Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(16): 5087-92, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25848010

RESUMO

Prey shifts in carnivorous predators are events that can initiate the accelerated generation of new biodiversity. However, it is seldom possible to reconstruct how the change in prey preference occurred. Here we describe an evolutionary "smoking gun" that illuminates the transition from worm hunting to fish hunting among marine cone snails, resulting in the adaptive radiation of fish-hunting lineages comprising ∼100 piscivorous Conus species. This smoking gun is δ-conotoxin TsVIA, a peptide from the venom of Conus tessulatus that delays inactivation of vertebrate voltage-gated sodium channels. C. tessulatus is a species in a worm-hunting clade, which is phylogenetically closely related to the fish-hunting cone snail specialists. The discovery of a δ-conotoxin that potently acts on vertebrate sodium channels in the venom of a worm-hunting cone snail suggests that a closely related ancestral toxin enabled the transition from worm hunting to fish hunting, as δ-conotoxins are highly conserved among fish hunters and critical to their mechanism of prey capture; this peptide, δ-conotoxin TsVIA, has striking sequence similarity to these δ-conotoxins from piscivorous cone snail venoms. Calcium-imaging studies on dissociated dorsal root ganglion (DRG) neurons revealed the peptide's putative molecular target (voltage-gated sodium channels) and mechanism of action (inhibition of channel inactivation). The results were confirmed by electrophysiology. This work demonstrates how elucidating the specific interactions between toxins and receptors from phylogenetically well-defined lineages can uncover molecular mechanisms that underlie significant evolutionary transitions.


Assuntos
Caramujo Conus/fisiologia , Peixes/fisiologia , Comportamento Predatório/fisiologia , Sequência de Aminoácidos , Animais , Bioensaio , Conotoxinas/química , Conotoxinas/toxicidade , Caramujo Conus/anatomia & histologia , Dados de Sequência Molecular , Peptídeos/metabolismo , Filogenia
2.
J Biol Chem ; 291(13): 7205-20, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-26817840

RESUMO

Cone snail toxins are well known blockers of voltage-gated sodium channels, a property that is of broad interest in biology and therapeutically in treating neuropathic pain and neurological disorders. Although most conotoxin channel blockers function by direct binding to a channel and disrupting its normal ion movement, conotoxin µO§-GVIIJ channel blocking is unique, using both favorable binding interactions with the channel and a direct tether via an intermolecular disulfide bond. Disulfide exchange is possible because conotoxin µO§-GVIIJ contains anS-cysteinylated Cys-24 residue that is capable of exchanging with a free cysteine thiol on the channel surface. Here, we present the solution structure of an analog of µO§-GVIIJ (GVIIJ[C24S]) and the results of structure-activity studies with synthetic µO§-GVIIJ variants. GVIIJ[C24S] adopts an inhibitor cystine knot structure, with two antiparallel ß-strands stabilized by three disulfide bridges. The loop region linking the ß-strands (loop 4) presents residue 24 in a configuration where it could bind to the proposed free cysteine of the channel (Cys-910, rat NaV1.2 numbering; at site 8). The structure-activity study shows that three residues (Lys-12, Arg-14, and Tyr-16) located in loop 2 and spatially close to residue 24 were also important for functional activity. We propose that the interaction of µO§-GVIIJ with the channel depends on not only disulfide tethering via Cys-24 to a free cysteine at site 8 on the channel but also the participation of key residues of µO§-GVIIJ on a distinct surface of the peptide.


Assuntos
Conotoxinas/química , Dissulfetos/química , Proteínas Musculares/química , Canal de Sódio Disparado por Voltagem NAV1.2/química , Bloqueadores dos Canais de Sódio/química , Canais de Sódio/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Conotoxinas/síntese química , Cristalografia por Raios X , Expressão Gênica , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Caramujos/química , Bloqueadores dos Canais de Sódio/síntese química , Canais de Sódio/genética , Canais de Sódio/metabolismo , Técnicas de Síntese em Fase Sólida , Relação Estrutura-Atividade
3.
Proc Natl Acad Sci U S A ; 111(7): 2758-63, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24497506

RESUMO

A cone snail venom peptide, µO§-conotoxin GVIIJ from Conus geographus, has a unique posttranslational modification, S-cysteinylated cysteine, which makes possible formation of a covalent tether of peptide to its target Na channels at a distinct ligand-binding site. µO§-conotoxin GVIIJ is a 35-aa peptide, with 7 cysteine residues; six of the cysteines form 3 disulfide cross-links, and one (Cys24) is S-cysteinylated. Due to limited availability of native GVIIJ, we primarily used a synthetic analog whose Cys24 was S-glutathionylated (abbreviated GVIIJSSG). The peptide-channel complex is stabilized by a disulfide tether between Cys24 of the peptide and Cys910 of rat (r) NaV1.2. A mutant channel of rNaV1.2 lacking a cysteine near the pore loop of domain II (C910L), was >10(3)-fold less sensitive to GVIIJSSG than was wild-type rNaV1.2. In contrast, although rNaV1.5 was >10(4)-fold less sensitive to GVIIJSSG than NaV1.2, an rNaV1.5 mutant with a cysteine in the homologous location, rNaV1.5[L869C], was >10(3)-fold more sensitive than wild-type rNaV1.5. The susceptibility of rNaV1.2 to GVIIJSSG was significantly altered by treating the channels with thiol-oxidizing or disulfide-reducing agents. Furthermore, coexpression of rNaVß2 or rNaVß4, but not that of rNaVß1 or rNaVß3, protected rNaV1.1 to -1.7 (excluding NaV1.5) against block by GVIIJSSG. Thus, GVIIJ-related peptides may serve as probes for both the redox state of extracellular cysteines and for assessing which NaVß- and NaVα-subunits are present in native neurons.


Assuntos
Conotoxinas/toxicidade , Dissulfetos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Neurônios/metabolismo , Bloqueadores do Canal de Sódio Disparado por Voltagem/toxicidade , Sequência de Aminoácidos , Animais , Sequência de Bases , Cromatografia Líquida de Alta Pressão , Conotoxinas/genética , Conotoxinas/metabolismo , Cisteína/metabolismo , Primers do DNA/genética , DNA Complementar/genética , Dados de Sequência Molecular , Oócitos/metabolismo , Técnicas de Patch-Clamp , Ratos , Análise de Sequência de DNA , Espectrometria de Massas em Tandem , Bloqueadores do Canal de Sódio Disparado por Voltagem/metabolismo
4.
Biochemistry ; 54(25): 3911-20, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26039939

RESUMO

µO§-Conotoxin GVIIJ is a 35-amino acid peptide that readily blocks six of eight tested NaV1 subunit isoforms of voltage-gated sodium channels. µO§-GVIIJ is unusual in having an S-cysteinylated cysteine (at residue 24). A proposed reaction scheme involves the peptide-channel complex stabilized by a disulfide bond formed via thiol-disulfide exchange between Cys24 of the peptide and a Cys residue at neurotoxin receptor site 8 in the pore module of the channel (specifically, Cys910 of rat NaV1.2). To examine this model, we synthesized seven derivatives of µO§-GVIIJ in which Cys24 was disulfide-bonded to various thiols (or SR groups) and tested them on voltage-clamped Xenopus laevis oocytes expressing NaV1.2. In the proposed model, the SR moiety is a leaving group that is no longer present in the final peptide-channel complex; thus, the same koff value should be obtained regardless of the SR group. We observed that all seven derivatives, whose kon values varied over a 30-fold range, had the same koff value. Concordant results were observed with NaV1.6, for which the koff was 17-fold larger. Additionally, we tested two µO§-GVIIJ derivatives (where SR was glutathione or a free thiol) on two NaV1.2 Cys replacement mutants (NaV1.2[C912A] and NaV1.2[C918A]) without and with reduction of channel disulfides by dithiothreitol. The results indicate that Cys910 in wild-type NaV1.2 has a free thiol and conversely suggest that in NaV1.2[C912A] and NaV1.2[C918A], Cys910 is disulfide-bonded to Cys918 and Cys912, respectively. Redox states of extracellular cysteines of sodium channels have hitherto received scant attention, and further experiments with GVIIJ may help fill this void.


Assuntos
Conotoxinas/química , Cisteína/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2/química , Animais , Sítios de Ligação , Conotoxinas/metabolismo , Cisteína/química , Cisteína/genética , Dissulfetos/química , Dissulfetos/metabolismo , Cinética , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Oócitos , Oxirredução , Ratos , Xenopus laevis
5.
J Neurophysiol ; 113(7): 2289-301, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25632083

RESUMO

We investigated the identities of the isoforms of the α (NaV1)- and ß (NaVß)-subunits of voltage-gated sodium channels, including those responsible for action potentials in rodent sciatic nerves. To examine α-subunits, we used seven µ-conotoxins, which target site 1 of the channel. With the use of exogenously expressed channels, we show that two of the µ-conotoxins, µ-BuIIIB and µ-SxIIIA, are 50-fold more potent in blocking NaV1.6 from mouse than that from rat. Furthermore, we observed that µ-BuIIIB and µ-SxIIIA are potent blockers of large, myelinated A-fiber compound action potentials (A-CAPs) [but not small, unmyelinated C-fiber CAPs (C-CAPs)] in the sciatic nerve of the mouse (unlike A-CAPs of the rat, previously shown to be insensitive to these toxins). To investigate ß-subunits, we used two synthetic derivatives of the recently discovered µO§-conotoxin GVIIJ that define site 8 of the channel, as previously characterized with cloned rat NaV1- and NaVß-subunits expressed in Xenopus laevis oocytes, where it was shown that µO§-GVIIJ is a potent inhibitor of several NaV1-isoforms and that coexpression of NaVß2 or -ß4 (but not NaVß1 or -ß3) totally protects against block by µO§-GVIIJ. We report here the effects of µO§-GVIIJ on 1) sodium currents of mouse NaV1.6 coexpressed with various combinations of NaVß-subunits in oocytes; 2) A- and C-CAPs of mouse and rat sciatic nerves; and 3) sodium currents of small and large neurons dissociated from rat dorsal root ganglia. Our overall results lead us to conclude that action potentials in A-fibers of the rodent sciatic nerve are mediated primarily by NaV1.6 associated with NaVß2 or NaVß4.


Assuntos
Potenciais de Ação/fisiologia , Conotoxinas/administração & dosagem , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Canais de Sódio Disparados por Voltagem/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Células Cultivadas , Conotoxinas/química , Relação Dose-Resposta a Droga , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Oócitos , Subunidades Proteicas , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo , Relação Estrutura-Atividade , Bloqueadores do Canal de Sódio Disparado por Voltagem , Canais de Sódio Disparados por Voltagem/química , Xenopus laevis
6.
Proc Natl Acad Sci U S A ; 109(5): 1388-95, 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22307590

RESUMO

We describe a functional profiling strategy to identify and characterize subtypes of neurons present in a peripheral ganglion, which should be extendable to neurons in the CNS. In this study, dissociated dorsal-root ganglion neurons from mice were exposed to various pharmacological agents (challenge compounds), while at the same time the individual responses of >100 neurons were simultaneously monitored by calcium imaging. Each challenge compound elicited responses in only a subset of dorsal-root ganglion neurons. Two general types of challenge compounds were used: agonists of receptors (ionotropic and metabotropic) that alter cytoplasmic calcium concentration (receptor-agonist challenges) and compounds that affect voltage-gated ion channels (membrane-potential challenges). Notably, among the latter are K-channel antagonists, which elicited unexpectedly diverse types of calcium responses in different cells (i.e., phenotypes). We used various challenge compounds to identify several putative neuronal subtypes on the basis of their shared and/or divergent functional, phenotypic profiles. Our results indicate that multiple receptor-agonist and membrane-potential challenges may be applied to a neuronal population to identify, characterize, and discriminate among neuronal subtypes. This experimental approach can uncover constellations of plasma membrane macromolecules that are functionally coupled to confer a specific phenotypic profile on each neuronal subtype. This experimental platform has the potential to bridge a gap between systems and molecular neuroscience with a cellular-focused neuropharmacology, ultimately leading to the identification and functional characterization of all neuronal subtypes at a given locus in the nervous system.


Assuntos
Neurônios/fisiologia , Animais , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/patologia , Gânglios Espinais/fisiologia , Camundongos , Venenos de Moluscos/química , Neurônios/efeitos dos fármacos , Neurônios/patologia , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Tetraetilamônio/farmacologia , Tetrodotoxina/farmacologia
7.
Proc Natl Acad Sci U S A ; 108(25): 10302-7, 2011 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-21652775

RESUMO

Voltage-gated sodium channels (VGSCs) are important for action potentials. There are seven major isoforms of the pore-forming and gate-bearing α-subunit (Na(V)1) of VGSCs in mammalian neurons, and a given neuron can express more than one isoform. Five of the neuronal isoforms, Na(V)1.1, 1.2, 1.3, 1.6, and 1.7, are exquisitely sensitive to tetrodotoxin (TTX), and a functional differentiation of these presents a serious challenge. Here, we examined a panel of 11 µ-conopeptides for their ability to block rodent Na(V)1.1 through 1.8 expressed in Xenopus oocytes. Although none blocked Na(V)1.8, a TTX-resistant isoform, the resulting "activity matrix" revealed that the panel could readily discriminate between the members of all pair-wise combinations of the tested isoforms. To examine the identities of endogenous VGSCs, a subset of the panel was tested on A- and C-compound action potentials recorded from isolated preparations of rat sciatic nerve. The results show that the major subtypes in the corresponding A- and C-fibers were Na(V)1.6 and 1.7, respectively. Ruled out as major players in both fiber types were Na(V)1.1, 1.2, and 1.3. These results are consistent with immunohistochemical findings of others. To our awareness this is the first report describing a qualitative pharmacological survey of TTX-sensitive Na(V)1 isoforms responsible for propagating action potentials in peripheral nerve. The panel of µ-conopeptides should be useful in identifying the functional contributions of Na(V)1 isoforms in other preparations.


Assuntos
Potenciais de Ação/fisiologia , Conotoxinas/metabolismo , Isoformas de Proteínas/metabolismo , Nervo Isquiático/fisiologia , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/metabolismo , Animais , Neurotoxinas/metabolismo , Oócitos/citologia , Oócitos/fisiologia , Técnicas de Patch-Clamp , Ratos , Xenopus laevis
8.
Biochemistry ; 51(49): 9826-35, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23167564

RESUMO

In the preparation of synthetic conotoxins containing multiple disulfide bonds, oxidative folding can produce numerous permutations of disulfide bond connectivities. Establishing the native disulfide connectivities thus presents a significant challenge when the venom-derived peptide is not available, as is increasingly the case when conotoxins are identified from cDNA sequences. Here, we investigate the disulfide connectivity of µ-conotoxin KIIIA, which was predicted originally to have a [C1-C9,C2-C15,C4-C16] disulfide pattern based on homology with closely related µ-conotoxins. The two major isomers of synthetic µ-KIIIA formed during oxidative folding were purified and their disulfide connectivities mapped by direct mass spectrometric collision-induced dissociation fragmentation of the disulfide-bonded polypeptides. Our results show that the major oxidative folding product adopts a [C1-C15,C2-C9,C4-C16] disulfide connectivity, while the minor product adopts a [C1-C16,C2-C9,C4-C15] connectivity. Both of these peptides were potent blockers of Na(V)1.2 (K(d) values of 5 and 230 nM, respectively). The solution structure for µ-KIIIA based on nuclear magnetic resonance data was recalculated with the [C1-C15,C2-C9,C4-C16] disulfide pattern; its structure was very similar to the µ-KIIIA structure calculated with the incorrect [C1-C9,C2-C15,C4-C16] disulfide pattern, with an α-helix spanning residues 7-12. In addition, the major folding isomers of µ-KIIIB, an N-terminally extended isoform of µ-KIIIA identified from its cDNA sequence, were isolated. These folding products had the same disulfide connectivities as µ-KIIIA, and both blocked Na(V)1.2 (K(d) values of 470 and 26 nM, respectively). Our results establish that the preferred disulfide pattern of synthetic µ-KIIIA and µ-KIIIB folded in vitro is 1-5/2-4/3-6 but that other disulfide isomers are also potent sodium channel blockers. These findings raise questions about the disulfide pattern(s) of µ-KIIIA in the venom of Conus kinoshitai; indeed, the presence of multiple disulfide isomers in the venom could provide a means of further expanding the snail's repertoire of active peptides.


Assuntos
Conotoxinas/farmacologia , Dissulfetos/química , Ativação do Canal Iônico , Isomerismo , Canais de Sódio/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Clonagem Molecular , Conotoxinas/química , Conotoxinas/genética , Espectrometria de Massas , Ressonância Magnética Nuclear Biomolecular , Oxirredução
9.
J Pharmacol Exp Ther ; 338(2): 687-93, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21586605

RESUMO

Voltage-gated sodium channels (VGSCs) consist of a pore-forming α-subunit and regulatory ß-subunits. Several families of neuroactive peptides of Conus snails target VGSCs, including µO-conotoxins and µ-conotoxins. Unlike µ-conotoxins and the guanidinium alkaloid saxitoxin (STX), which are pore blockers, µO-conotoxins MrVIA and MrVIB inhibit VGSCs by modifying channel gating. µO-MrVIA/B can block Na(v)1.8 (a tetrodotoxin-resistant isoform of VGSCs) and have analgesic properties. The effect of Na(v)ß-subunit coexpression on susceptibility to block by µO-MrVIA/B and STX has, until now, not been reported. Here, we show that ß1-, ß2-, ß3-, and ß4-subunits, when individually coexpressed with Na(v)1.8 in Xenopus laevis oocytes, increased the k(on) of the block produced by µO-MrVIB (by 3-, 32-, 2-, and 7-fold, respectively) and modestly decreased the apparent k(off). Strong depolarizing prepulses markedly accelerated MrVIB washout with rates dependent on ß-subunit coexpression. Thus, coexpression of ß-subunits with Na(v)1.8 can strongly influence the affinity of the conopeptide for the channel. This observation is of particular interest because ß-subunit expression can be dynamic, e.g., ß2-expression is up-regulated after nerve injury (J Neurosci, 25:10970-10980, 2005); therefore, the effectiveness of a µO-conotoxin as a channel blocker could be enhanced by the conditions that may call for its use therapeutically. In contrast to MrVIB's action, the STX-induced block of Na(v)1.8 was only marginally, if at all, affected by coexpression of any of the ß-subunits. Our results raise the possibility that µO-conotoxins and perhaps other gating modifiers may provide a means to functionally assess the ß-subunit composition of VGSC complexes in neurons.


Assuntos
Analgésicos não Narcóticos/farmacologia , Conotoxinas/farmacologia , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Animais , Relação Dose-Resposta a Droga , Feminino , Canal de Sódio Disparado por Voltagem NAV1.8 , Oócitos/metabolismo , Biossíntese Peptídica/efeitos dos fármacos , Ligação Proteica , Subunidades Proteicas/biossíntese , Ratos , Canais de Sódio/fisiologia , Xenopus laevis
10.
Biochemistry ; 49(23): 4804-12, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20459109

RESUMO

The possibility of independently manipulating the affinity and efficacy of pore-blocking ligands of sodium channels is of interest for the development of new drugs for the treatment of pain. The analgesic mu-conotoxin KIIIA (KIIIA), a 16-residue peptide with three disulfide bridges, is a pore blocker of voltage-gated sodium channels, including neuronal subtype Na(V)1.2 (K(d) = 5 nM). At saturating concentrations, KIIIA incompletely blocks the sodium current of Na(V)1.2, leaving a 5% residual current (rI(Na)). Lys7 is an important residue: the K7A mutation decreases both the efficacy (i.e., increases rI(Na) to 23%) and the affinity of the peptide (K(d) = 115 nM). In this report, various replacements of residue 7 were examined to determine whether affinity and efficacy were inexorably linked. Because of their facile chemical synthesis, KIIIA analogues that had as a core structure the disulfide-depleted KIIIA[C1A,C2U,C9A,C15U] (where U is selenocysteine) or ddKIIIA were used. Analogues ddKIIIA and ddKIIIA[K7X], where X represents one of nine different amino acids, were tested on voltage-clamped Xenopus oocytes expressing rat Na(V)1.2 or Na(V)1.4. Their affinities ranged from 0.01 to 36 muM and rI(Na) values from 2 to 42%, and these two variables appeared to be uncorrelated. Instead, rI(Na) varied inversely with side chain size, and remarkably charge and hydrophobicity appeared to be inconsequential. The ability to manipulate a mu-conopeptide's affinity and efficacy, as well as its capacity to interfere with subsequent tetrodotoxin binding, greatly expands its scope as a reagent for probing sodium channel structure and function and may also lead to the development of mu-conotoxins as safe analgesics.


Assuntos
Conotoxinas/metabolismo , Proteínas Musculares/fisiologia , Prótons , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Conotoxinas/síntese química , Dissulfetos/metabolismo , Indicadores e Reagentes , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.2 , Proteínas do Tecido Nervoso , Oócitos/metabolismo , Ligação Proteica , Ratos , Selenocisteína/genética , Bloqueadores dos Canais de Sódio/síntese química , Xenopus
11.
J Neurophysiol ; 104(1): 88-97, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20410356

RESUMO

The guanidinium alkaloids tetrodotoxin (TTX) and saxitoxin (STX) are classic ligands of voltage-gated sodium channels (VGSCs). Like TTX and STX, micro-conotoxin peptides are pore blockers but with greater VGSC subtype selectivity. micro-Conotoxin KIIIA blocks the neuronal subtype Na(V)1.2 with nanomolar affinity and we recently discovered that KIIIA and its mutant with one fewer positive charge, KIIIA[K7A], could act synergistically with TTX in a ternary peptide x TTX x Na(V) complex. In the complex, the peptide appeared to trap TTX in its normal binding site such that TTX could not readily dissociate from the channel until the peptide had done so; in turn, the presence of TTX accelerated the rate at which peptide dissociated from the channel. In the present study we examined the inhibition of Na(V)1.2, exogenously expressed in Xenopus oocytes, by STX (a divalent cation) and its sulfated congener GTX2/3 (with a net +1 charge). Each could form a ternary complex with KIIIA and Na(V)1.2, as previously found with TTX (a monovalent cation), but only when STX or GTX2/3 was added before KIIIA. The KIIIA x alkaloid x Na(V) complex was considerably less stable with STX than with either GTX2/3 or TTX. In contrast, ternary KIIIA[K7A] x alkaloid x Na(V) complexes could be formed with either order of ligand addition and were about equally stable with STX, GTX2/3, or TTX. The most parsimonious interpretation of the overall results is that the alkaloid and peptide are closely apposed in the ternary complex. The demonstration that two interacting ligands ("syntoxins") occupy adjacent sites raises the possibility of evolving a much more sophisticated neuropharmacology of VGSCs.


Assuntos
Conotoxinas/metabolismo , Conotoxinas/farmacologia , Saxitoxina/metabolismo , Saxitoxina/farmacologia , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/fisiologia , Tetrodotoxina/metabolismo , Tetrodotoxina/farmacologia , Vestíbulo do Labirinto/efeitos dos fármacos , Vestíbulo do Labirinto/fisiologia , Algoritmos , Animais , Fenômenos Eletrofisiológicos , Feminino , Cinética , Modelos Neurológicos , Modelos Estatísticos , Canal de Sódio Disparado por Voltagem NAV1.2 , Proteínas do Tecido Nervoso , Oócitos/efeitos dos fármacos , Técnicas de Patch-Clamp , Gravidez , Ratos , Canais de Sódio/metabolismo
12.
Mar Drugs ; 8(7): 2153-61, 2010 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-20714429

RESUMO

Neurotoxin receptor site 1, in the outer vestibule of the conducting pore of voltage-gated sodium channels (VGSCs), was first functionally defined by its ability to bind the guanidinium-containing agents, tetrodotoxin (TTX) and saxitoxin (STX). Subsequent studies showed that peptide micro-conotoxins competed for binding at site 1. All of these natural inhibitors block single sodium channels in an all-or-none manner on binding. With the discovery of an increasing variety of micro-conotoxins, and the synthesis of numerous derivatives, observed interactions between the channel and these different ligands have become more complex. Certain micro-conotoxin derivatives block single-channel currents partially, rather than completely, thus enabling the demonstration of interactions between the bound toxin and the channel's voltage sensor. Most recently, the relatively small micro-conotoxin KIIIA (16 amino acids) and its variants have been shown to bind simultaneously with TTX and exhibit both synergistic and antagonistic interactions with TTX. These interactions raise new pharmacological possibilities and place new constraints on the possible structures of the bound complexes of VGSCs with these toxins.


Assuntos
Conotoxinas/metabolismo , Canais de Sódio/metabolismo , Animais , Sítios de Ligação , Conotoxinas/química , Humanos , Ligação Proteica , Saxitoxina/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Tetrodotoxina/metabolismo
13.
Toxins (Basel) ; 12(3)2020 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-32245200

RESUMO

We report the discovery and functional characterization of αM-Conotoxin MIIIJ, a peptide from the venom of the fish-hunting cone snail Conus magus. Injections of αM-MIIIJ induced paralysis in goldfish (Carassius auratus) but not mice. Intracellular recording from skeletal muscles of fish (C. auratus) and frog (Xenopus laevis) revealed that αM-MIIIJ inhibited postsynaptic nicotinic acetylcholine receptors (nAChRs) with an IC50 of ~0.1 µM. With comparable potency, αM-MIIIJ reversibly blocked ACh-gated currents (IACh) of voltage-clamped X. laevis oocytes exogenously expressing nAChRs cloned from zebrafish (Danio rerio) muscle. αM-MIIIJ also protected against slowly-reversible block of IACh by α-bungarotoxin (α-BgTX, a snake neurotoxin) and α-conotoxin EI (α-EI, from Conus ermineus another fish hunter) that competitively block nAChRs at the ACh binding site. Furthermore, assessment by fluorescence microscopy showed that αM-MIIIJ inhibited the binding of fluorescently-tagged α-BgTX at neuromuscular junctions of X. laevis,C. auratus, and D. rerio. (Note, we observed that αM-MIIIJ can block adult mouse and human muscle nAChRs exogenously expressed in X. laevis oocytes, but with IC50s ~100-times higher than those of zebrafish nAChRs.) Taken together, these results indicate that αM-MIIIJ inhibits muscle nAChRs and furthermore apparently does so by interfering with the binding of ACh to its receptor. Comparative alignments with homologous sequences identified in other fish hunters revealed that αM-MIIIJ defines a new class of muscle nAChR inhibitors from cone snails.


Assuntos
Conotoxinas/farmacologia , Músculo Esquelético/efeitos dos fármacos , Junção Neuromuscular/efeitos dos fármacos , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/metabolismo , Potenciais de Ação/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Conotoxinas/química , Relação Dose-Resposta a Droga , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Carpa Dourada , Camundongos , Músculo Esquelético/metabolismo , Junção Neuromuscular/metabolismo , Antagonistas Nicotínicos/química , Paresia/induzido quimicamente , Comportamento Predatório/efeitos dos fármacos , Ligação Proteica , Alinhamento de Sequência , Especificidade da Espécie , Xenopus laevis
14.
Biochemistry ; 48(6): 1210-9, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19170536

RESUMO

Mu-conotoxin mu-KIIIA, from Conus kinoshitai, blocks mammalian neuronal voltage-gated sodium channels (VGSCs) and is a potent analgesic following systemic administration in mice. We have determined its solution structure using NMR spectroscopy. Key residues identified previously as being important for activity against VGSCs (Lys7, Trp8, Arg10, Asp11, His12, and Arg14) all reside on an alpha-helix with the exception of Arg14. To further probe structure-activity relationships of this toxin against VGSC subtypes, we have characterized the analogue mu-KIIIA[C1A,C9A], in which the Cys residues involved in one of the three disulfides in mu-KIIIA were replaced with Ala. Its structure is quite similar to that of mu-KIIIA, indicating that the Cys1-Cys9 disulfide bond could be removed without any significant distortion of the alpha-helix bearing the key residues. Consistent with this, mu-KIIIA[C1A,C9A] retained activity against VGSCs, with its rank order of potency being essentially the same as that of mu-KIIIA, namely, Na(V)1.2 > Na(V)1.4 > Na(V)1.7 >or= Na(V)1.1 > Na(V)1.3 > Na(V)1.5. Kinetics of block were obtained for Na(V)1.2, Na(V)1.4, and Na(V)1.7, and in each case, both k(on) and k(off) values of mu-KIIIA[C1A,C9A] were larger than those of mu-KIIIA. Our results show that the key residues for VGSC binding lie mostly on an alpha-helix and that the first disulfide bond can be removed without significantly affecting the structure of this helix, although the modification accelerates the on and off rates of the peptide against all tested VGSC subtypes. These findings lay the groundwork for the design of minimized peptides and helical mimetics as novel analgesics.


Assuntos
Analgésicos/química , Analgésicos/farmacologia , Conotoxinas/química , Conotoxinas/metabolismo , Dissulfetos/metabolismo , Sequência de Aminoácidos , Analgésicos/metabolismo , Animais , Simulação por Computador , Conotoxinas/farmacologia , Cristalografia por Raios X , Espectroscopia de Ressonância Magnética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Ratos , Canais de Sódio/metabolismo , Soluções , Relação Estrutura-Atividade , Xenopus
15.
Toxicon ; 53(1): 90-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18950653

RESUMO

Described herein is a general approach to identify novel compounds using the biodiversity of a megadiverse group of animals; specifically, the phylogenetic lineage of the venomous gastropods that belong to the genus Conus ("cone snails"). Cone snail biodiversity was exploited to identify three new mu-conotoxins, BuIIIA, BuIIIB and BuIIIC, encoded by the fish-hunting species Conus bullatus. BuIIIA, BuIIIB and BuIIIC are strikingly divergent in their amino acid composition compared to previous mu-conotoxins known to target the voltage-gated Na channel skeletal muscle subtype Na(v)1.4. Our preliminary results indicate that BuIIIB and BuIIIC are potent inhibitors of Na(v)1.4 (average block approximately 96%, at a 1muM concentration of peptide), displaying a very slow off-rate not seen in previously characterized mu-conotoxins that block Na(v)1.4. In addition, the three new C. bullatus mu-conopeptides help to define a new branch of the M-superfamily of conotoxins, namely M-5. The exogene strategy used to discover these Na channel-inhibiting peptides was based on both understanding the phylogeny of Conus, as well as the molecular genetics of venom mu-conotoxin peptides previously shown to generally target voltage-gated Na channels. The discovery of BuIIIA, BuIIIB and BuIIIC Na channel blockers expands the diversity of ligands useful in determining the structure-activity relationship of voltage-gated sodium channels.


Assuntos
Biodiversidade , Conotoxinas/química , Caramujo Conus/genética , Caramujo Conus/fisiologia , Bloqueadores dos Canais de Sódio/química , Aminoácido N-Acetiltransferase , Animais , Sequência de Bases , Clonagem Molecular , Conotoxinas/genética , Conotoxinas/metabolismo , DNA Complementar , Venenos de Moluscos/química , Oócitos , Filogenia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Xenopus
16.
Angew Chem Int Ed Engl ; 48(12): 2221-4, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19206132

RESUMO

Building bridges: The use of diselenide and selectively ((15)N/(13)C)-labeled disulfide bridges is combined to give improvements in oxidative folding and disulfide mapping. Conotoxin analogues, each with a pair of selenocysteines (Sec) and labeled cysteines (see scheme, red), exhibited significantly improved folding and the labeled cysteines allow correctly folded species to be rapidly identified by NMR spectroscopy.


Assuntos
Conotoxinas/síntese química , Cisteína/química , Peptídeos/química , Selenocisteína/química , Conotoxinas/química , Dissulfetos/química , Espectroscopia de Ressonância Magnética , Dobramento de Proteína
17.
Biochemistry ; 47(41): 10940-9, 2008 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-18798648

RESUMO

mu-SIIIA, a novel mu-conotoxin from Conus striatus, appeared to be a selective blocker of tetrodotoxin-resistant sodium channels in frog preparations. It also exhibited potent analgesic activity in mice, although its selectivity profile against mammalian sodium channels remains unknown. We have determined the structure of mu-SIIIA in aqueous solution and characterized its backbone dynamics by NMR and its functional properties electrophysiologically. Consistent with the absence of hydroxyprolines, mu-SIIIA adopts a single conformation with all peptide bonds in the trans conformation. The C-terminal region contains a well-defined helix encompassing residues 11-16, while residues 3-5 in the N-terminal region form a helix-like turn resembling 3 10-helix. The Trp12 and His16 side chains are close together, as in the related conotoxin mu-SmIIIA, but Asn2 is more distant. Dynamics measurements show that the N-terminus and Ser9 have larger-magnitude motions on the subnanosecond time scale, while the C-terminus is more rigid. Cys4, Trp12, and Cys13 undergo significant conformational exchange on microsecond to millisecond time scales. mu-SIIIA is a potent, nearly irreversible blocker of Na V1.2 but also blocks Na V1.4 and Na V1.6 with submicromolar potency. The selectivity profile of mu-SIIIA, including poor activity against the cardiac sodium channel, Na V1.5, is similar to that of the closely related mu-KIIIA, suggesting that the C-terminal regions of both are critical for blocking neuronal Na V1.2. The structural and functional characterization described in this paper of an analgesic mu-conotoxin that targets neuronal subtypes of mammalian sodium channels provides a basis for the design of novel analogues with an improved selectivity profile.


Assuntos
Conotoxinas/química , Bloqueadores dos Canais de Sódio/química , Animais , Cromatografia Líquida de Alta Pressão , Conotoxinas/farmacologia , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Bloqueadores dos Canais de Sódio/farmacologia , Xenopus
18.
J Am Chem Soc ; 130(43): 14280-6, 2008 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-18831583

RESUMO

Disulfide-rich peptides represent a megadiverse group of natural products with very promising therapeutic potential. To accelerate their functional characterization, high-throughput chemical synthesis and folding methods are required, including efficient mapping of multiple disulfide bridges. Here, we describe a novel approach for such mapping and apply it to a three-disulfide-bridged conotoxin, mu-SxIIIA (from the venom of Conus striolatus), whose discovery is also reported here for the first time. Mu-SxIIIA was chemically synthesized with three cysteine residues labeled 100% with (15)N/(13)C, while the remaining three cysteine residues were incorporated using a mixture of 70%/30% unlabeled/labeled Fmoc-protected residues. After oxidative folding, the major product was analyzed by NMR spectroscopy. Sequence-specific resonance assignments for the isotope-enriched Cys residues were determined with 2D versions of standard triple-resonance ((1)H, (13)C, (15)N) NMR experiments and 2D [(13)C, (1)H] HSQC. Disulfide patterns were directly determined with cross-disulfide NOEs confirming that the oxidation product had the disulfide connectivities characteristic of mu-conotoxins. Mu-SxIIIA was found to be a potent blocker of the sodium channel subtype Na(V)1.4 (IC50 = 7 nM). These results suggest that differential incorporation of isotope-labeled cysteine residues is an efficient strategy to map disulfides and should facilitate the discovery and structure-function studies of many bioactive peptides.


Assuntos
Conotoxinas/química , Cisteína/química , Dissulfetos/química , Espectroscopia de Ressonância Magnética/métodos , Peptídeos Cíclicos/química , Animais , Caramujo Conus , Espectroscopia de Ressonância Magnética/normas , Oxirredução , Dobramento de Proteína , Padrões de Referência , Fatores de Tempo
19.
J Neurochem ; 105(5): 1761-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18248619

RESUMO

Striatal dopamine (DA) plays a major role in the regulation of motor coordination and in the processing of salient information. We used voltammetry to monitor DA-release evoked by electrical stimulation in striatal slices, where interneurons continuously release acetylcholine. Use of the alpha6-selective antagonist alpha-conotoxin MII[E11A] and alpha4 knockout mice enabled identification of two populations of DA-ergic fibers. The first population had a low action potential threshold, and action potential-evoked DA-release from these fibers was modulated by alpha6. The second population had a higher action potential threshold, and only alpha4(non-alpha6) modulated action potential-evoked DA-release. Striatal DA-ergic neurons fire in both tonic and phasic patterns. When stimuli were applied in a train to mimic phasic firing, more DA-release was observed in alpha4 knockout versus wild-type mice. Furthermore, block of alpha4(non-alpha6), but not of alpha6, increased DA release evoked by a train. These results indicate that there are different classes of striatal DA-ergic fibers that express different subtypes of nicotinic receptors.


Assuntos
Corpo Estriado/metabolismo , Dopamina/metabolismo , Receptores Nicotínicos/biossíntese , Receptores Nicotínicos/fisiologia , Animais , Estimulação Elétrica , Potenciais Evocados/genética , Potenciais Evocados/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nicotínicos/genética
20.
Chem Biol ; 14(4): 399-407, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17462575

RESUMO

Disulfide-rich neurotoxins from venomous animals continue to provide compounds with therapeutic potential. Minimizing neurotoxins often results in removal of disulfide bridges or critical amino acids. To address this drug-design challenge, we explored the concept of disulfide-rich scaffolds consisting of isostere polymers and peptidic pharmacophores. Flexible spacers, such as amino-3-oxapentanoic or 6-aminohexanoic acids, were used to replace conformationally constrained parts of a three-disulfide-bridged conotoxin, SIIIA. The peptide-polymer hybrids, polytides, were designed based on cladistic identification of nonconserved loci in related peptides. After oxidative folding, the polytides appeared to be better inhibitors of sodium currents in dorsal root ganglia and sciatic nerves in mice. Moreover, the polytides appeared to be significantly more potent and longer-lasting analgesics in the inflammatory pain model in mice, when compared to SIIIA. The resulting polytides provide a promising strategy for transforming disulfide-rich peptides into therapeutics.


Assuntos
Analgésicos não Narcóticos/síntese química , Conotoxinas/síntese química , Desenho de Fármacos , Peptídeos/síntese química , Sequência de Aminoácidos , Analgésicos não Narcóticos/química , Analgésicos não Narcóticos/farmacologia , Animais , Conotoxinas/química , Conotoxinas/farmacologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Dor/tratamento farmacológico , Medição da Dor , Peptídeos/química , Peptídeos/farmacologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Bloqueadores dos Canais de Sódio/síntese química , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa