoxin-resistant sodium channels. Upon oral administration to rats, compound 52 produced dose- and exposure-dependent efficacy in the formalin model of pain.


Assuntos
Acetamidas/síntese química , Analgésicos/síntese química , Proteínas do Tecido Nervoso/antagonistas & inibidores , Dor/tratamento farmacológico , Bloqueadores dos Canais de Sódio/síntese química , Triazinas/síntese química , Acetamidas/farmacocinética , Acetamidas/farmacologia , Administração Oral , Analgésicos/farmacocinética , Analgésicos/farmacologia , Animais , Sítios de Ligação , Linhagem Celular , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Formaldeído , Gânglios Espinais/citologia , Humanos , Técnicas In Vitro , Microssomos Hepáticos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.1 , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Medição da Dor , Técnicas de Patch-Clamp , Ratos , Bloqueadores dos Canais de Sódio/farmacocinética , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio , Solubilidade , Relação Estrutura-Atividade , Tetrodotoxina/farmacologia , Triazinas/farmacocinética , Triazinas/farmacologia
13.
Pain ; 149(1): 33-49, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20167427

RESUMO

Nicotinic acetylcholine receptors (nAChRs) are longstanding targets for a next generation of pain therapeutics, but the nAChR subtypes that govern analgesia remain unknown. We tested a series of nicotinic agonists, including many molecules used or tried clinically, on a panel of cloned neuronal nAChRs for potency and selectivity using patch-clamp electrophysiology and a live cell-based fluorescence assay. Nonselective nicotinic agonists as well as compounds selective either for alpha4beta2 or for alpha7 nAChRs were then tested in the formalin and complete Freund's adjuvant models of pain. Nonselective nAChR agonists ABT-594 and varenicline were effective analgesics. By contrast, the selective alpha4beta2 agonist ispronicline and a novel alpha4beta2-selective potentiator did not appear to produce analgesia in either model. alpha7-selective agonists reduced the pain-related endpoint, but the effect could be ascribed to nonspecific reduction of movement rather than to analgesia. Neither selective nor nonselective alpha7 nicotinic agonists affected the release of pro-inflammatory cytokines in response to antigen challenge. Electrophysiological recordings from spinal cord slice showed a strong nicotine-induced increase in inhibitory synaptic transmission that was mediated partially by alpha4beta2 and only minimally by alpha7 subtypes. Taken with previous studies, the results suggest that agonism of alpha4beta2 nAChRs is necessary but not sufficient to produce analgesia, and that the spinal cord is a key site where the molecular action of nAChRs produces analgesia.


Assuntos
Analgésicos/administração & dosagem , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Agonistas Nicotínicos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Animais , Doença Crônica , Humanos , Hiperalgesia/diagnóstico , Masculino , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
14.
Bioorg Med Chem Lett ; 18(19): 5209-12, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18789861

RESUMO

The discovery of a series of small molecule alpha4beta2 nAChR potentiators is reported. The structure-activity relationship leads to potent compounds selective against nAChRs including alpha3beta2 and alpha3beta4 and optimized for CNS penetrance. Compounds increased currents through recombinant alpha4beta2 nAChRs, yet did not compete for binding with the orthosteric ligand cytisine. High potency and efficacy on the rat channel combined with good PK properties will allow testing of the alpha4beta2 potentiator mechanism in animal models of disease.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Agonistas Nicotínicos/farmacologia , Piperidinas/síntese química , Piperidinas/farmacologia , Receptores Nicotínicos/efeitos dos fármacos , Animais , Técnicas de Química Combinatória , Modelos Animais de Doenças , Humanos , Estrutura Molecular , Piperidinas/química , Ratos , Receptores Nicotínicos/química , Relação Estrutura-Atividade
15.
Curr Pharm Des ; 14(24): 2480-91, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18781996

RESUMO

Many peptides are potent and highly selective blockers or modulators of calcium channel function, and as such are valuable pharmacological tools and potentially valuable leads for the development of human therapeutics. Cone shells and spiders are rich sources of such peptides, although they are also found in scorpions and insects. In this article we compare the amino acid sequences of toxins active against calcium channels and describe their three-dimensional structures and structure-function relationships. Certain structural motifs, in particular the inhibitor cystine knot, prove to be quite common amongst this class of toxins. Aspects of the pharmacology and physiology of these toxins in mammalian systems are also discussed, with an emphasis on their application in the treatment of chronic pain. We then consider the prospects for peptide-based therapeutics targeting calcium channels for this and other indications, including the development of non-peptide (peptidomimetic) compounds based on a detailed understanding of toxin structure-function relationships.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Desenho de Fármacos , Peptídeos/farmacologia , Peçonhas/química , Sequência de Aminoácidos , Animais , Bloqueadores dos Canais de Cálcio/isolamento & purificação , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio/química , Canais de Cálcio/fisiologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/isolamento & purificação , Peptídeos/uso terapêutico , Conformação Proteica , Alinhamento de Sequência , Relação Estrutura-Atividade
16.
Bioorg Med Chem Lett ; 18(20): 5643-7, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18805006

RESUMO

The synthesis and structure-activity relationship of a series of carbamate potentiators of alpha4beta2 nAChR is reported herein. These compounds were highly selective for alpha4beta2 over other nAChR subtypes. In addition, compounds increased the response of alpha4beta2 nAChRs to acetylcholine, as measured with patch-clamp electrophysiology.


Assuntos
Química Farmacêutica/métodos , Receptores Nicotínicos/química , Acetilcolina/química , Cálcio/química , Carbamatos/química , Desenho de Fármacos , Humanos , Modelos Químicos , Sistema Nervoso/metabolismo , Neurônios/metabolismo , Técnicas de Patch-Clamp , Pirazóis/química , Piridinas/química , Receptores Nicotínicos/metabolismo , Relação Estrutura-Atividade
17.
Toxicon ; 49(2): 202-12, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17097124

RESUMO

Some of the most potent and specific inhibitors of voltage-gated calcium channels are peptide toxins that inhibit channel function not by occlusion of the channel pore, but rather by interfering with the voltage dependence and kinetics of channel opening and closing. Many such gating modifier toxins conform to the inhibitor cystine knot structural family and have primary sequence or functional mechanism similar to toxins that target voltage-gated sodium or potassium channels. This review introduces known gating modifiers of calcium channels, discusses the selectivity, binding sites, and mechanism of the toxin-channel interaction, and reviews the usefulness of these toxins as research tools and as the basis for novel calcium channel pharmacology and therapeutics.


Assuntos
Canais de Cálcio/farmacologia , Ativação do Canal Iônico , Peçonhas/genética , Peçonhas/farmacologia , Sequência de Aminoácidos , Animais , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Dados de Sequência Molecular , Alinhamento de Sequência
18.
Biophys J ; 88(1): 211-23, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15501945

RESUMO

FPL 64176 (FPL) is a nondihydropyridine compound that dramatically increases macroscopic inward current through L-type calcium channels and slows activation and deactivation. To understand the mechanism by which channel behavior is altered, we compared the effects of the drug on the kinetics and voltage dependence of ionic currents and gating currents. Currents from a homogeneous population of channels were obtained using cloned rabbit Ca(V)1.2 (alpha1C, cardiac L-type) channels stably expressed in baby hamster kidney cells together with beta1a and alpha2delta1 subunits. We found a striking dissociation between effects of FPL on ionic currents, which were modified strongly, and on gating currents, which were not detectably altered. Inward ionic currents were enhanced approximately 5-fold for a voltage step from -90 mV to +10 mV. Kinetics of activation and deactivation were slowed dramatically at most voltages. Curiously, however, at very hyperpolarized voltages (< -250 mV), deactivation was actually faster in FPL than in control. Gating currents were measured using a variety of inorganic ions to block ionic current and also without blockers, by recording gating current at the reversal potential for ionic current (+50 mV). Despite the slowed kinetics of ionic currents, FPL had no discernible effect on the fundamental movements of gating charge that drive channel gating. Instead, FPL somehow affects the coupling of charge movement to opening and closing of the pore. An intriguing possibility is that the drug causes an inactivated state to become conducting without otherwise affecting gating transitions.


Assuntos
Biofísica/métodos , Agonistas dos Canais de Cálcio/farmacologia , Íons , Pirróis/farmacologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Canais de Cálcio/química , Linhagem Celular , Césio/química , Cricetinae , Eletrofisiologia , Ativação do Canal Iônico , Cinética , Potenciais da Membrana , Modelos Teóricos , Plasmídeos/metabolismo , Canais de Potássio/química , Coelhos , Fatores de Tempo , Transfecção
19.
Curr Drug Targets CNS Neurol Disord ; 3(6): 457-78, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15578964

RESUMO

This review focuses on the importance of voltage-gated calcium channels in modulating and controlling the function of peripheral and central neurons involved in nociceptive processing. We describe the different families of voltage-gated calcium channels that are expressed in pain pathway neurons, how the expression levels of calcium channel currents change in chronic pain conditions, and the validation of N-type, T-type, and P-type calcium channels as targets for the treatment of pain. The molecular mechanism of action is reviewed for the most prominent calcium channel-targeted drugs including gabapentin and ziconotide as well as antiepileptics administered off-label for the treatment of pain. We discuss how the major genetic, functional, and pharmacological differences between subtypes of neuronal calcium channels can be leveraged to identify new molecular targets and to discover and develop new therapeutic agents for the treatment of chronic pain syndromes.


Assuntos
Vias Aferentes/fisiopatologia , Canais de Cálcio/genética , Neurônios Aferentes/fisiologia , Nociceptores/fisiologia , Dor Intratável/fisiopatologia , Vias Aferentes/efeitos dos fármacos , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio/efeitos dos fármacos , Desenho de Fármacos , Humanos , Neurônios Aferentes/efeitos dos fármacos , Nociceptores/efeitos dos fármacos , Dor Intratável/tratamento farmacológico
20.
J Neurophysiol ; 92(5): 2820-30, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15201306

RESUMO

The CaV2.2 gene encodes the functional core of the N-type calcium channel. This gene has the potential to generate thousands of CaV2.2 splice isoforms with different properties. However, the functional significance of most sites of alternative splicing is not established. The IVS3-IVS4 region contains an alternative splice site that is conserved evolutionarily among CaValpha1 genes from Drosophila to human. In CaV2.2, inclusion of exon 31a in the IVS3-IVS4 region is restricted to the peripheral nervous system, and its inclusion slows the speed of channel activation. To investigate the effects of exon 31a in more detail, we generated four tsA201 cell lines stably expressing CaV2.2 splice isoforms. Coexpression of auxiliary CaVbeta and CaValpha2delta subunits was required to reconstitute currents with the kinetics of N-type channels from neurons. Channels including exon 31a activated and deactivated more slowly at all voltages. Current densities were high enough in the stable cell lines co-expressing CaValpha2delta to resolve gating currents. The steady-state voltage dependence of charge movement was not consistently different between splice isoforms, but on gating currents from the exon 31a-containing CaV2.2 isoform decayed with a slower time course, corresponding to slower movement of the charge sensor. Exon 31a-containing CaV2.2 is restricted to peripheral ganglia; and the slower gating kinetics of CaV2.2 splice isoforms containing exon 31a correlated reasonably well with the properties of native N-type currents in sympathetic neurons. Our results suggest that alternative splicing in the S3-S4 linker influences the kinetics but not the voltage dependence of N-type channel gating.


Assuntos
Processamento Alternativo/genética , Canais de Cálcio Tipo N/genética , Animais , Canais de Cálcio Tipo N/fisiologia , Linhagem Celular , Drosophila/genética , Humanos , Ativação do Canal Iônico , Potenciais da Membrana , Subunidades Proteicas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...