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
Intervalo de ano de publicação
1.
J Neurosci ; 29(14): 4408-19, 2009 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-19357268

RESUMO

Neurogenesis in the subventricular zone (SVZ), which gives rise to new neurons in the olfactory bulb, continues throughout life but declines with increasing age. Little is known about how aging affects the intrinsic properties of the neural stem and progenitor cells (NSCs) in SVZ and the functional characteristics of their neuronal progeny. Here, we have compared the properties of NSCs isolated from embryonic lateral ganglionic eminence and adult and aged SVZ in mice using in vivo and in vitro systems, analyzed their gene expression profile, and studied their electrophysiological characteristics before and after differentiation into neurons. We show a loss of NSCs in SVZ from aged mice accompanied by reduced expression of genes for NSC markers, developmentally important transcription factors, and neurogenic factors. However, when isolated in vitro, the NSCs from SVZ of aged animals have capacity for proliferation and multilineage differentiation, including production of functional neurons, similar to that of NSCs in adult mice, albeit with lower efficacy. These properties are of major importance when considering therapeutic applications of neuronal replacement from endogenous NSCs in the injured, aged brain.


Assuntos
Envelhecimento/fisiologia , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular/fisiologia , Proliferação de Células , Neurônios/fisiologia , Células-Tronco/fisiologia , Animais , Encéfalo/citologia , Contagem de Células/métodos , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Células-Tronco/citologia
2.
Bioorg Med Chem Lett ; 19(8): 2299-304, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19282171
3.
Biochem J ; 385(Pt 1): 95-104, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15588251

RESUMO

OSK1 (alpha-KTx3.7) is a 38-residue toxin cross-linked by three disulphide bridges that was initially isolated from the venom of the Asian scorpion Orthochirus scrobiculosus. OSK1 and several structural analogues were produced by solid-phase chemical synthesis, and were tested for lethality in mice and for their efficacy in blocking a series of 14 voltage-gated and Ca2+-activated K+ channels in vitro. In the present paper, we report that OSK1 is lethal in mice by intracerebroventricular injection, with a LD50 (50% lethal dose) value of 2 microg/kg. OSK1 blocks K(v)1.1, K(v)1.2, K(v)1.3 channels potently and K(Ca)3.1 channel moderately, with IC50 values of 0.6, 5.4, 0.014 and 225 nM respectively. Structural analogues of OSK1, in which we mutated positions 16 (Glu16-->Lys) and/or 20 (Lys20-->Asp) to amino acid residues that are conserved in all other members of the alpha-KTx3 toxin family except OSK1, were also produced and tested. Among the OSK1 analogues, [K16,D20]-OSK1 (OSK1 with Glu16-->Lys and Lys20-->Asp mutations) shows an increased potency on K(v)1.3 channel, with an IC50 value of 0.003 nM, without loss of activity on K(Ca)3.1 channel. These data suggest that OSK1 or [K16,D20]-OSK1 could serve as leads for the design and production of new immunosuppressive drugs.


Assuntos
Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Venenos de Escorpião/síntese química , Venenos de Escorpião/farmacologia , Escorpiões/química , Toxinas Biológicas/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Humanos , Concentração Inibidora 50 , Injeções Intraventriculares , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/toxicidade , Venenos de Escorpião/química , Venenos de Escorpião/toxicidade , Toxinas Biológicas/síntese química , Toxinas Biológicas/química , Toxinas Biológicas/toxicidade
4.
Proteins ; 60(3): 401-11, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15971207

RESUMO

Scorpion toxins interact with their target ion channels through multiple molecular contacts. Because a "gain of function" approach has never been described to evaluate the importance of the molecular contacts in defining toxin affinity, we experimentally examined whether increasing the molecular contacts between a toxin and an ion channel directly impacts toxin affinity. For this purpose, we focused on two scorpion peptides, the well-characterized maurotoxin with its variant Pi1-like disulfide bridging (MTX(Pi1)), used as a molecular template, and butantoxin (BuTX), used as an N-terminal domain provider. BuTX is found to be 60-fold less potent than MTX(Pi1) in blocking Kv1.2 (IC(50) values of 165 nM for BuTX versus 2.8 nM for MTX(Pi1)). Removal of its N-terminal domain (nine residues) further decreases BuTX affinity for Kv1.2 by 5.6-fold, which is in agreement with docking simulation data showing the importance of this domain in BuTX-Kv1.2 interaction. Transfer of the BuTX N-terminal domain to MTX(Pi1) results in a chimera with five disulfide bridges (BuTX-MTX(Pi1)) that exhibits 22-fold greater affinity for Kv1.2 than MTX(Pi1) itself, in spite of the lower affinity of BuTX as compared to MTX(Pi1). Docking experiments performed with the 3-D structure of BuTX-MTX(Pi1) in solution, as solved by (1)H-NMR, reveal that the N-terminal domain of BuTX participates in the increased affinity for Kv1.2 through additional molecular contacts. Altogether, the data indicate that acting on molecular contacts between a toxin and a channel is an efficient strategy to modulate toxin affinity.


Assuntos
Biologia Computacional/métodos , Canal de Potássio Kv1.2/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Proteômica/métodos , Venenos de Escorpião/química , Sequência de Aminoácidos , Animais , Dicroísmo Circular , Cisteína/química , Dissulfetos/química , Eletrofisiologia , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Escorpiões , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Ácido Trifluoracético/química
5.
Biochem J ; 377(Pt 1): 25-36, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12962541

RESUMO

Pi1 is a 35-residue scorpion toxin cross-linked by four disulphide bridges that acts potently on both small-conductance Ca2+-activated (SK) and voltage-gated (Kv) K+ channel subtypes. Two approaches were used to investigate the relative contribution of the Pi1 functional dyad (Tyr-33 and Lys-24) to the toxin action: (i) the chemical synthesis of a [A24,A33]-Pi1 analogue, lacking the functional dyad, and (ii) the production of a Pi1 analogue that is phosphorylated on Tyr-33 (P-Pi1). According to molecular modelling, this phosphorylation is expected to selectively impact the two amino acid residues belonging to the functional dyad without altering the nature and three-dimensional positioning of other residues. P-Pi1 was directly produced by peptide synthesis to rule out any possibility of trace contamination by the unphosphorylated product. Both Pi1 analogues were compared with synthetic Pi1 for bioactivity. In vivo, [A24,A33]-Pi1 and P-Pi1 are lethal by intracerebroventricular injection in mice (LD50 values of 100 and 40 microg/mouse, respectively). In vitro, [A24,A33]-Pi1 and P-Pi1 compete with 125I-apamin for binding to SK channels of rat brain synaptosomes (IC50 values of 30 and 10 nM, respectively) and block rat voltage-gated Kv1.2 channels expressed in Xenopus laevis oocytes (IC50 values of 22 microM and 75 nM, respectively), whereas they are inactive on Kv1.1 or Kv1.3 channels at micromolar concentrations. Therefore, although both analogues are less active than Pi1 both in vivo and in vitro, the integrity of the Pi1 functional dyad does not appear to be a prerequisite for the recognition and binding of the toxin to the Kv1.2 channels, thereby highlighting the crucial role of other toxin residues with regard to Pi1 action on these channels. The computed simulations detailing the docking of Pi1 peptides on to the Kv1.2 channels support an unexpected key role of specific basic amino acid residues, which form a basic ring (Arg-5, Arg-12, Arg-28 and Lys-31 residues), in toxin binding.


Assuntos
Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Venenos de Escorpião/química , Venenos de Escorpião/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Cultivadas , Canal de Potássio Kv1.2 , Lisina/fisiologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/metabolismo , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/química , Estrutura Terciária de Proteína , Ratos , Venenos de Escorpião/farmacologia , Alinhamento de Sequência , Tirosina/fisiologia , Xenopus laevis
6.
Biochem J ; 377(Pt 1): 37-49, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14498829

RESUMO

CoTX1 (cobatoxin 1) is a 32-residue toxin with three disulphide bridges that has been isolated from the venom of the Mexican scorpion Centruroides noxius Hoffmann. Here we report the chemical synthesis, disulphide bridge organization, 3-D (three-dimensional) solution structure determination, pharmacology on K+ channel subtypes (voltage-gated and Ca2+-activated) and docking-simulation experiments. An enzyme-based cleavage of the synthetic folded/oxidized CoTX1 indicated half-cystine pairs between Cys3-Cys22, Cys8-Cys27 and Cys12-Cys29. The 3-D structure of CoTX1 (solved by 1H-NMR) showed that it folds according to the common alpha/beta scaffold of scorpion toxins. In vivo, CoTX1 was lethal after intracerebroventricular injection to mice (LD50 value of 0.5 microg/mouse). In vitro, CoTX1 tested on cells expressing various voltage-gated or Ca2+-activated (IKCa1) K+ channels showed potent inhibition of currents from rat K(v)1.2 ( K(d) value of 27 nM). CoTX1 also weakly competed with 125I-labelled apamin for binding to SKCa channels (small-conductance Ca2+-activated K+ channels) on rat brain synaptosomes (IC50 value of 7.2 microM). The 3-D structure of CoTX1 was used in docking experiments which suggests a key role of Arg6 or Lys10, Arg14, Arg18, Lys21 (dyad), Ile23, Asn24, Lys28 and Tyr30 (dyad) residues of CoTX1 in its interaction with the rat K(v)1.2 channel. In addition, a [Pro7,Gln9]-CoTX1 analogue (ACoTX1) was synthesized. The two residue replacements were selected aiming to restore the RPCQ motif in order to increase peptide affinity towards SKCa channels, and to alter the CoTX1 dipole moment such that it is expected to decrease peptide activity on K(v) channels. Unexpectedly, ACoTX1 exhibited an activity similar to that of CoTX1 towards SKCa channels, while it was markedly more potent on IKCa1 and several voltage-gated K+ channels.


Assuntos
Bloqueadores dos Canais de Potássio , Canais de Potássio/metabolismo , Venenos de Escorpião , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Dicroísmo Circular , Simulação por Computador , Dissulfetos/química , Humanos , Canal de Potássio Kv1.2 , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/química , Canais de Potássio Cálcio-Ativados/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ratos , Venenos de Escorpião/síntese química , Venenos de Escorpião/química , Venenos de Escorpião/metabolismo , Venenos de Escorpião/farmacologia , Homologia de Sequência de Aminoácidos
7.
Neurosci Lett ; 327(3): 153-6, 2002 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-12113900

RESUMO

The entorhinal cortex (EC) is an essential relay station in neocortical - hippocampal information transfer and memory functions. Layer II stellate and layer III pyramidal neurons show specific damage in Alzheimer's disease and epilepsy, respectively. Using whole cell patch clamp recording in rat brain slices we here demonstrate that high voltage activated Ca(2+)-currents (I(Ca)(2+)) are about 1.6-fold bigger in stellate cells than in pyramidal neurons while current density is equal in both cell types. In stellate cells I(Ca)(2+) shows stronger inactivation with depolarization, block of I(Ca)(2+) by Ni(2+) (300 and 600 microM) is more effective, and this block decreases more for currents evoked from a less negative holding potential than in layer III pyramidal neurons. These data indicate distinct molecular composition of Ca(2+)-channels and can partially explain stronger increases of [Ca(2+)](i) during 10 Hz firing activity in EC pyramidal versus stellate neurons.


Assuntos
Canais de Cálcio/fisiologia , Córtex Entorrinal/fisiologia , Neurônios/fisiologia , Animais , Canais de Cálcio/efeitos dos fármacos , Níquel/farmacologia , Técnicas de Patch-Clamp , Células Piramidais/fisiologia , Ratos , Ratos Wistar , Gânglio Estrelado/fisiologia
8.
Toxicon ; 43(8): 973-80, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15208030

RESUMO

Using human intermediate-conductance calcium-activated potassium (hIKCa1) channels as a model we aimed to characterize structural differences between maurotoxin (MTX) and charybdotoxin (CTX) and to gain new insights into the molecular determinants that define the interaction of these pore-blocking peptides with hIKCa1 channel. We report here that the block of MTX, but not of CTX on current through hIKCa1 channels is pH0 dependent. The replacement of histidine 236 from hIKCa1 channel with a smaller amino acid, cystein, did not change MTX binding affinity, however, partially affected the pH0 dependency of its block at low pH0. In contrast, CTX binding affinity to the hIKCa1_H236C channel mutant was increased suggesting that His236 might play a role in the binding of CTX, but has only a weak influence in the binding of MTX to hIKCa1 channels.


Assuntos
Charibdotoxina/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/efeitos dos fármacos , Venenos de Escorpião/farmacologia , Escorpiões/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação/efeitos dos fármacos , Charibdotoxina/metabolismo , Relação Dose-Resposta a Droga , Eletrofisiologia , Humanos , Concentração de Íons de Hidrogênio , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Bloqueadores dos Canais de Potássio/metabolismo , Canais de Potássio/metabolismo , Ligação Proteica/efeitos dos fármacos , Venenos de Escorpião/metabolismo , Alinhamento de Sequência , Transfecção
9.
Mol Pharmacol ; 69(1): 354-62, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16234482

RESUMO

OSK1, a toxin from the venom of the Asian scorpion Orthochirus scrobiculosus, is a 38-residue peptide cross-linked by three disulfide bridges. A structural analog of OSK1, [Lys(16),Asp(20)]-OSK1, was found previously to be one of the most potent blockers of the voltage-gated K(+) channel Kv1.3 hitherto characterized. Here, we demonstrate that progressive trimming of the N-terminal domain of [Lys(16),Asp(20)]-OSK1 results in marked changes in its pharmacological profile, in terms of both K(+) channel affinity and selectivity. Whereas the affinity to Kv1.1 and Kv1.3 did not change significantly, the affinity to Kv1.2 and K(Ca)3.1 was drastically reduced with the truncations. It is surprising that a striking gain in potency was observed for Kv3.2. In contrast, a truncation of the C-terminal domain, expected to partially disrupt the toxin beta-sheet structure, resulted in a significant decrease or a complete loss of activity on all channel types tested. These data highlight the value of structure-function studies on the extended N-terminal domain of [Lys(16),Asp(20)]-OSK1 to identify new analogs with unique pharmacological properties.


Assuntos
Peptídeos/farmacologia , Venenos de Escorpião/química , Toxinas Biológicas/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Humanos , Camundongos , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química , Homologia de Sequência de Aminoácidos , Toxinas Biológicas/química
10.
Mol Pharmacol ; 66(5): 1103-12, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15286210

RESUMO

Maurotoxin (MTX) is a potent blocker of human voltage-activated Kv1.2 and intermediate-conductance calcium-activated potassium channels, hIKCa1. Because its blocking affinity on both channels is similar, although the pore region of these channels show only few conserved amino acids, we aimed to characterize the binding sites of MTX in these channels. Investigating the pH(o) dependence of MTX block on current through hKv1.2 channels, we concluded that the block is less pH(o) - sensitive than for hIKCa1 channels. Using mutant cycle analysis and computer docking, we tried to identify the amino acids through which MTX binds to hKv1.2 and hIKCa1 channels. We report that MTX interacts with hKv1.2 mainly through six strong interactions. Lys(23) from MTX protrudes into the channel pore interacting with the GYGD motif, whereas Tyr(32) and Lys(7) interact with Val(381), Asp(363), and Glu(355), stabilizing the toxin onto the channel pore. Because only Val(381), Asp(363), and the GYGD motif are conserved in hIKCa1 channels, and the replacement of His(399) from hKv1.3 channels with a threonine makes this channel MTX-sensitive, we concluded that MTX binds to all three channels through the same amino acids. Glu(355), although important, is not essential in MTX recognition. A negatively charged amino acid in this position could better stabilize the toxin-channel interaction and could explain the pH(o) sensitivity of MTX block on current through hIKCa1 versus hKv1.2 channels.


Assuntos
Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Venenos de Escorpião/farmacologia , Sequência de Aminoácidos , Animais , Sítios de Ligação/efeitos dos fármacos , Células COS , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Canal de Potássio Kv1.2 , Canal de Potássio Kv1.3 , Dados de Sequência Molecular , Canais de Potássio Cálcio-Ativados , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
11.
J Biol Chem ; 277(40): 37406-13, 2002 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-12133841

RESUMO

A structural model of BgK, a sea anemone toxin, complexed with the S5-S6 region of Kv1.1, a voltage-gated potassium channel, was determined by flexible docking under distance restraints identified by a double mutant cycles approach. This structure provides the molecular basis for identifying the major determinants of the BgK-Kv1.1 channel interactions involving the BgK dyad residues Lys(25) and Tyr(26). These interactions are (i) electrostatic interactions between the extremity of Lys(25) side chain and carbonyl oxygen atoms of residues from the channel selectivity filter that may be strengthened by solvent exclusion provided by (ii) hydrophobic interactions involving BgK residues Tyr(26) and Phe(6) and Kv1.1 residue Tyr(379) whose side chain protrudes in the channel vestibule. In other Kv1 channel-BgK complexes, these interactions are likely to be conserved, implicating both conserved and variable residues from the channels. The data suggest that the conservation in sea anemone and scorpion potassium channel blockers of a functional dyad composed of a lysine, and a hydrophobic residue reflects their use of convergent binding solutions based on a crucial interplay between these important conserved interactions.


Assuntos
Venenos de Cnidários/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Venenos de Cnidários/farmacologia , Canal de Potássio Kv1.1 , Potenciais da Membrana/efeitos dos fármacos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia , Conformação Proteica , Estrutura Secundária de Proteína , Ratos , Anêmonas-do-Mar , Transcrição Gênica , Células Tumorais Cultivadas
12.
Mol Pharmacol ; 65(3): 788-801, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14978258

RESUMO

We have isolated an hSK3 isoform from a human embryonic cDNA library that we have named hSK3_ex4. This isoform contains a 15 amino acid insertion within the S5 to P-loop segment. Transcripts encoding hSK3_ex4 are coexpressed at lower levels with hSK3 in neuronal as well as in non-neuronal tissues. To investigate the pharmacokinetic properties of hSK3_ex4, we expressed the isoforms hSK3 and hSK3_ex4 in tsA cells. Both isoforms were similarly activated by cytosolic Ca2+ (hSK3, EC50=0.91 +/- 0.4 microM; hSK3_ex4, EC50=0.78 +/- 0.2 microM) and by 1-ethyl-2-benzimidazolinone (hSK3, EC50=0.17 mM; hSK3_ex4, 0.19 mM). They were both blocked by tetraethylammonium (hSK3, Kd=2.2 mM; hSK3_ex4, 2.6 mM) and showed similar permeabilities relative to K+ for Cs+ (hSK3, 0.17 +/- 0.04, n=3; hSK3_ex4, 0.17 +/- 0.05, n=3) and Rb+ (hSK3, 0.79 +/- 0.04, n=3; hSK3_ex4, 0.8 +/- 0.07, n=3). Ba2+ blocked both isoforms, and in both cases, the block was strongest at hyperpolarizing membrane potentials. However, the voltage-dependence of hSK3 was stronger than that of hSK3_ex4. The most obvious distinguishing feature of this new isoform was that whereas hSK3 was blocked by apamin (Kd=0.8 nM), scyllatoxin (Kd=2.1 nM), and d-tubocurarine (Kd=33.4 microM), hSK3_ex4 was not affected by apamin up to 100 nM, scyllatoxin up to 500 nM, and d-tubocurarine up to 500 microM. So far, isoform hSK3_ex4 forms the only small-conductance calcium-activated potassium (SK) channels, which are insensitive to the classic SK blockers.


Assuntos
Apamina/farmacologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Isoformas de Proteínas/metabolismo , Venenos de Escorpião/farmacologia , Processamento Alternativo , Bário/farmacologia , Benzimidazóis/farmacologia , Cálcio/metabolismo , Humanos , Canais de Potássio/efeitos dos fármacos , Isoformas de Proteínas/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Tetraetilamônio/farmacologia , Tubocurarina/farmacologia
13.
J Biol Chem ; 278(33): 31095-104, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12783861

RESUMO

Maurotoxin (MTX) is a 34-residue toxin that has been isolated initially from the venom of the scorpion Scorpio maurus palmatus. It presents a large number of pharmacological targets, including small conductance Ca2+-activated and voltage-gated K+ channels. Contrary to other toxins of the alpha-KTx6 family (Pi1, Pi4, Pi7, and HsTx1), MTX exhibits a unique disulfide bridge organization of the type C1-C5, C2-C6, C3-C4, and C7-C8 (instead of the conventional C1-C5, C2-C6, C3-C7, and C4-C8, herein referred to as Pi1-like) that does not prevent its folding along the classic alpha/beta scaffold of scorpion toxins. Here, we developed an innovative strategy of chemical peptide synthesis to produce an MTX variant (MTXPi1) with a conventional pattern of disulfide bridging without any alteration of the toxin chemical structure. This strategy was used solely to address the impact of half-cystine pairings on MTX structural properties and pharmacology. The data indicate that MTXPi1 displays some marked changes in affinities toward the target K+ channels. Computed docking analyses using molecular models of both MTXPi1 and the various voltage-gated K+ channel subtypes (Shaker B, Kv1.2, and Kv1.3) were found to correlate with MTXPi1 pharmacology. A functional map detailing the interaction between MTXPi1 and Shaker B channel was generated in line with docking experiments.


Assuntos
Dissulfetos/química , Venenos de Escorpião/química , Venenos de Escorpião/toxicidade , Escorpiões/química , Sequência de Aminoácidos , Animais , Apamina/metabolismo , Apamina/farmacologia , Sítios de Ligação , Ligação Competitiva , Radioisótopos do Iodo , Potenciais da Membrana/efeitos dos fármacos , Dados de Sequência Molecular , Oócitos/fisiologia , Canais de Potássio/química , Canais de Potássio/metabolismo , Estrutura Terciária de Proteína , Ratos , Venenos de Escorpião/metabolismo , Análise de Sequência de Proteína , Superfamília Shaker de Canais de Potássio , Sinaptossomos/efeitos dos fármacos , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA