Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
Proc Natl Acad Sci U S A ; 116(3): 1059-1064, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30593566

RESUMO

The vast complexity of native heteromeric K+ channels is largely unexplored. Defining the composition and subunit arrangement of individual subunits in native heteromeric K+ channels and establishing their physiological roles is experimentally challenging. Here we systematically explored this "zone of ignorance" in molecular neuroscience. Venom components, such as peptide toxins, appear to have evolved to modulate physiologically relevant targets by discriminating among closely related native ion channel complexes. We provide proof-of-principle for this assertion by demonstrating that κM-conotoxin RIIIJ (κM-RIIIJ) from Conus radiatus precisely targets "asymmetric" Kv channels composed of three Kv1.2 subunits and one Kv1.1 or Kv1.6 subunit with 100-fold higher apparent affinity compared with homomeric Kv1.2 channels. Our study shows that dorsal root ganglion (DRG) neurons contain at least two major functional Kv1.2 channel complexes: a heteromer, for which κM-RIIIJ has high affinity, and a putative Kv1.2 homomer, toward which κM-RIIIJ is less potent. This conclusion was reached by (i) covalent linkage of members of the mammalian Shaker-related Kv1 family to Kv1.2 and systematic assessment of the potency of κM-RIIIJ block of heteromeric K+ channel-mediated currents in heterologous expression systems; (ii) molecular dynamics simulations of asymmetric Kv1 channels providing insights into the molecular basis of κM-RIIIJ selectivity and potency toward its targets; and (iii) evaluation of calcium responses of a defined population of DRG neurons to κM-RIIIJ. Our study demonstrates that bioactive molecules present in venoms provide essential pharmacological tools that systematically target specific heteromeric Kv channel complexes that operate in native tissues.


Assuntos
Conotoxinas , Gânglios Espinais , Potenciais da Membrana , Simulação de Dinâmica Molecular , Neurônios , Superfamília Shaker de Canais de Potássio , Conotoxinas/química , Conotoxinas/metabolismo , Gânglios Espinais/química , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Transporte de Íons , Neurônios/química , Neurônios/metabolismo , Ligação Proteica , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Superfamília Shaker de Canais de Potássio/química , Superfamília Shaker de Canais de Potássio/metabolismo
3.
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
4.
Toxicon ; 89: 45-54, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24997406

RESUMO

The toxinology of the crassispirine snails, a major group of venomous marine gastropods within the superfamily Conoidea, is largely unknown. Here we define the first venom peptide superfamily, the P-like crassipeptides, and show that the organization of their gene sequences is similar to conotoxin precursors. We provide evidence that one peptide family within the P-like crassipeptide superfamily includes potassium-channel (K-channel) blockers, the κP-crassipeptides. Three of these peptides were chemically synthesized (cce9a, cce9b and iqi9a). Using conventional electrophysiology, cce9b was shown to be an antagonist of both a human Kv1.1 channel isoform (Shaker subfamily of voltage-gated K channels) and a Drosophila K-channel isoform. We assessed the bioactivity of these peptides in native mammalian dorsal root ganglion neurons in culture. We demonstrate that two of these crassipeptides, cce9a and cce9b, elicited an excitatory phenotype in a subset of small-diameter capsaicin-sensitive mouse DRG neurons that were also affected by κJ-conotoxin PlXIVA (pl14a), a blocker of Kv1.6 channels. Given the vast complexity of heteromeric K-channel isoforms, this study demonstrates that the crassispirine venoms are a potentially rich source for discovering novel peptides that can help to identify and characterize the diversity of K-channel subtypes expressed in native neurons and other cell types.


Assuntos
Venenos de Moluscos/química , Peptídeos/química , Caramujos/química , Animais , Clonagem Molecular , Drosophila , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Venenos de Moluscos/isolamento & purificação , Venenos de Moluscos/toxicidade , Peptídeos/isolamento & purificação , Peptídeos/toxicidade , Filogenia , Canais de Potássio/química , Caramujos/genética , Xenopus
5.
Curr Opin Pharmacol ; 15: 115-21, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24632326

RESUMO

Voltage activated potassium (Kv) channels are intensely investigated targets within the pharmacological strategies to treat cardiac arrhythmia. For atrial fibrillation (AF) substances inhibiting the ultra rapid outward rectifying Kv current (IKur) and its underlying Kv1.5 channel have been developed. Here we describe potential limitations of this approach with respect to critical parameters of Kv channel pharmacology. In healthy tissue IKur/Kv1.5 inhibition can unexpectedly lead to action potential shortening with corresponding arrhythmogenic effects. In tissue with chronic AF, electrical remodeling occurs which is accompanied with changes in ion channel expression and composition. As a consequence atrial tissue exhibits a different pharmacological fingerprint. New strategies to obtain more mechanistic insight into drug target interaction are needed for better understanding the pharmacological potential of IKur/Kv1.5 inhibition for AF treatment.


Assuntos
Canal de Potássio Kv1.5/antagonistas & inibidores , Animais , Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/metabolismo , Humanos , Canal de Potássio Kv1.5/metabolismo
6.
EMBO Mol Med ; 4(5): 424-34, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438204

RESUMO

Glucose-stimulated insulin secretion (GSIS) relies on repetitive, electrical spiking activity of the beta cell membrane. Cyclic activation of voltage-gated potassium channels (K(v) ) generates an outward, 'delayed rectifier' potassium current, which drives the repolarizing phase of each spike and modulates insulin release. Although several K(v) channels are expressed in pancreatic islets, their individual contributions to GSIS remain incompletely understood. We take advantage of a naturally occurring cone-snail peptide toxin, Conkunitzin-S1 (Conk-S1), which selectively blocks K(v) 1.7 channels to provide an intrinsically limited, finely graded control of total beta cell delayed rectifier current and hence of GSIS. Conk-S1 increases GSIS in isolated rat islets, likely by reducing K(v) 1.7-mediated delayed rectifier currents in beta cells, which yields increases in action potential firing and cytoplasmic free calcium. In rats, Conk-S1 increases glucose-dependent insulin secretion without decreasing basal glucose. Thus, we conclude that K(v) 1.7 contributes to the membrane-repolarizing current of beta cells during GSIS and that block of this specific component of beta cell K(v) current offers a potential strategy for enhancing GSIS with minimal risk of hypoglycaemia during metabolic disorders such as Type 2 diabetes.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Potássio/metabolismo , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Animais , Secreções Corporais , Humanos , Secreção de Insulina , Masculino , Venenos de Moluscos/toxicidade , Ratos , Ratos Wistar
7.
Cardiovasc Res ; 93(1): 50-9, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21972180

RESUMO

AIMS: Cardiovascular research requires complex and functionally intact experimental models. Due to major differences in the cellular and subcellular composition of the myocardium between species, the use of human heart tissue is highly desirable. To enhance the experimental use of the human myocardium, we established methods for the preparation of vital tissue slices from the adult ventricular myocardium as well as conditions for their long-term preservation in organotypic culture. METHODS AND RESULTS: Human ventricular heart samples were derived from surgical specimens excised during a therapeutic Morrow myectomy and cut into 300 µm thick slices. Slices were either characterized in acute experiments or cultured at a liquid-air interface. Viability and functionality were proven by viability staining, enzyme activity tests, intracellular potential recordings, and force measurements. Precision-cut slices showed high viability throughout 28 days in culture and displayed typical cardiomyocyte action potential characteristics, which enabled pharmacological safety testing on the rapid component of the delayed rectifier potassium current (I(Kr)) and ATP-dependent potassium channels throughout the whole culture period. Constant expression of major ion channels was confirmed by quantitative PCR. Acute slices developed excitation-dependent contractions with a clear preload dependency and a ß-adrenergic response. Contractility and myosin light chain expression decreased during the first days in culture but reached a steady state with reactivity upon ß-adrenergic stimulation being preserved. CONCLUSION: Organotypic heart slices represent a multicellular model of the human myocardium and a novel platform for studies ranging from the investigation of molecular interactions to tissue engineering.


Assuntos
Ventrículos do Coração/anatomia & histologia , Modelos Cardiovasculares , Miocárdio/metabolismo , Técnicas de Cultura de Órgãos/métodos , Adulto , Fenômenos Fisiológicos Cardiovasculares , Fenômenos Eletrofisiológicos , Ventrículos do Coração/cirurgia , Humanos , Contração Miocárdica
8.
Cell Physiol Biochem ; 27(1): 1-12, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21325816

RESUMO

BACKGROUND/AIMS: Cellular models are an interesting tool to study human heart diseases. To date, research groups mainly focus on mouse models, but important murine physiology is different from human characteristics. Recently, scientists found that the electrophysiology of fish cardiomyocytes largely resembles that of humans. So far, cardiomyocyte models were generated using differentiation medium, were stimulated electrically or, when contracting spontaneously, only did so over a short time period. We established an in vitro spontaneously, long-term beating heart model generated from rainbow trout, with the potential to be used as a new human heart model system because of its electrophysiology. METHODS: Spontaneously contracting 3D cell layers from rainbow trout were generated in vitro and analyzed using PCR and immunochemistry. Further, electrophysiology was measured via intra - and extracellular recordings. RESULTS: Contracting cardiomyogenic aggregates were generated without differentiation medium and were beating autonomously for more than one month. Electrophysiological measurements exhibit that the action potential properties of fish cardiomyocytes in part resemble the characteristics of human cardiomyocytes. The sensitivity of the beating cell aggregates to drugs could also be confirmed. CONCLUSION: Spontaneously contracting cardiomyogenic cell aggregates from rainbow trout generated in vitro are suitable for human heart research and pharmacology.


Assuntos
Miócitos Cardíacos/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Antiarrítmicos/farmacologia , Biomarcadores/metabolismo , Células Cultivadas , Cromanos/farmacologia , Coração/fisiologia , Humanos , Isoproterenol/farmacologia , Modelos Biológicos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Oncorhynchus mykiss , Canais de Potássio/química , Canais de Potássio/metabolismo , Pirrolidinas/farmacologia , Receptores Adrenérgicos beta/química , Receptores Adrenérgicos beta/metabolismo
9.
J Biol Chem ; 285(20): 14882-14889, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20220134

RESUMO

Conus snail (Conus) venoms are a valuable source of pharmacologically active compounds; some of the peptide toxin families from the snail venoms are known to interact with potassium channels. We report the purification, synthesis, and characterization of kappaM-conotoxin RIIIJ from the venom of a fish-hunting species, Conus radiatus. This conopeptide, like a previously characterized peptide in the same family, kappaM-RIIIK, inhibits the homotetrameric human Kv1.2 channels. When tested in Xenopus oocytes, kappaM-RIIIJ has an order of magnitude higher affinity (IC(50) = 33 nm) to Kv1.2 than kappaM-RIIIK (IC(50) = 352 nm). Chimeras of RIIIK and RIIIJ tested on the human Kv1.2 channels revealed that Lys-9 from kappaM-RIIIJ is a determinant of its higher potency against hKv1.2. However, when compared in a model of ischemia/reperfusion, kappaM-RIIIK (100 mug/kg of body weight), administered just before reperfusion, significantly reduces the infarct size in rat hearts in vivo without influencing hemodynamics, providing a potential compound for cardioprotective therapeutics. In contrast, kappaM-RIIIJ does not exert any detectable cardioprotective effect. kappaM-RIIIJ shows more potency for Kv1.2-Kv1.5 and Kv1.2-Kv1.6 heterodimers than kappaM-RIIIK, whereas the affinity of kappaM-RIIIK to Kv1.2-Kv1.7 heterodimeric channels is higher (IC(50) = 680 nm) than that of kappaM-RIIIJ (IC(50) = 3.15 mum). Thus, the cardioprotection seems to correlate to antagonism to heteromultimeric channels, involving the Kv1.2 alpha-subunit rather than antagonism to Kv1.2 homotetramers. Furthermore, kappaM-RIIIK and kappaM-RIIIJ provide a valuable set of probes for understanding the underlying mechanism of cardioprotection.


Assuntos
Conotoxinas/farmacologia , Canal de Potássio Kv1.2/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Conotoxinas/química , Humanos , Masculino , Dados de Sequência Molecular , Ratos , Ratos Wistar , Espectrometria de Massas por Ionização por Electrospray
10.
J Biol Chem ; 283(34): 23026-32, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18505731

RESUMO

Two venom peptides, CPY-Pl1 (EU000528) and CPY-Fe1 (EU000529), characterized from the vermivorous marine snails Conus planorbis and Conus ferrugineus, define a new class of conopeptides, the conopeptide Y (CPY) family. The peptides have no disulfide cross-links and are 30 amino acids long; the high content of tyrosine is unprecedented for any native gene product. The CPY peptides were chemically synthesized and shown to be biologically active upon injection into both mice and Caenorhabditis elegans; activity on mammalian Kv1 channel isoforms was demonstrated using an oocyte heterologous expression system, and selectivity for Kv1.6 was found. NMR spectroscopy revealed that the peptides were unstructured in aqueous solution; however, a helical region including residues 12-18 for one peptide, CPY-Pl1, formed in trifluoroethanol buffer. Clones obtained from cDNA of both species encoded prepropeptide precursors that shared a unique signal sequence, indicating that these peptides are encoded by a novel gene family. This is the first report of tyrosine-rich bioactive peptides in Conus venom.


Assuntos
Peptídeos/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Tirosina/química , Sequência de Aminoácidos , Animais , Caramujo Conus , DNA Complementar/metabolismo , Canal de Potássio Kv1.6/química , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Venenos de Moluscos/metabolismo , Oócitos/metabolismo , Homologia de Sequência de Aminoácidos , Frações Subcelulares , Trifluoretanol/química
11.
Appl Microbiol Biotechnol ; 79(1): 1-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18340446

RESUMO

Cone snails are marine predators that use venoms to immobilize their prey. The venoms of these mollusks contain a cocktail of peptides that mainly target different voltage- and ligand-gated ion channels. Typically, conopeptides consist of ten to 30 amino acids but conopeptides with more than 60 amino acids have also been described. Due to their extraordinary pharmacological properties, conopeptides gained increasing interest in recent years. There are several conopeptides used in clinical trials and one peptide has received approval for the treatment of pain. Accordingly, there is an increasing need for the production of these peptides. So far, most individual conopeptides are synthesized using solid phase peptide synthesis. Here, we describe that at least some of these peptides can be obtained using prokaryotic or eukaryotic expression systems. This opens the possibility for biotechnological production of also larger amounts of long chain conopeptides for the use of these peptides in research and medical applications.


Assuntos
Conotoxinas/biossíntese , Conotoxinas/síntese química , Conotoxinas/farmacologia , Caramujo Conus/química , Analgésicos não Narcóticos/farmacologia , Animais , Canais de Cálcio Tipo N/metabolismo , Escherichia coli/metabolismo , Pichia/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/biossíntese , Relação Estrutura-Atividade , Especificidade por Substrato , ômega-Conotoxinas/farmacologia
12.
J Biol Chem ; 282(42): 30699-706, 2007 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17724025

RESUMO

Peptide neurotoxins from cone snails continue to supply compounds with therapeutic potential. Although several analgesic conotoxins have already reached human clinical trials, a continuing need exists for the discovery and development of novel non-opioid analgesics, such as subtype-selective sodium channel blockers. Micro-conotoxin KIIIA is representative of micro-conopeptides previously characterized as inhibitors of tetrodotoxin (TTX)-resistant sodium channels in amphibian dorsal root ganglion neurons. Here, we show that KIIIA has potent analgesic activity in the mouse pain model. Surprisingly, KIIIA was found to block most (>80%) of the TTX-sensitive, but only approximately 20% of the TTX-resistant, sodium current in mouse dorsal root ganglion neurons. KIIIA was tested on cloned mammalian channels expressed in Xenopus oocytes. Both Na(V)1.2 and Na(V)1.6 were strongly blocked; within experimental wash times of 40-60 min, block was reversed very little for Na(V)1.2 and only partially for Na(V)1.6. Other isoforms were blocked reversibly: Na(V)1.3 (IC50 8 microM), Na(V)1.5 (IC50 284 microM), and Na(V)1.4 (IC50 80 nM). "Alanine-walk" and related analogs were synthesized and tested against both Na(V)1.2 and Na(V)1.4; replacement of Trp-8 resulted in reversible block of Na(V)1.2, whereas replacement of Lys-7, Trp-8, or Asp-11 yielded a more profound effect on the block of Na(V)1.4 than of Na(V)1.2. Taken together, these data suggest that KIIIA is an effective tool to study structure and function of Na(V)1.2 and that further engineering of micro-conopeptides belonging to the KIIIA group may provide subtype-selective pharmacological compounds for mammalian neuronal sodium channels and potential therapeutics for the treatment of pain.


Assuntos
Analgésicos não Narcóticos/farmacologia , Conotoxinas/farmacologia , Neurônios/metabolismo , Dor/tratamento farmacológico , Peptídeos/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Substituição de Aminoácidos , Animais , Conotoxinas/genética , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Camundongos , Mutação de Sentido Incorreto , Neurônios/patologia , Oócitos , Dor/metabolismo , Dor/patologia , Peptídeos/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canais de Sódio/genética , Xenopus laevis
13.
Channels (Austin) ; 1(4): 253-62, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18698149

RESUMO

The muO-conotoxins MrVIA and MrVIB are 31-residue peptides from Conus marmoreus, belonging to the O-superfamily of conotoxins with three disulfide bridges. They have attracted attention because they are inhibitors of tetrodotoxin-insensitive voltage-gated sodium channels (Na(V)1.8) and could therefore serve as lead structure for novel analgesics. The aim of this study was to elucidate the molecular mechanism by which muO-conotoxins affect Na(V) channels. Rat Na(V)1.4 channels and mutants thereof were expressed in mammalian cells and were assayed with the whole-cell patch-clamp method. Unlike for the M-superfamily mu-conotoxin GIIIA from Conus geographus, channel block by MrVIA was strongly diminished after activating the Na(V) channels by depolarizing voltage steps. Searching for the source of this voltage dependence, the gating charges in all four-voltage sensors were reduced by site-directed mutagenesis showing that alterations of the voltage sensor in domain-2 have the strongest impact on MrVIA action. These results, together with previous findings that the effect of MrVIA depends on the structure of the pore-loop in domain-3, suggest a functional similarity with scorpion beta-toxins. In fact, MrVIA functionally competed with the scorpion beta-toxin Ts1 from Tityus serrulatus, while it did not show competition with mu-GIIIA. Ts1 and mu-GIIIA did not compete either. Thus, similar to scorpion beta-toxins, muO-conotoxins are voltage-sensor toxins targeting receptor site-4 on Na(V) channels. They "block" Na(+) flow most likely by hindering the voltage sensor in domain-2 from activating and, hence, the channel from opening.


Assuntos
Conotoxinas/farmacologia , Proteínas Musculares/antagonistas & inibidores , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Linhagem Celular , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Técnicas de Patch-Clamp , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Venenos de Escorpião/farmacologia , Sódio/metabolismo , Canais de Sódio/química , Canais de Sódio/genética , Canais de Sódio/metabolismo , Transfecção
14.
Toxicon ; 49(3): 318-28, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17118419

RESUMO

We have characterized the defining members of a novel subfamily of excitatory conotoxins, the short kappaA-conotoxins (kappaA(S)-conotoxins). kappaA-conotoxins PIVE and PIVF (kappaA-PIVE and kappaA-PIVF) were purified from Conus purpurascens venom. Both peptides elicited excitatory activity upon injection into fish. kappaA-PIVE was synthesized for further characterization. The excitatory effects of kappaA-PIVE in vivo were dose dependent, causing hyperactivity at low doses and rapid immobilization at high doses, symptomatic of a type of excitotoxic shock. Consistent with these observations, kappaA-PIVE caused repetitive action potentials in frog motor axons in vitro. Similar results have been reported for other structurally distinct conotoxin families; such peptides appear to be required by most fish-hunting cone snails for the rapid immobilization of prey. Unexpected structure-function relationships were revealed between these peptides and two families of homologous conotoxins: the alphaA-conotoxins (muscle nAChR antagonists) and kappaA-conotoxins (excitotoxins), which all share a common arrangement of cysteine residues (CC-C-C-C-C). Biochemically, the kappaA(S)-conotoxins more closely resemble the alphaA(S)-conotoxins than the other kappaA-conotoxin subfamily, the long kappaA-conotoxins (kappaA(L)-conotoxins); however, kappaA(S)- and alphaA(S)-conotoxins produce different physiological effects. In contrast, the kappaA(S)-and kappaA(L)-conotoxins that diverge in several biochemical characteristics are clearly more similar in their physiological effects.


Assuntos
Conotoxinas/isolamento & purificação , Caramujo Conus , Neurotoxinas/isolamento & purificação , Sequência de Aminoácidos , Animais , Bioensaio , Encéfalo/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Conotoxinas/química , Conotoxinas/farmacologia , Relação Dose-Resposta a Droga , Carpa Dourada , Camundongos , Dados de Sequência Molecular , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Neurotoxinas/química , Neurotoxinas/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Técnicas de Patch-Clamp , Rana pipiens , Relação Estrutura-Atividade , Xenopus
15.
Biochemistry ; 45(27): 8331-40, 2006 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-16819832

RESUMO

Using assay-directed fractionation of the venom from the vermivorous cone snail Conus planorbis, we isolated a new conotoxin, designated pl14a, with potent activity at both nicotinic acetylcholine receptors and a voltage-gated potassium channel subtype. pl14a contains 25 amino acid residues with an amidated C-terminus, an elongated N-terminal tail (six residues), and two disulfide bonds (1-3, 2-4 connectivity) in a novel framework distinct from other conotoxins. The peptide was chemically synthesized, and its three-dimensional structure was demonstrated to be well-defined, with an alpha-helix and two 3(10)-helices present. Analysis of a cDNA clone encoding the prepropeptide precursor of pl14a revealed a novel signal sequence, indicating that pl14a belongs to a new gene superfamily, the J-conotoxin superfamily. Five additional peptides in the J-superfamily were identified. Intracranial injection of pl14a in mice elicited excitatory symptoms that included shaking, rapid circling, barrel rolling, and seizures. Using the oocyte heterologous expression system, pl14a was shown to inhibit both a K+ channel subtype (Kv1.6, IC50 = 1.59 microM) and neuronal (IC50 = 8.7 microM for alpha3beta4) and neuromuscular (IC50 = 0.54 microM for alpha1beta1 epsilondelta) subtypes of the nicotinic acetylcholine receptor (nAChR). Similarities in sequence and structure are apparent between the middle loop of pl14a and the second loop of a number of alpha-conotoxins. This is the first conotoxin shown to affect the activity of both voltage-gated and ligand-gated ion channels.


Assuntos
Conotoxinas/farmacologia , Caramujo Conus/química , Canal de Potássio Kv1.6/antagonistas & inibidores , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Conotoxinas/química , Conotoxinas/isolamento & purificação , Caramujo Conus/genética , DNA Complementar/química , Camundongos , Dados de Sequência Molecular , Músculos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Antagonistas Nicotínicos/química , Antagonistas Nicotínicos/isolamento & purificação , Filogenia , Conformação Proteica
16.
J Gen Physiol ; 128(1): 133-45, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16801386

RESUMO

Ion channels are membrane-spanning proteins that allow ions to permeate at high rates. The kinetic characteristics of the channels present in a cell determine the cell signaling profile and therefore cell function in many different physiological processes. We found that Kv1.7 channels from mouse heart muscle have two putative translation initiation start sites that generate two channel isoforms with different functional characteristics, mKv1.7L (489 aa) and a shorter mKv1.7S (457 aa). The electrophysiological analysis of mKv1.7L and mKv1.7S channels revealed that the two channel isoforms have different inactivation kinetics. The channel resulting from the longer protein (L) inactivates faster than the shorter channels (S). Our data supports the hypothesis that mKv1.7L channels inactivate predominantly due to an N-type related mechanism, which is impaired in the mKv1.7S form. Furthermore, only the longer version mKv1.7L is regulated by the cell redox state, whereas the shorter form mKv1.7S is not. Thus, expression starting at each translation initiation site results in significant functional divergence. Our data suggest that the redox modulation of mKv1.7L may occur through a site in the cytoplasmic N-terminal domain that seems to encompass a metal coordination motif resembling those found in many redox-sensitive proteins. The mRNA expression profile and redox modulation of mKv1.7 kinetics identify these channels as molecular entities of potential importance in cellular redox-stress states such as hypoxia.


Assuntos
Miocárdio/metabolismo , Superfamília Shaker de Canais de Potássio/fisiologia , Animais , Sequência de Bases , Clonagem Molecular , Dissulfetos/farmacologia , Ditiotreitol/farmacologia , Feminino , Cinética , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Mutação/genética , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oxirredução , Potássio/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , RNA Complementar/genética , Homologia de Sequência do Ácido Nucleico , Superfamília Shaker de Canais de Potássio/genética , Xenopus laevis
17.
Biochemistry ; 45(23): 7404-14, 2006 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-16752929

RESUMO

MuO-conotoxin MrVIB is a blocker of voltage-gated sodium channels, including TTX-sensitive and -resistant subtypes. A comprehensive characterization of this peptide has been hampered by the lack of sufficient synthetic material. Here, we describe the successful chemical synthesis and oxidative folding of MrVIB that has made an investigation of the pharmacological properties and therapeutic potential of the peptide feasible. We show for the first time that synthetic MrVIB blocks rat NaV1.8 sodium channels and has potent and long-lasting local anesthetic effects when tested in two pain assays in rats. Furthermore, MrVIB can block propagation of action potentials in A- and C-fibers in sciatic nerve as well as skeletal muscle in isolated preparations from rat. Our work provides the first example of analgesia produced by a conotoxin that blocks sodium channels. The emerging diversity of antinociceptive mechanisms targeted by different classes of conotoxins is discussed.


Assuntos
Analgésicos/farmacologia , Conotoxinas/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Tetrodotoxina/farmacologia , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Conotoxinas/química , Masculino , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.8 , Ratos , Ratos Sprague-Dawley , Canais de Sódio
18.
Protein Expr Purif ; 47(2): 640-4, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16542850

RESUMO

Conkunitzin-S1 from the cone snail Conus striatus is the first member of a new neurotoxin family with a canonical Kunitz domain fold. Conk-S1 is 60 amino acids long and lacks one of the three conserved disulfide bonds typically found in Kunitz domain modules. It binds specifically to voltage activated potassium channels of the Shaker family. The peptide was expressed in insoluble form in fusion with an N-terminal intein. Refolding in the presence of glutathione followed by pH shift-induced cleavage of the fusion protein resulted in a functional toxin as demonstrated by voltage-clamp measurements.


Assuntos
Escherichia coli , Venenos de Moluscos/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Animais , Escherichia coli/metabolismo , Feminino , Concentração de Íons de Hidrogênio , Corpos de Inclusão/metabolismo , Inteínas/genética , Venenos de Moluscos/química , Oócitos/citologia , Oócitos/metabolismo , Técnicas de Patch-Clamp , Proteínas Recombinantes de Fusão/química , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Xenopus
19.
FEBS Lett ; 580(5): 1360-4, 2006 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-16458302

RESUMO

Several families of peptide toxins from cone snails affect voltage-gated sodium (Na(V)) channels: mu-conotoxins block the pore, delta-conotoxins inhibit channel inactivation, and muO-conotoxins inhibit Na(V) channels by an unknown mechanism. The only currently known muO-conotoxins MrVIA and MrVIB from Conus marmoreus were applied to cloned rat skeletal muscle (Na(V)1.4) and brain (Na(V)1.2) sodium channels in mammalian cells. A systematic domain-swapping strategy identified the C-terminal pore loop of domain-3 as the major determinant for Na(V)1.4 being more potently blocked than Na(V)1.2 channels. muO-conotoxins therefore show an interaction pattern with Na(V) channels that is clearly different from the related mu- and delta-conotoxins, indicative of a distinct molecular mechanism of channel inhibition.


Assuntos
Conotoxinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Animais , Sítios de Ligação , Química Encefálica , Clonagem Molecular , Conotoxinas/metabolismo , Caramujo Conus , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/química , Músculo Esquelético/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.2 , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ratos , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/química , Canais de Sódio/genética
20.
FEBS Lett ; 579(18): 3881-4, 2005 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-15990094

RESUMO

Various neurotoxic peptides modulate voltage-gated sodium (Na(V)) channels and thereby affect cellular excitability. Delta-conotoxins from predatory cone snails slow down inactivation of Na(V) channels, but their interaction site and mechanism of channel modulation are unknown. Here, we show that delta-conotoxin SVIE from Conus striatus interacts with a conserved hydrophobic triad (YFV) in the domain-4 voltage sensor of Na(V) channels. This site overlaps with that of the scorpion alpha-toxin Lqh-2, but not with the alpha-like toxin Lqh-3 site. Delta-SVIE functionally competes with Lqh-2, but exhibits strong cooperativity with Lqh-3, presumably by synergistically trapping the voltage sensor in its "on" position.


Assuntos
Conotoxinas/metabolismo , Canais de Sódio/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Cisteína/química , Eletrofisiologia , Humanos , Dados de Sequência Molecular , Proteínas Musculares/metabolismo , Neurotoxinas/química , Ligação Proteica , Ratos , Venenos de Escorpião/química , Venenos de Escorpião/metabolismo , Escorpiões , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA