Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
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.
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
4.
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
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.
FEBS J ; 281(13): 2885-98, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24814369

RESUMO

Inhibitors of the α-subunit of the voltage-gated sodium channel subtype 1.3 (NaV 1.3) are of interest as pharmacological tools for the study of neuropathic pain associated with spinal cord injury and have potential therapeutic applications. The recently described µ-conotoxin BuIIIB (µ-BuIIIB) from Conus bullatus was shown to block NaV 1.3 with submicromolar potency (Kd = 0.2 µm), making it one of the most potent peptidic inhibitors of this subtype described to date. However, oxidative folding of µ-BuIIIB results in numerous folding isoforms, making it difficult to obtain sufficient quantities of the active form of the peptide for detailed structure-activity studies. In the present study, we report the synthesis and characterization of µ-BuIIIB analogs incorporating a disulfide-deficient, diselenide-containing scaffold designed to simplify synthesis and facilitate structure-activity studies directed at identifying amino acid residues involved in NaV 1.3 blockade. Our results indicate that, similar to other µ-conotoxins, the C-terminal residues (Trp16, Arg18 and His20) are most crucial for NaV 1 blockade. At the N-terminus, replacement of Glu3 by Ala resulted in an analog with an increased potency for NaV 1.3 (Kd = 0.07 µm), implicating this position as a potential site for modification for increased potency and/or selectivity. Further examination of this position showed that increased negative charge, through γ-carboxyglutamate replacement, decreased potency (Kd = 0.33 µm), whereas replacement with positively-charged 2,4-diamonobutyric acid increased potency (Kd = 0.036 µm). These results provide a foundation for the design and synthesis of µ-BuIIIB-based analogs with increased potency against NaV 1.3.


Assuntos
Analgésicos/síntese química , Conotoxinas/síntese química , Canal de Sódio Disparado por Voltagem NAV1.3/metabolismo , Selenocisteína/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/síntese química , Sequência de Aminoácidos , Substituição de Aminoácidos , Analgésicos/química , Analgésicos/farmacologia , Animais , Células Cultivadas , Conotoxinas/química , Conotoxinas/farmacologia , Cistina/química , Espectroscopia de Ressonância Magnética , Potenciais da Membrana , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Xenopus laevis
7.
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
8.
Eur J Med Chem ; 65: 144-50, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23707919

RESUMO

The µ-conotoxin KIIIA is a three disulfide-bridged blocker of voltage-gated sodium channels (VGSCs). The Lys(7) residue in KIIIA is an attractive target for manipulating the selectivity and efficacy of this peptide. Here, we report the design and chemical synthesis of µ-conopeptoid analogues (peptomers) in which we replaced Lys(7) with peptoid monomers of increasing side-chain size: N-methylglycine, N-butylglycine and N-octylglycine. In the first series of analogues, the peptide core contained all three disulfide bridges; whereas in the second series, a disulfide-depleted selenoconopeptide core was used to simplify oxidative folding. The analogues were tested for functional activity in blocking the Nav1.2 subtype of mammalian VGSCs exogenously expressed in Xenopus oocytes. All six analogues were active, with the N-methylglycine analogue, [Sar(7)]KIIIA, the most potent in blocking the channels while favouring lower efficacy. Our findings demonstrate that the use of N-substituted Gly residues in conotoxins show promise as a tool to optimize their pharmacological properties as potential analgesic drug leads.


Assuntos
Conotoxinas/química , Peptídeos/química , Peptoides/química , Selenoproteínas/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo , Estrutura Molecular , Selenoproteínas/síntese química , Selenoproteínas/química , Relação Estrutura-Atividade , Bloqueadores do Canal de Sódio Disparado por Voltagem/síntese química , Bloqueadores do Canal de Sódio Disparado por Voltagem/química
9.
ACS Chem Biol ; 8(6): 1344-51, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23557677

RESUMO

Among the µ-conotoxins that block vertebrate voltage-gated sodium channels (VGSCs), some have been shown to be potent analgesics following systemic administration in mice. We have determined the solution structure of a new representative of this family, µ-BuIIIB, and established its disulfide connectivities by direct mass spectrometric collision induced dissociation fragmentation of the peptide with disulfides intact. The major oxidative folding product adopts a 1-4/2-5/3-6 pattern with the following disulfide bridges: Cys5-Cys17, Cys6-Cys23, and Cys13-Cys24. The solution structure reveals that the unique N-terminal extension in µ-BuIIIB, which is also present in µ-BuIIIA and µ-BuIIIC but absent in other µ-conotoxins, forms part of a short α-helix encompassing Glu3 to Asn8. This helix is packed against the rest of the toxin and stabilized by the Cys5-Cys17 and Cys6-Cys23 disulfide bonds. As such, the side chain of Val1 is located close to the aromatic rings of Trp16 and His20, which are located on the canonical helix that displays several residues found to be essential for VGSC blockade in related µ-conotoxins. Mutations of residues 2 and 3 in the N-terminal extension enhanced the potency of µ-BuIIIB for NaV1.3. One analogue, [d-Ala2]BuIIIB, showed a 40-fold increase, making it the most potent peptide blocker of this channel characterized to date and thus a useful new tool with which to characterize this channel. On the basis of previous results for related µ-conotoxins, the dramatic effects of mutations at the N-terminus were unanticipated and suggest that further gains in potency might be achieved by additional modifications of this region.


Assuntos
Conotoxinas/química , Conotoxinas/farmacologia , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/farmacologia , Sequência de Aminoácidos , Animais , Dissulfetos/química , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.3/metabolismo , Xenopus
10.
Br J Pharmacol ; 169(1): 102-14, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23351163

RESUMO

BACKGROUND AND PURPOSE: Adult rat dorsal root ganglion (DRG) neurons normally express transcripts for five isoforms of the α-subunit of voltage-gated sodium channels: NaV 1.1, 1.6, 1.7, 1.8 and 1.9. Tetrodotoxin (TTX) readily blocks all but NaV 1.8 and 1.9, and pharmacological agents that discriminate among the TTX-sensitive NaV 1-isoforms are scarce. Recently, we used the activity profile of a panel of µ-conotoxins in blocking cloned rodent NaV 1-isoforms expressed in Xenopus laevis oocytes to conclude that action potentials of A- and C-fibres in rat sciatic nerve were, respectively, mediated primarily by NaV 1.6 and NaV 1.7. EXPERIMENTAL APPROACH: We used three µ-conotoxins, µ-TIIIA, µ-PIIIA and µ-SmIIIA, applied individually and in combinations, to pharmacologically differentiate the TTX-sensitive INa of voltage-clamped neurons acutely dissociated from adult rat DRG. We examined only small and large neurons whose respective INa were >50% and >80% TTX-sensitive. KEY RESULTS: In both small and large neurons, the ability of the toxins to block TTX-sensitive INa was µ-TIIIA < µ-PIIIA < µ-SmIIIA, with the latter blocking ≳90%. Comparison of the toxin-susceptibility profiles of the neuronal INa with recently acquired profiles of rat NaV 1-isoforms, co-expressed with various NaV ß-subunits in X. laevis oocytes, were consistent: NaV 1.1, 1.6 and 1.7 could account for all of the TTX-sensitive INa , with NaV 1.1 < NaV 1.6 < NaV 1.7 for small neurons and NaV 1.7 < NaV 1.1 < NaV 1.6 for large neurons. CONCLUSIONS AND IMPLICATIONS: Combinations of µ-conotoxins can be used to determine the probable NaV 1-isoforms underlying the INa in DRG neurons. Preliminary experiments with sympathetic neurons suggest that this approach is extendable to other neurons.


Assuntos
Conotoxinas/farmacologia , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Tetrodotoxina/farmacologia , Animais , Conotoxinas/administração & dosagem , Feminino , Gânglios Espinais/metabolismo , Masculino , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Neurônios/metabolismo , Oócitos/metabolismo , Ratos , Ratos Sprague-Dawley , Xenopus laevis
11.
Br J Pharmacol ; 168(7): 1597-610, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23146020

RESUMO

BACKGROUND AND PURPOSE: Voltage-gated sodium channels (VGSCs) are assembled from two classes of subunits, a pore-bearing α-subunit (NaV 1) and one or two accessory ß-subunits (NaV ßs). Neurons in mammals can express one or more of seven isoforms of NaV 1 and one or more of four isoforms of NaV ß. The peptide µ-conotoxins, like the guanidinium alkaloids tetrodotoxin (TTX) and saxitoxin (STX), inhibit VGSCs by blocking the pore in NaV 1. Hitherto, the effects of NaV ß-subunit co-expression on the activity of these toxins have not been comprehensively assessed. EXPERIMENTAL APPROACH: Four µ-conotoxins (µ-TIIIA, µ-PIIIA, µ-SmIIIA and µ-KIIIA), TTX and STX were tested against NaV 1.1, 1.2, 1.6 or 1.7, each co-expressed in Xenopus laevis oocytes with one of NaV ß1, ß2, ß3 or ß4 and, for NaV 1.7, binary combinations of thereof. KEY RESULTS: Co-expression of NaV ß-subunits modifies the block by µ-conotoxins: in general, NaV ß1 or ß3 co-expression tended to increase kon (in the most extreme instance by ninefold), whereas NaV ß2 or ß4 co-expression decreased kon (in the most extreme instance by 240-fold). In contrast, the block by TTX and STX was only minimally, if at all, affected by NaV ß-subunit co-expression. Tests of NaV ß1 : ß2 chimeras co-expressed with NaV 1.7 suggest that the extracellular portion of the NaV ß subunit is largely responsible for altering µ-conotoxin kinetics. CONCLUSIONS AND IMPLICATIONS: These results are the first indication that NaV ß subunit co-expression can markedly influence µ-conotoxin binding and, by extension, the outer vestibule of the pore of VGSCs. µ-Conotoxins could, in principle, be used to pharmacologically probe the NaV ß subunit composition of endogenously expressed VGSCs.


Assuntos
Conotoxinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Feminino , Cinética , Oócitos/metabolismo , Isoformas de Proteínas/metabolismo , Ratos , Subunidades beta do Canal de Sódio Disparado por Voltagem/metabolismo , Xenopus laevis
12.
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
13.
mBio ; 3(3)2012.
Artigo em Inglês | MEDLINE | ID: mdl-22647788

RESUMO

UNLABELLED: The structure, assembly, and function of the bacterial flagellum involves about 60 different proteins, many of which are selectively secreted via a specific type III secretion system (T3SS) (J. Frye et al., J. Bacteriol. 188:2233-2243, 2006). The T3SS is reported to secrete proteins at rates of up to 10,000 amino acid residues per second. In this work, we showed that the flagellar T3SS of Salmonella enterica serovar Typhimurium could be manipulated to export recombinant nonflagellar proteins through the flagellum and into the surrounding medium. We translationally fused various neuroactive peptides and proteins from snails, spiders, snakes, sea anemone, and bacteria to the flagellar secretion substrate FlgM. We found that all tested peptides of various sizes were secreted via the bacterial flagellar T3SS. We subsequently purified the recombinant µ-conotoxin SIIIA (rSIIIA) from Conus striatus by affinity chromatography and confirmed that T3SS-derived rSIIIA inhibited mammalian voltage-gated sodium channel Na(V)1.2 comparably to chemically synthesized SIIIA. IMPORTANCE: Manipulation of the flagellar secretion system bypasses the problems of inclusion body formation and cellular degradation that occur during conventional recombinant protein expression. This work serves as a proof of principle for the use of engineered bacterial cells for rapid purification of recombinant neuroactive peptides and proteins by exploiting secretion via the well-characterized flagellator type III secretion system.


Assuntos
Proteínas de Bactérias/metabolismo , Conotoxinas/metabolismo , Flagelos/metabolismo , Peptídeos/metabolismo , Salmonella typhimurium/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Conotoxinas/genética , Conotoxinas/isolamento & purificação , Flagelos/genética , Dados de Sequência Molecular , Peptídeos/genética , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Salmonella typhimurium/genética , Anêmonas-do-Mar , Caramujos , Serpentes , Aranhas
14.
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
15.
J Med Chem ; 54(21): 7558-66, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21962108

RESUMO

µ-Conotoxin KIIIA (µ-KIIIA) blocks mammalian voltage-gated sodium channels (VGSCs) and is a potent analgesic following systemic administration in mice. Previous structure-activity studies of µ-KIIIA identified a helical pharmacophore for VGSC blockade. This suggested a route for designing truncated analogues of µ-KIIIA by incorporating the key residues into an α-helical scaffold. As (i, i+4) lactam bridges constitute a proven approach for stabilizing α-helices, we designed and synthesized six truncated analogues of µ-KIIIA containing single lactam bridges at various locations. The helicity of these lactam analogues was analyzed by NMR spectroscopy, and their activities were tested against mammalian VGSC subtypes Na(V)1.1 through 1.7. Two of the analogues, Ac-cyclo9/13[Asp9,Lys13]KIIIA7-14 and Ac-cyclo9/13[Lys9,Asp13]KIIIA7-14, displayed µM activity against VGSC subtypes Na(V)1.2 and Na(V)1.6; importantly, the subtype selectivity profile for these peptides matched that of µ-KIIIA. Our study highlights structure-activity relationships within these helical mimetics and provides a basis for the design of additional truncated peptides as potential analgesics.


Assuntos
Analgésicos/química , Conotoxinas/química , Lactamas/química , Oligopeptídeos/química , Bloqueadores dos Canais de Sódio/química , Analgésicos/síntese química , Analgésicos/farmacologia , Animais , Feminino , Lactamas/síntese química , Lactamas/farmacologia , Espectroscopia de Ressonância Magnética , Camundongos , Simulação de Dinâmica Molecular , Mimetismo Molecular , Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Técnicas de Patch-Clamp , Estrutura Secundária de Proteína , Ratos , Bloqueadores dos Canais de Sódio/síntese química , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/fisiologia , Relação Estrutura-Atividade , Xenopus laevis
16.
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
17.
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
18.
ACS Med Chem Lett ; 1(4): 140-144, 2010 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-20676359

RESUMO

Despite the therapeutic promise of disulfide-rich, peptidic natural products, their discovery and structure/function studies have been hampered by inefficient oxidative folding methods for their synthesis. Here we report that converting the three disulfide-bridged mu-conopeptide KIIIA into a disulfide-depleted selenoconopeptide (by removal of a noncritical disulfide bridge and substitution of a disulfide- with a diselenide-bridge) dramatically simplified its oxidative folding while preserving the peptide's ability to block voltage-gated sodium channels. The simplicity of synthesizing disulfide-depleted selenopeptide analogs containing a single disulfide bridge allowed rapid positional scanning at Lys7 of mu-KIIIA, resulting in the identification of K7L as a mutation that improved the peptide's selectivity in blocking a neuronal (Na(v)1.2) over a muscle (Na(v)1.4) subtype of sodium channel. The disulfide-depleted selenopeptide strategy offers regioselective folding compatible with high throughput chemical synthesis and on-resin oxidation methods, and thus shows great promise to accelerate the use of disulfide-rich peptides as research tools and drugs.

19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...