Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biochemistry ; 52(30): 5103-5116, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23837633

RESUMO

We have shown previously, using confocal imaging and transport assays, that the N-terminus of sodium-dependent vitamin C transporter 2 (SVCT2) can redirect apical SVCT1 to the basolateral membrane. Here, the SVCT model was used to further characterize the basolateral targeting peptide signal. Both the length (31 amino acids) and sequence accuracy of the N-terminus of SVCT2 were found to be important in basolateral targeting activity, suggesting a structural requirement. However, the N-terminal basolateral targeting sequence did not appear to act alone, based on analyses of heterologous chimeras. Although diverse N-terminal basolateral targeting signals from multipass membrane proteins can all redirect apical protein from the same gene family to the basolateral membrane, none of the N-terminal basolateral targeting signals can redirect the transmembrane and C-terminal regions from a different gene family. Instead, the presence of these heterologous N-terminal basolateral targeting signals affected the trafficking of otherwise apical protein, causing their accumulation in a stable tubulin-like non-actin structure. Nontargeting N-terminal sequences had no effect. Similar protein retention was observed previously and in this study when the C-terminus of apical or basolateral protein was mutated. These results suggest that the N- and C-termini interact, directly or indirectly, within each gene family for basolateral targeting. Circular dichroism and two-dimensional nuclear magnetic resonance analyses both found a lack of regular secondary structure in the conserved N-terminus of SVCT2, consistent with the presence of partner(s) in the targeting unit. Our finding, a departure from the prevailing single-peptide motif model, is consistent with the evolution of basolateral transporters from the corresponding apical genes. The interaction among the N-terminus, its partner(s), and the cellular basolateral targeting machinery needs to be further elucidated.


Assuntos
Membrana Celular/metabolismo , Modelos Biológicos , Sinais Direcionadores de Proteínas , Transportadores de Sódio Acoplados à Vitamina C/metabolismo , Sequência de Aminoácidos , Animais , Polaridade Celular , Sequência Conservada , Cães , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células Madin Darby de Rim Canino , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Transportadores de Sódio Acoplados à Vitamina C/química , Transportadores de Sódio Acoplados à Vitamina C/genética
2.
Circ Res ; 102(1): 103-12, 2008 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17967786

RESUMO

Hibernating myocardium is accompanied by a downregulation in energy utilization that prevents the immediate development of ischemia during stress at the expense of an attenuated level of regional contractile function. We used a discovery based proteomic approach to identify novel regional molecular adaptations responsible for this phenomenon in subendocardial samples from swine instrumented with a chronic LAD stenosis. After 3 months (n=8), hibernating myocardium was present as reflected by reduced resting LAD flow (0.75+/-0.14 versus 1.19+/-0.14 mL x min(-1) x g(-1) in remote) and wall thickening (1.93+/-0.46 mm versus 5.46+/-0.41 mm in remote, P<0.05). Regionally altered proteins were quantified with 2D Differential-in-Gel Electrophoresis (2D-DIGE) using normal myocardium as a reference with identification of candidates using MALDI-TOF mass spectrometry. Hibernating myocardium developed a significant downregulation of many mitochondrial proteins and an upregulation of stress proteins. Of particular note, the major entry points to oxidative metabolism (eg, pyruvate dehydrogenase complex and Acyl-CoA dehydrogenase) and enzymes involved in electron transport (eg, complexes I, III, and V) were reduced (P<0.05). Multiple subunits within an enzyme complex frequently showed a concordant downregulation in abundance leading to an amplification of their cumulative effects on activity (eg, "total" LAD PDC activity was 21.9+/-3.1 versus 42.8+/-1.9 mU, P<0.05). After 5-months (n=10), changes in mitochondrial and stress proteins persisted whereas cytoskeletal proteins (eg, desmin and vimentin) normalized. These data indicate that the proteomic phenotype of hibernating myocardium is dynamic and has similarities to global changes in energy substrate metabolism and function in the advanced failing heart. These proteomic changes may limit oxidative injury and apoptosis and impact functional recovery after revascularization.


Assuntos
Metabolismo Energético/genética , Regulação da Expressão Gênica/fisiologia , Miocárdio Atordoado/genética , Proteínas/análise , Proteômica/métodos , Estresse Fisiológico/genética , Adaptação Fisiológica/genética , Animais , Doença Crônica , Modelos Animais de Doenças , Regulação para Baixo , Enzimas , Regulação Enzimológica da Expressão Gênica , Proteínas Mitocondriais , Proteínas/genética , Suínos , Regulação para Cima
3.
Toxicon ; 52(3): 489-500, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18657562

RESUMO

The peptide toxin ProTxII, recently isolated from the venom of the tarantula spider Thrixopelma pruriens, modifies gating in voltage-gated Na+ and Ca2+ channels. ProTxII is distinct from other known Na+ channel gating modifier toxins in that it affects activation, but not inactivation. It shifts activation gating positively and decreases current magnitude such that the dose-dependence of toxin action measured at a single potential reflects both effects. To test the extent to which these effects were independent, we tracked several different measures of current amplitude, voltage-dependent activation, and current kinetics in Na(V)1.5 in a range of toxin concentrations. Changes in voltage dependence and a decrease in G(max) appeared at relatively low concentrations (40-100 nM) while a positive shift in the voltage range of activation was apparent at higher toxin concentrations (> or =500 nM). Because ProTxII carries a net +4 charge we tested whether electrostatic interactions contributed to toxin action. We examined the effects of ProTxII in the presence of high extracellular Ba2+, known to screen and/or bind to surface charge. Some, but not all aspects of ProTxII modification were sensitive to the presence of Ba2+ indicating the contribution of an electrostatic, surface charge-like mechanism and supporting the idea of a multi-faceted toxin-channel interaction.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Proteínas Musculares/metabolismo , Canais de Sódio/metabolismo , Venenos de Aranha/farmacologia , Animais , Relação Dose-Resposta a Droga , Humanos , Proteínas Musculares/genética , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5 , Canais de Sódio/genética , Venenos de Aranha/química , Venenos de Aranha/metabolismo , Aranhas/fisiologia
4.
Toxicon ; 49(2): 159-70, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17095031

RESUMO

Sea anemone toxins, whose biological function is the capture of marine prey, are invaluable tools for studying the structure and function of mammalian voltage-gated sodium channels. Their high degree of specificity and selectivity have allowed for detailed analysis of inactivation gating and assignment of molecular entities responsible for this process. Because of their ability to discriminate among channel isoforms, and their high degree of structural conservation, these toxins could serve as important lead compounds for future pharmaceutical design.


Assuntos
Venenos de Cnidários/genética , Venenos de Cnidários/farmacologia , Ativação do Canal Iônico , Anêmonas-do-Mar , Canais de Sódio/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Sondas Moleculares , Dados de Sequência Molecular
5.
Toxicon ; 49(2): 194-201, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17087985

RESUMO

The tarantula venom peptides ProTx-I and ProTx-II inhibit voltage-gated sodium channels by shifting their voltage dependence of activation to a more positive potential, thus acting by a mechanism similar to that of potassium channel gating modifiers such as hanatoxin and VSTX1. ProTx-I and ProTx-II inhibit all sodium channel (Nav1) subtypes tested with similar potency and represent the first potent peptidyl inhibitors of TTX-resistant sodium channels. Like gating modifiers of potassium channels, ProTx-I and ProTx-II conform to the inhibitory cystine knot motif, and ProTx-II was demonstrated to bind to sodium channels in the closed state. Both toxins have been synthesized chemically, and ProTx-II, produced by recombinant means, has been used to map the interaction surface of the peptide with the Nav1.5 channel. In comparison, beta-scorpion toxins activate sodium channels by shifting the voltage dependence of activation to more negative potentials, and together these peptides represent valuable tools for exploring the gating mechanism of sodium channels.


Assuntos
Ativação do Canal Iônico , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Venenos de Aranha/farmacologia , Animais , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia
6.
Hear Res ; 226(1-2): 140-56, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17321087

RESUMO

To identify early changes in protein expression associated with cisplatin ototoxicity, we used two dimensional-difference gel electrophoresis (2D-DIGE) and matrix-assisted laser desorption-time-of-flight (MALDI-TOF) mass spectrometry to analyze proteins from P3 rat cochleae that were cultured for 3h with or without 1mM cisplatin. Replicate analysis of fluorescent images from six gels revealed significant (p<0.01) cisplatin-induced changes (greater than 1.5-fold) in expression of 22 cochlear proteins. These include increases in the expression of five proteins, four of which were identified as nucleobindin 1, a nuclear calcium signaling and homeostasis protein (2.1-fold), heterogeneous nuclear ribonucleoprotein C, an RNA processing protein (1.8-fold), a 55 kDa protein that is either endothelial differentiation-related factor 1 or alpha-6 tubulin (1.7-fold), and calreticulin, a calcium binding chaperone of the endoplasmic reticulum (ER, 1.6-fold). The expression of 17 proteins was significantly (p<0.01) decreased by greater than 1.5-fold. These include ribonuclease/angiogenin inhibitor 1 (1.6-fold), RAS-like, family 12 (predicted), ras association (RalGDS/AF-6) domain family 5 (4.5-fold), homologous the RAS family of GTPase signaling proteins (2.4-fold), and Protein tyrosine phosphatase domain containing 1 (predicted, 6.1-fold). We identified seven cochlear proteins with either smaller (1.2-1.5-fold) or less significant (p<0.05) cisplatin-induced changes in expression. Notably, heat shock 70 kDa protein 5 (Hspa5, Grp78, and BiP), an ER chaperone protein involved in stress response, decreased 1.7-fold. We observed changes consistent with phosphorylation in the level of isoforms of another ER stress-induced protein, glucose-regulated protein Grp58. Changes in cisplatin-induced protein expression are discussed with respect to known or hypothesized functions of the identified proteins.


Assuntos
Cisplatino/toxicidade , Cóclea/efeitos dos fármacos , Cóclea/metabolismo , Proteínas/metabolismo , Animais , Antineoplásicos/toxicidade , Proteínas de Ligação ao Cálcio/metabolismo , Cóclea/patologia , Proteínas de Ligação a DNA/metabolismo , Eletroforese em Gel Bidimensional , Proteínas de Choque Térmico/metabolismo , Ponto Isoelétrico , Chaperonas Moleculares/metabolismo , Peso Molecular , Proteínas do Tecido Nervoso , Nucleobindinas , Mapeamento de Peptídeos , Proteínas/isolamento & purificação , Proteômica , Ratos , Ratos Endogâmicos F344 , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Técnicas de Cultura de Tecidos
7.
Cell Biochem Biophys ; 38(2): 215-38, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12777715

RESUMO

Neurotoxins have served as invaluable agents for identification, purification, and functional characterization of voltage-gated ion channels. Multiple classes of these toxins, which target voltage- gated Na+ channels via high-affinity binding to distinct but allosterically coupled sites, have been identified. The toxins are chemically diverse, including guanidinium heterocycles, a variety of structurally unrelated alkaloids, and multiple families of nonhomologous polypeptides having either related or distinct functions. This review describes the biochemistry and pharmacology of these agents, and summarizes the structure-function relationships underlying their interaction with molecular targets. In addition, we explore recent advances in the use of these toxins as molecular scaffolding agents, drugs, and insecticides.


Assuntos
Ativação do Canal Iônico/fisiologia , Neurotoxinas/química , Neurotoxinas/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Venenos de Cnidários/química , Desenho de Fármacos , Matriz Extracelular/química , Inseticidas/síntese química , Inseticidas/química , Ativação do Canal Iônico/efeitos dos fármacos , Substâncias Macromoleculares , Dados de Sequência Molecular , Neurotoxinas/classificação , Neurotoxinas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Ligação Proteica , Conformação Proteica , Subunidades Proteicas , Venenos de Escorpião/química , Venenos de Aranha/química , Relação Estrutura-Atividade
8.
Toxicon ; 56(4): 624-36, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20600227

RESUMO

Toxins have been used extensively to probe the gating mechanisms of voltage-gated ion channels. Relatively few such tools are available to study the low-voltage activated T-type Ca channels, which underlie thalamic neuron firing and affect sleep, resistance to seizures, and weight gain. Here we show that ProTxII, a peptide toxin recently isolated from the venom of the tarantula spider Thrixopelma pruriens, dose-dependently inhibited Ca(V)3.1 causing a decrease in current (81.6% +/- 3.1% at -30 mV in 5 microM toxin) and a positive shift in the voltage range of activation (+34.5 mV +/- 4.4 mV). Toxin-modified currents were slower to activate and faster to deactivate and they displayed a longer lag in the onset of current, i.e. the Cole-Moore shift, consistent with the inhibition of gating transitions along the activation pathway, particularly the final opening transition. Single-channel current amplitude and total gating charge were unaffected by toxin, ruling out a change in ion flux or channel dropout as mechanisms for the decrease in macroscopic conductance. A positive shift in the voltage range of gating charge movement (+30.6 mV +/- 2.6 mV shift in the voltage of half maximal charge movement in the presence of 5 microM toxin) confirmed that ProTxII-induced gating perturbations in this channel occur at the level of the voltage sensors, and kinetic modeling based on these findings suggested that reductions in current magnitude could be largely accounted for by kinetic perturbations of activation.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/química , Venenos de Aranha/química , Bloqueadores dos Canais de Cálcio/isolamento & purificação , Canais de Cálcio Tipo T/fisiologia , Linhagem Celular , Condutividade Elétrica , Humanos , Técnicas de Patch-Clamp , Venenos de Aranha/isolamento & purificação , Venenos de Aranha/farmacologia
9.
J Biol Chem ; 282(17): 12687-97, 2007 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-17339321

RESUMO

Voltage-gated Na(+) channels are critical components in the generation of action potentials in excitable cells, but despite numerous structure-function studies on these proteins, their gating mechanism remains unclear. Peptide toxins often modify channel gating, thereby providing a great deal of information about these channels. ProTx-II is a 30-amino acid peptide toxin from the venom of the tarantula, Thrixopelma pruriens, that conforms to the inhibitory cystine knot motif and which modifies activation kinetics of Na(v) and Ca(v), but not K(v), channels. ProTx-II inhibits current by shifting the voltage dependence of activation to more depolarized potentials and, therefore, differs from the classic site 4 toxins that shift voltage dependence of activation in the opposite direction. Despite this difference in functional effects, ProTx-II has been proposed to bind to neurotoxin site 4 because it modifies activation. Here, we investigate the bioactive surface of ProTx-II by alanine-scanning the toxin and analyzing the interactions of each mutant with the cardiac isoform, Na(v)1.5. The active face of the toxin is largely composed of hydrophobic and cationic residues, joining a growing group of predominantly K(v) channel gating modifier toxins that are thought to interact with the lipid environment. In addition, we performed extensive mutagenesis of Na(v)1.5 to locate the receptor site with which ProTx-II interacts. Our data establish that, contrary to prior assumptions, ProTx-II does not bind to the previously characterized neurotoxin site 4, thus making it a novel probe of activation gating in Na(v) channels with potential to shed new light on this process.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Proteínas Musculares/metabolismo , Canais de Sódio/metabolismo , Venenos de Aranha/farmacologia , Substituição de Aminoácidos , Sítios de Ligação/genética , Linhagem Celular , Humanos , Potenciais da Membrana/efeitos dos fármacos , Mutagênese , Canal de Sódio Disparado por Voltagem NAV1.5 , Mapeamento de Peptídeos , Venenos de Aranha/genética
10.
J Biol Chem ; 281(41): 30455-62, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16912051

RESUMO

Epithelial Na+ channels (ENaCs) comprise three subunits that have been proposed to be arranged in either an alpha2betagamma or a higher ordered configuration. Each subunit has two putative membrane-spanning segments (M1 and M2), intracellular amino and carboxyl termini, and a large extracellular loop. We have used the TOXCAT assay (a reporter assay for transmembrane segment homodimerization) to identify residues within the transmembrane segments of ENaC that may participate in important structural interactions within ENaC, with which we identified a candidate site within alphaM1. We performed site-directed mutagenesis at this site and found that, although the mutants reduced channel activity, ENaC protein expression at the plasma membrane was unaffected. To deduce the role of alphaM1 in the pore structure of ENaC, we performed tryptophan-scanning mutagenesis throughout alphaM1 (residues 110-130). We found that mutations within the amino-terminal part of alphaM1 had effects on activity and selectivity with a periodicity consistent with a helical structure but no effect on channel surface expression. We also observed that mutations within the carboxyl-terminal part of alphaM1 had effects on activity and selectivity but with no apparent periodicity. Additionally, these mutants reduced channel surface expression. Our data support a model in which the amino-terminal half of alphaM1 is alpha-helical and packs against structural element(s) that contribute to the ENaC pore. Furthermore, these data suggest that the carboxyl-terminal half of alphaM1 may be helical or assume a different conformation and may be involved in tertiary interactions essential to proper channel folding or assembly. Together, our data suggest that alphaM1 is divided into two distinct regions.


Assuntos
Canais Epiteliais de Sódio/química , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Triptofano/química
11.
J Biol Chem ; 280(12): 11127-33, 2005 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-15632158

RESUMO

It has been shown recently that polypeptide toxins that modulate the gating properties of voltage-sensitive cation channels are able to bind to phospholipid membranes, leading to the suggestion that these toxins are able to access a channel-binding site that remains membrane-restricted (Lee, S.-Y., and MacKinnon, R. (2004) Nature 430, 232-235). We therefore examined the ability of anthopleurin B (ApB), a sea anemone toxin that selectively modifies inactivation kinetics of Na(V)1.x channels, and ProTx-II, a spider toxin that modifies activation kinetics of the same channels, to bind to liposomes. Whereas ProTx-II can be quantitatively depleted from solution upon incubation with phosphatidylcholine/phosphatidylserine liposomes, ApB displays no discernible phospholipid binding activity. We therefore examined the activities of structurally unrelated site 3 and site 4 toxins derived from Leiurus and Centruroides venoms, respectively, in the same assay. Like ApB, the site 3 toxin LqqV shows no lipid binding activity, whereas the site 4 toxin Centruroides toxin II, like ProTx-II, is completely bound. We conclude that toxins that modify inactivation kinetics via binding to Na(V)1.x site 3 lack the ability to bind phospholipids, whereas site 4 toxins, which modify activation, have this activity. This inherent difference suggests that the conformation of domain II more closely resembles that of the K(V)AP channel than does the conformation of domain IV.


Assuntos
Proteínas Musculares/química , Peptídeos/metabolismo , Fosfolipídeos/metabolismo , Venenos de Escorpião/metabolismo , Canais de Sódio/química , Venenos de Aranha/metabolismo , Sítios de Ligação , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas Musculares/efeitos dos fármacos , Proteínas Musculares/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Conformação Proteica , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo
12.
Biochemistry ; 43(22): 7082-9, 2004 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-15170345

RESUMO

Anthopleurin B (ApB) is a type 1 sea anemone toxin, which binds to voltage-sensitive sodium channels (Na(V)'s), thereby delaying channel inactivation. Previous work from our laboratories has demonstrated that the structurally unconstrained region involving residues 8-17 of this polypeptide, designated the Arg-14 loop, is important for full toxin affinity (Seibert et al., (2003) Biochemistry 42, 14515). Within this region, important contributions are made by residues Arg-12 and Leu-18 (Gallagher and Blumenthal, (1994) J. Biol. Chem. 269, 254; Dias-Kadambi et al., (1996) J. Biol. Chem. 271, 23828). Moreover, replacement of glycine residues found at positions 10 or 15 of the loop by alanine has been shown to have profound, isoform-selective effects on toxin-binding kinetics (Seibert et al., (2003)Biochemistry 42, 14515). To thoroughly understand the importance of this entire region, the work described here investigates the contribution of ApB residues Asn-16, Thr-17, and Ser-19 to toxin affinity and isoform selectivity. Our results demonstrate that residues within and proximal to the C terminus of the Arg-14 loop are important modulators of ApB affinity for Na(V) channels, indicating that the loop and channel site 3 are likely in close contact. A comparison of the effects of multiple replacements at each position reveals that Asn-16 and Ser-19 are involved in binding, whereas Thr-17 is not. The fact that anionic replacements for Asn-16 or Ser-19 are highly deleterious for toxin binding strongly suggests that site 3 contains either formal anionic residues or regions of high electron density, which could be formed by aromatic clusters. These data represent the first indication of the presence of such residues or regions within Na(V) site 3.


Assuntos
Substituição de Aminoácidos/genética , Asparagina/química , Venenos de Cnidários/metabolismo , Peptídeos/metabolismo , Serina/química , Canais de Sódio/metabolismo , Animais , Asparagina/genética , Asparagina/metabolismo , Sítios de Ligação , Células Cultivadas , Venenos de Cnidários/genética , Venenos de Cnidários/isolamento & purificação , Eletrofisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ligação Proteica , Anêmonas-do-Mar/química , Serina/genética , Serina/metabolismo , Eletricidade Estática
13.
Biochemistry ; 42(49): 14515-21, 2003 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-14661964

RESUMO

Anthopleurin B (ApB) is a high-affinity sea anemone neurotoxin that interacts with voltage-sensitive sodium (Na(V)) channels, causing a delay in channel inactivation. The solution structures of all known anemone toxins having this activity include a poorly defined region encompassing ApB residues 8-17, which we call the Arg-14 loop. We propose that the inherent mobility of the Arg-14 loop is necessary for the toxins' ability to maintain a high-affinity channel complex throughout the continual conformational transitions experienced by the channel during its functional cycle. We have previously shown that Arg-12, located in this loop, and Leu-18, which is adjacent, are important for ApB activity. Here, we characterized the role of two glycines located within the loop (Gly-10 and Gly-15) and an additional glycine positioned immediately C-terminal to it (Gly-20). We used site-directed replacement by alanine to assess the functional contribution to toxin binding of each of these residues singly and in combination. Gly-20 was found to be an essential toxin folding determinant; Gly-10 and Gly-15 were important for determining toxin affinity. Compared to wild-type toxin, the G10A and G15A toxins displayed significantly higher K(D) values for both cardiac (Na(V)1.5) and neuronal (Na(V)1.2) channels, although both demonstrated greater isoform discrimination for Na(V)1.5 than did wild-type ApB. For both G10A and G15A, significant Na(V) isoform differences were evident for on- and off-rates, with the most dramatic effect of a single mutation being the 467-fold reduction in the on-rate for G10A binding to Na(V)1.2, suggestive of a more accommodating binding site on Na(V)1.5 as compared to Na(V)1.2. Because alanine replacement of glycines is known to be associated with reduced backbone freedom, these results suggest an essential role for Arg-14 loop flexibility in toxin function, although a direct steric effect of the mutant methyl group cannot be excluded.


Assuntos
Substituição de Aminoácidos/genética , Arginina/genética , Venenos de Cnidários/metabolismo , Glicina/genética , Peptídeos/metabolismo , Alanina/genética , Animais , Arginina/metabolismo , Sítios de Ligação/genética , Linhagem Celular , Linhagem Celular Tumoral , Venenos de Cnidários/genética , Venenos de Cnidários/isolamento & purificação , Glicina/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Cinética , Camundongos , Modelos Moleculares , Proteínas Musculares/metabolismo , Mutagênese Sítio-Dirigida , Canal de Sódio Disparado por Voltagem NAV1.2 , Canal de Sódio Disparado por Voltagem NAV1.5 , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Patch-Clamp , Peptídeos/genética , Peptídeos/isolamento & purificação , Estrutura Terciária de Proteína/genética , Canais de Sódio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA