Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Mol Neurosci ; 73(7-8): 664-677, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37580644

RESUMEN

The serotonin receptor subtype 5-HTR1B is widely distributed in the brain with an important role in various behavioral implications including neurological conditions and psychiatric disorders. The neuromodulatory action of 5-HTR1B largely depends upon its arrestin mediated signaling pathway. In this study, we tried to investigate the role of unusually long intracellular loop 3 (ICL3) region of the serotonin receptor 5-HTR1B in interaction with ß-arrestin1 (Arr2) to compensate for the absence of the long cytoplasmic tail. Molecular modeling and docking tools were employed to obtain a suitable molecular conformation of the ICL3 region in complex with Arr2 which dictates the specific complex formation of 5-HTR1B with Arr2. This reveals the novel molecular mechanism of phosphorylated ICL3 mediated GPCR-arrestin interaction in the absence of the long cytoplasmic tail. The in-cell disulfide cross-linking experiments and molecular dynamics simulations of the complex further validate the model of 5-HTR1B-ICL3-Arr2 complex. Two serine residues (Ser281 and Ser295) within the 5-HTR1B-ICL3 region were found to be occupying the electropositive pocket of Arr2 in our model and might be crucial for phosphorylation and specific Arr2 binding. The alignment studies of these residues showed them to be conserved only across 5-HTR1B mammalian species. Thus, our studies were able to predict a molecular conformation of 5-HTR1B-Arr2 and identify the role of long ICL3 in the signaling process which might be crucial in designing targeted drugs (biased agonists) that promote GPCR-Arr2 signaling to deter the effects of stress and anxiety-like disorders.


Asunto(s)
Receptores de Serotonina , Transducción de Señal , Humanos , Animales , beta-Arrestina 1/metabolismo , Fosforilación , Receptores de Serotonina/metabolismo , Trastornos de Ansiedad , Arrestina beta 2/metabolismo , Arrestina beta 2/farmacología , beta-Arrestinas/metabolismo , Mamíferos
2.
PNAS Nexus ; 2(3): pgad068, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37007714

RESUMEN

Store-operated calcium entry (SOCE) is a vital process aimed at refilling cellular internal Ca2+ stores and a primary cellular signaling driver for transcription factors' entry to the nucleus. SOCE-associated regulatory factor (SARAF)/TMEM66 is an endoplasmic reticulum (ER)-resident transmembrane protein that promotes SOCE inactivation and prevents Ca2+ overfilling of the cell. Here, we demonstrate that mice deficient in SARAF develop age-dependent sarcopenic obesity with decreased energy expenditure, lean mass, and locomotion without affecting food consumption. Moreover, SARAF ablation reduces hippocampal proliferation, modulates the activity of the hypothalamus-pituitary-adrenal (HPA) axis, and mediates changes in anxiety-related behaviors. Interestingly, selective SARAF ablation in the hypothalamus's paraventricular nucleus (PVN) neurons reduces old age-induced obesity and preserves locomotor activity, lean mass, and energy expenditure, suggesting a possible central control with a site-specific role for SARAF. At the cellular level, SARAF ablation in hepatocytes leads to elevated SOCE, elevated vasopressin-induced Ca2+ oscillations, and an increased mitochondrial spare respiratory capacity (SPC), thus providing insights into the cellular mechanisms that may affect the global phenotypes. These effects may be mediated via the liver X receptor (LXR) and IL-1 signaling metabolic regulators explicitly altered in SARAF ablated cells. In short, our work supports both central and peripheral roles of SARAF in regulating metabolic, behavioral, and cellular responses.

3.
Gigascience ; 112022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35640874

RESUMEN

Venoms have evolved >100 times in all major animal groups, and their components, known as toxins, have been fine-tuned over millions of years into highly effective biochemical weapons. There are many outstanding questions on the evolution of toxin arsenals, such as how venom genes originate, how venom contributes to the fitness of venomous species, and which modifications at the genomic, transcriptomic, and protein level drive their evolution. These questions have received particularly little attention outside of snakes, cone snails, spiders, and scorpions. Venom compounds have further become a source of inspiration for translational research using their diverse bioactivities for various applications. We highlight here recent advances and new strategies in modern venomics and discuss how recent technological innovations and multi-omic methods dramatically improve research on venomous animals. The study of genomes and their modifications through CRISPR and knockdown technologies will increase our understanding of how toxins evolve and which functions they have in the different ontogenetic stages during the development of venomous animals. Mass spectrometry imaging combined with spatial transcriptomics, in situ hybridization techniques, and modern computer tomography gives us further insights into the spatial distribution of toxins in the venom system and the function of the venom apparatus. All these evolutionary and biological insights contribute to more efficiently identify venom compounds, which can then be synthesized or produced in adapted expression systems to test their bioactivity. Finally, we critically discuss recent agrochemical, pharmaceutical, therapeutic, and diagnostic (so-called translational) aspects of venoms from which humans benefit.


Asunto(s)
Proteómica , Ponzoñas , Animales , Investigación , Serpientes/genética , Transcriptoma , Ponzoñas/química , Ponzoñas/genética
4.
Methods Enzymol ; 654: 169-201, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34120712

RESUMEN

Animal venom is a rich source for peptide toxins that bind and modulate the function of ion channels. Owing to their ability to bind receptor sites on the channel protein with high affinity and specificity, peptide neurotoxins have become an indispensable tool for ion channel research. Recent breakthroughs in structural biology and advances in computer simulations of biomolecules have sparked a new interest in animal toxins as probes of channel protein structure and function. Here, we focus on methods used to produce animal toxins for research purposes using recombinant expression. The specific challenges associated with heterologous production of venom peptides are discussed, and several methods targeting these issues are presented with an emphasis on E. coli based systems. An efficient protocol for the bacterial expression, folding, and purification of recombinant venom peptides is described.


Asunto(s)
Escherichia coli , Ponzoñas , Animales , Escherichia coli/genética , Escherichia coli/metabolismo , Canales Iónicos/genética , Péptidos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
J Gen Physiol ; 153(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34014250

RESUMEN

It has been reported earlier that the slow (C-type) inactivated conformation in Kv channels is stabilized by a multipoint hydrogen-bond network behind the selectivity filter. Furthermore, MD simulations revealed that structural water molecules are also involved in the formation of this network locking the selectivity filter in its inactive conformation. We found that the application of an extracellular, but not intracellular, solution based on heavy water (D2O) dramatically slowed entry into the slow inactivated state in Shaker-IR mutants (T449A, T449A/I470A, and T449K/I470C, displaying a wide range of inactivation kinetics), consistent with the proposed effect of the dynamics of structural water molecules on the conformational stability of the selectivity filter. Alternative hypotheses capable of explaining the observed effects of D2O were examined. Increased viscosity of the external solution mimicked by the addition of glycerol had a negligible effect on the rate of inactivation. In addition, the inactivation time constants of K+ currents in the outward and the inward directions in asymmetric solutions were not affected by a H2O/D2O exchange, negating an indirect effect of D2O on the rate of K+ rehydration. The elimination of the nonspecific effects of D2O on our macroscopic current measurements supports the hypothesis that the rate of structural water exchange at the region behind the selectivity filter determines the rate of slow inactivation, as proposed by molecular modeling.


Asunto(s)
Activación del Canal Iónico , Agua , Óxido de Deuterio , Enlace de Hidrógeno , Cinética
6.
J Mol Biol ; 433(17): 166957, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-33771569

RESUMEN

Many venomous organisms carry in their arsenal short polypeptides that block K+ channels in a highly selective manner. These toxins may compete with the permeating ions directly via a "plug" mechanism or indirectly via a "pore-collapse" mechanism. An alternative "lid" mechanism was proposed but remained poorly defined. Here we study the Drosophila Shaker channel block by Conkunitzin-S1 and Conkunitzin-C3, two highly similar toxins derived from cone venom. Despite their similarity, the two peptides exhibited differences in their binding poses and biophysical assays, implying discrete action modes. We show that while Conkunitzin-S1 binds tightly to the channel turret and acts via a "pore-collapse" mechanism, Conkunitzin-C3 does not contact this region. Instead, Conk-C3 uses a non-conserved Arg to divert the permeant ions and trap them in off-axis cryptic sites above the SF, a mechanism we term a "molecular-lid". Our study provides an atomic description of the "lid" K+ blocking mode and offers valuable insights for the design of therapeutics based on venom peptides.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Péptidos/farmacología , Canales de Potasio/metabolismo , Potasio/metabolismo , Venenos de Escorpión/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión/efectos de los fármacos , Biofisica/métodos , Xenopus laevis/metabolismo
7.
Cell ; 184(2): 299-301, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33482096

RESUMEN

Ion channels can tailor their activity to the particular cellular context by incorporating auxiliary subunits that are channel-type specific. In this issue of Cell, Ávalos Prado et al. now find that a well-characterized voltage-gated K+ channel auxiliary subunit can also modulate the gating of Ca2+-activated Cl- channels.


Asunto(s)
Canales Iónicos , Humanos
8.
J Physiol ; 599(2): 521-545, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33124684

RESUMEN

KEY POINTS: G-protein inwardly rectifying K+ (GIRK) channels consist of four homologous subunits (GIRK1-4) and are essential regulators of electrical excitability in the nervous system. GIRK2-null mice have been widely investigated for their distinct behaviour and altered depotentiation following long-term potentiation (LTP), whereas GIRK1 mice are less well characterized. Here we utilize a novel knockin mouse strain in which the GIRK1 subunit is fluorescently tagged with yellow fluorescent protein (YFP-GIRK1) and the GIRK1-null mouse line to investigate the role of GIRK1 in neuronal processes such as spatial learning and memory, locomotion and depotentiation following LTP. Neurons dissected from YFP-GIRK1 mice had significantly reduced potassium currents and this mouse line phenotypically resembled GIRK1-null mice, making it a 'functional knockdown' model of GIRK1-containing channels. YFP-GIRK1 and GIRK1-null mice had increased locomotion, reduced spatial learning and memory and blunted depotentiation following LTP. ABSTRACT: GIRK channels are essential for the slow inhibition of electrical activity in the nervous system and heart rate regulation via the parasympathetic system. The implications of individual GIRK isoforms in specific physiological activities are based primarily on studies conducted with GIRK-null mouse lines. Here we utilize a novel knockin mouse line in which YFP was fused in-frame to the N-terminus of GIRK1 (YFP-GIRK1) to correlate GIRK1 spatial distribution with physiological activities. These mice, however, displayed spontaneous seizure-like activity and thus were investigated for the origin of such activity. We show that GIRK tetramers containing YFP-GIRK1 are correctly assembled and trafficked to the plasma membrane, but are functionally impaired. A battery of behavioural assays conducted on YFP-GIRK1 and GIRK1-null (GIRK1-/- ) mice revealed similar phenotypes, including impaired nociception, reduced anxiety and hyperactivity in an unfamiliar environment. However, YFP-GIRK1 mice exhibited increased home-cage locomotion while GIRK1-/- mice did not. In addition, we show that the GIRK1 subunit is essential for intact spatial learning and memory and synaptic plasticity in hippocampal brain slices. This study expands our knowledge regarding the role of GIRK1 in neuronal processes and underlines the importance of GIRK1-containing heterotetramers.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Aprendizaje Espacial , Animales , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Hipocampo/metabolismo , Ratones , Plasticidad Neuronal , Neuronas/metabolismo
9.
Cell ; 178(4): 776-778, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398335

RESUMEN

Voltage sensing by ion channels is the key event enabling the generation and propagation of electrical activity in excitable cells. In this issue of Cell, Wisedchaisri et al. provide a structural view of a voltage-gated sodium channel in its resting closed conformation.


Asunto(s)
Canales Iónicos , Sodio , Conformación Molecular , Canales de Sodio Activados por Voltaje
10.
Proc Natl Acad Sci U S A ; 116(37): 18700-18709, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31444298

RESUMEN

Voltage-dependent potassium channels (Kvs) gate in response to changes in electrical membrane potential by coupling a voltage-sensing module with a K+-selective pore. Animal toxins targeting Kvs are classified as pore blockers, which physically plug the ion conduction pathway, or as gating modifiers, which disrupt voltage sensor movements. A third group of toxins blocks K+ conduction by an unknown mechanism via binding to the channel turrets. Here, we show that Conkunitzin-S1 (Cs1), a peptide toxin isolated from cone snail venom, binds at the turrets of Kv1.2 and targets a network of hydrogen bonds that govern water access to the peripheral cavities that surround the central pore. The resulting ectopic water flow triggers an asymmetric collapse of the pore by a process resembling that of inherent slow inactivation. Pore modulation by animal toxins exposes the peripheral cavity of K+ channels as a novel pharmacological target and provides a rational framework for drug design.


Asunto(s)
Membrana Celular/efectos de los fármacos , Proteínas de Drosophila/antagonistas & inhibidores , Activación del Canal Iónico/efectos de los fármacos , Canal de Potasio Kv.1.2/antagonistas & inhibidores , Venenos de Moluscos/toxicidad , Canales de Potasio de la Superfamilia Shaker/antagonistas & inhibidores , Animales , Membrana Celular/metabolismo , Cristalografía por Rayos X , Proteínas de Drosophila/genética , Proteínas de Drosophila/aislamiento & purificación , Proteínas de Drosophila/metabolismo , Diseño de Fármacos , Femenino , Enlace de Hidrógeno/efectos de los fármacos , Canal de Potasio Kv.1.2/genética , Canal de Potasio Kv.1.2/aislamiento & purificación , Canal de Potasio Kv.1.2/metabolismo , Dosificación Letal Mediana , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Venenos de Moluscos/química , Mutación , Oocitos , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Canales de Potasio de la Superfamilia Shaker/genética , Canales de Potasio de la Superfamilia Shaker/aislamiento & purificación , Canales de Potasio de la Superfamilia Shaker/metabolismo , Agua/química , Agua/metabolismo , Xenopus laevis
11.
Br J Pharmacol ; 176(17): 3161-3179, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31116876

RESUMEN

BACKGROUND AND PURPOSE: A second-generation antihistamine, terfenadine, is known to induce arrhythmia by blocking hERG channels. In this study, we have shown that terfenadine also inhibits the activity of G-protein-gated inwardly rectifying K+ (GIRK) channels, which regulate the excitability of neurons and cardiomyocytes. To clarify the underlying mechanism(s), we examined the effects of several antihistamines on GIRK channels and identified the structural determinant for the inhibition. EXPERIMENTAL APPROACH: Electrophysiological recordings were made in Xenopus oocytes and rat atrial myocytes to analyse the effects of antihistamines on various GIRK subunits (Kir 3.x). Mutagenesis analyses identified the residues critical for inhibition by terfenadine and the regulation of ion selectivity. The potential docking site of terfenadine was analysed by molecular docking. KEY RESULTS: GIRK channels containing Kir 3.1 subunits heterologously expressed in oocytes and native GIRK channels in atrial myocytes were inhibited by terfenadine and other non-sedating antihistamines. In Kir 3.1 subunits, mutation of Phe137, located in the centre of the pore helix, to the corresponding Ser in Kir 3.2 subunits reduced the inhibition by terfenadine. Introduction of an amino acid with a large side chain in Kir 3.2 subunits at Ser148 increased the inhibition. When this residue was mutated to a non-polar amino acid, the channel became permeable to Na+ . Phosphoinositide-mediated activity was also decreased by terfenadine. CONCLUSION AND IMPLICATIONS: The Phe137 residue in Kir 3.1 subunits is critical for inhibition by terfenadine. This study provides novel insights into the regulation of GIRK channels by the pore helix and information for drug design.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Antagonistas de los Receptores Histamínicos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Antagonistas de los Receptores Histamínicos/química , Masculino , Simulación del Acoplamiento Molecular , Mutación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Ratas , Ratas Wistar , Relación Estructura-Actividad , Xenopus laevis
12.
J Mol Biol ; 431(15): 2869-2883, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31082439

RESUMEN

Store-Operated Calcium Entry (SOCE) plays key roles in cell proliferation, muscle contraction, immune responses, and memory formation. The coordinated interactions of a number of proteins from the plasma and endoplasmic reticulum membranes control SOCE to replenish internal Ca2+ stores and generate intracellular Ca2+ signals. SARAF, an endoplasmic reticulum resident component of the SOCE pathway having no homology to any characterized protein, serves as an important brake on SOCE. Here, we describe the X-ray crystal structure of the SARAF luminal domain, SARAFL. This domain forms a novel 10-stranded ß-sandwich fold that includes a set of three conserved disulfide bonds, denoted the "SARAF-fold." The structure reveals a domain-swapped dimer in which the last two ß-strands (ß9 and ß10) are exchanged forming a region denoted the "SARAF luminal switch" that is essential for dimerization. Sequence comparisons reveal that the SARAF-fold is highly conserved in vertebrates and in a variety of pathologic fungi. Förster resonance energy transfer experiments using full-length SARAF validate the formation of the domain-swapped dimer in cells and demonstrate that dimerization is reversible. A designed variant lacking the SARAF luminal switch shows that the domain swapping is essential to function and indicates that the SARAF dimer accelerates SOCE inactivation.


Asunto(s)
Calcio/metabolismo , Proteínas Sensoras del Calcio Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Señalización del Calcio , Cristalografía por Rayos X , Células HEK293 , Humanos , Proteínas Sensoras del Calcio Intracelular/química , Proteínas de la Membrana/química , Modelos Moleculares , Conformación Proteica en Lámina beta , Dominios Proteicos , Pliegue de Proteína , Multimerización de Proteína
14.
J Biol Chem ; 287(36): 30719-28, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22761417

RESUMEN

Activation of voltage-gated sodium (Na(v)) channels initiates and propagates action potentials in electrically excitable cells. ß-Scorpion toxins, including toxin IV from Centruroides suffusus suffusus (CssIV), enhance activation of Na(V) channels. CssIV stabilizes the voltage sensor in domain II in its activated state via a voltage-sensor trapping mechanism. Amino acid residues required for the action of CssIV have been identified in the S1-S2 and S3-S4 extracellular loops of domain II. The extracellular loops of domain III are also involved in toxin action, but individual amino acid residues have not been identified. We used site-directed mutagenesis and voltage clamp recording to investigate amino acid residues of domain III that are involved in CssIV action. In the IIISS2-S6 loop, five substitutions at four positions altered voltage-sensor trapping by CssIV(E15A). Three substitutions (E1438A, D1445A, and D1445Y) markedly decreased voltage-sensor trapping, whereas the other two substitutions (N1436G and L1439A) increased voltage-sensor trapping. These bidirectional effects suggest that residues in IIISS2-S6 make both positive and negative interactions with CssIV. N1436G enhanced voltage-sensor trapping via increased binding affinity to the resting state, whereas L1439A increased voltage-sensor trapping efficacy. Based on these results, a three-dimensional model of the toxin-channel interaction was developed using the Rosetta modeling method. These data provide additional molecular insight into the voltage-sensor trapping mechanism of toxin action and define a three-point interaction site for ß-scorpion toxins on Na(V) channels. Binding of α- and ß-scorpion toxins to two distinct, pseudo-symmetrically organized receptor sites on Na(V) channels acts synergistically to modify channel gating and paralyze prey.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.2/metabolismo , Venenos de Escorpión/farmacología , Sustitución de Aminoácidos , Animales , Línea Celular , Activación del Canal Iónico/genética , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.2/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas
15.
J Biol Chem ; 286(40): 35209-17, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21832067

RESUMEN

Neurotoxin receptor site-3 at voltage-gated Na(+) channels is recognized by various peptide toxin inhibitors of channel inactivation. Despite extensive studies of the effects of these toxins, their mode of interaction with the channel remained to be described at the molecular level. To identify channel constituents that interact with the toxins, we exploited the opposing preferences of LqhαIT and Lqh2 scorpion α-toxins for insect and mammalian brain Na(+) channels. Construction of the DIV/S1-S2, DIV/S3-S4, DI/S5-SS1, and DI/SS2-S6 external loops of the rat brain rNa(v)1.2a channel (highly sensitive to Lqh2) in the background of the Drosophila DmNa(v)1 channel (highly sensitive to LqhαIT), and examination of toxin activity on the channel chimera expressed in Xenopus oocytes revealed a substantial decrease in LqhαIT effect, whereas Lqh2 was as effective as at rNa(v)1.2a. Further substitutions of individual loops and specific residues followed by examination of gain or loss in Lqh2 and LqhαIT activities highlighted the importance of DI/S5-S6 (pore module) and the C-terminal region of DIV/S3 (gating module) of rNa(v)1.2a for Lqh2 action and selectivity. In contrast, a single substitution of Glu-1613 to Asp at DIV/S3-S4 converted rNa(v)1.2a to high sensitivity toward LqhαIT. Comparison of depolarization-driven dissociation of Lqh2 and mutant derivatives off their binding site at rNa(v)1.2a mutant channels has suggested that the toxin core domain interacts with the gating module of DIV. These results constitute the first step in better understanding of the way scorpion α-toxins interact with voltage-gated Na(+)-channels at the molecular level.


Asunto(s)
Venenos de Escorpión/metabolismo , Escorpiones/metabolismo , Canales de Sodio/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , ADN Complementario/metabolismo , Drosophila , Conformación Molecular , Datos de Secuencia Molecular , Mutagénesis , Mutación , Neurotoxinas/metabolismo , Ratas , Anémonas de Mar , Homología de Secuencia de Aminoácido , Xenopus
16.
J Biol Chem ; 286(38): 33641-51, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21795675

RESUMEN

Voltage-gated sodium (Na(v)) channels are the molecular targets of ß-scorpion toxins, which shift the voltage dependence of activation to more negative membrane potentials by a voltage sensor-trapping mechanism. Molecular determinants of ß-scorpion toxin (CssIV) binding and action on rat brain sodium channels are located in the S1-S2 (IIS1-S2) and S3-S4 (IIS3-S4) extracellular linkers of the voltage-sensing module in domain II. In IIS1-S2, mutations of two amino acid residues (Glu(779) and Pro(782)) significantly altered the toxin effect by reducing binding affinity. In IIS3-S4, six positions surrounding the key binding determinant, Gly(845), define a hot spot of high-impact residues. Two of these substitutions (A841N and L846A) reduced voltage sensor trapping. The other three substitutions (N842R, V843A, and E844N) increased voltage sensor trapping. These bidirectional effects suggest that the IIS3-S4 loop plays a primary role in determining both toxin affinity and efficacy. A high resolution molecular model constructed with the Rosetta-Membrane modeling system reveals interactions of amino acid residues in sodium channels that are crucial for toxin action with residues in CssIV that are required for its effects. In this model, the wedge-shaped CssIV inserts between the IIS1-S2 and IIS3-S4 loops of the voltage sensor, placing key amino acid residues in position to interact with binding partners in these extracellular loops. These results provide new molecular insights into the voltage sensor-trapping model of toxin action and further define the molecular requirements for the development of antagonists that can prevent or reverse toxicity of scorpion toxins.


Asunto(s)
Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Venenos de Escorpión/química , Venenos de Escorpión/metabolismo , Canales de Sodio/química , Canales de Sodio/metabolismo , Animales , Activación del Canal Iónico , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Canal de Sodio Activado por Voltaje NAV1.2 , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Relación Estructura-Actividad
17.
J Biol Chem ; 285(40): 30531-8, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20682774

RESUMEN

Scorpion ß-toxin 4 from Centruroides suffusus suffusus (Css4) enhances the activation of voltage-gated sodium channels through a voltage sensor trapping mechanism by binding the activated state of the voltage sensor in domain II and stabilizing it in its activated conformation. Here we describe the antagonist and partial agonist properties of a mutant derivative of this toxin. Substitution of seven different amino acid residues for Glu(15) in Css4 yielded toxin derivatives with both increased and decreased affinities for binding to neurotoxin receptor site 4 on sodium channels. Css4(E15R) is unique among this set of mutants in that it retained nearly normal binding affinity but lost its functional activity for modification of sodium channel gating in our standard electrophysiological assay for voltage sensor trapping. More detailed analysis of the functional effects of Css4(E15R) revealed weak voltage sensor trapping activity, which was very rapidly reversed upon repolarization and therefore was not observed in our standard assay of toxin effects. This partial agonist activity of Css4(E15R) is observed clearly in voltage sensor trapping assays with brief (5 ms) repolarization between the conditioning prepulse and the test pulse. The effects of Css4(E15R) are fit well by a three-step model of toxin action involving concentration-dependent toxin binding to its receptor site followed by depolarization-dependent activation of the voltage sensor and subsequent voltage sensor trapping. Because it is a partial agonist with much reduced efficacy for voltage sensor trapping, Css4(E15R) can antagonize the effects of wild-type Css4 on sodium channel activation and can prevent paralysis by Css4 when injected into mice. Our results define the first partial agonist and antagonist activities for scorpion toxins and open new avenues of research toward better understanding of the structure-function relationships for toxin action on sodium channel voltage sensors and toward potential toxin-based therapeutics to prevent lethality from scorpion envenomation.


Asunto(s)
Sustitución de Aminoácidos , Activación del Canal Iónico/efectos de los fármacos , Modelos Biológicos , Venenos de Escorpión/genética , Venenos de Escorpión/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Animales , Sitios de Unión , Mordeduras y Picaduras/terapia , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Ratones , Mutación , Ratas , Ratas Wistar , Venenos de Escorpión/antagonistas & inhibidores , Venenos de Escorpión/uso terapéutico , Escorpiones
18.
J Biol Chem ; 284(31): 20684-91, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19509294

RESUMEN

The scorpion alpha-toxin Lqh2 (from Leiurus quinquestriatus hebraeus) is active at various mammalian voltage-gated sodium channels (Na(v)s) and is inactive at insect Na(v)s. To resolve the molecular basis of this preference we used the following strategy: 1) Lqh2 was expressed in recombinant form and key residues important for activity at the rat brain channel rNa(v)1.2a were identified by mutagenesis. These residues form a bipartite functional surface made of a conserved "core domain" (residues of the loops connecting the secondary structure elements of the molecule core), and a variable "NC domain" (five-residue turn and the C-tail) as was reported for other scorpion alpha-toxins. 2) The functional role of the two domains was validated by their stepwise construction on the similar scaffold of the anti-insect toxin LqhalphaIT. Analysis of the activity of the intermediate constructs highlighted the critical role of Phe(15) of the core domain in toxin potency at rNa(v)1.2a, and has suggested that the shape of the NC-domain is important for toxin efficacy. 3) Based on these findings and by comparison with other scorpion alpha-toxins we were able to eliminate the activity of Lqh2 at rNa(v)1.4 (skeletal muscle), hNa(v)1.5 (cardiac), and rNa(v)1.6 channels, with no hindrance of its activity at Na(v)1.1-1.3. These results suggest that by employing a similar approach the design of further target-selective sodium channel modifiers is imminent.


Asunto(s)
Activación del Canal Iónico , Venenos de Escorpión/química , Venenos de Escorpión/metabolismo , Canales de Sodio/metabolismo , Animales , Humanos , Modelos Moleculares , Mutagénesis , Proteínas Mutantes/metabolismo , Estructura Secundaria de Proteína , Ratas , Ratas Wistar , Propiedades de Superficie , Xenopus
19.
J Biol Chem ; 283(22): 15169-76, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18339620

RESUMEN

The bioactive surface of scorpion beta-toxins that interact with receptor site-4 at voltage-gated sodium channels is constituted of residues of the conserved betaalphabetabeta core and the C-tail. In an attempt to evaluate the extent by which residues of the toxin core contribute to bioactivity, the anti-insect and anti-mammalian beta-toxins Bj-xtrIT and Css4 were truncated at their N and C termini, resulting in miniature peptides composed essentially of the core secondary structure motives. The truncated beta-toxins (DeltaDeltaBj-xtrIT and DeltaDeltaCss4) were non-toxic and did not compete with the parental toxins on binding at receptor site-4. Surprisingly, DeltaDeltaBj-xtrIT and DeltaDeltaCss4 were capable of modulating in an allosteric manner the binding and effects of site-3 scorpion alpha-toxins in a way reminiscent of that of brevetoxins, which bind at receptor site-5. While reducing the binding and effect of the scorpion alpha-toxin Lqh2 at mammalian sodium channels, they enhanced the binding and effect of LqhalphaIT at insect sodium channels. Co-application of DeltaDeltaBj-xtrIT or DeltaDeltaCss4 with brevetoxin abolished the brevetoxin effect, although they did not compete in binding. These results denote a novel surface at DeltaDeltaBj-xtrIT and DeltaDeltaCss4 capable of interaction with sodium channels at a site other than sites 3, 4, or 5, which prior to the truncation was masked by the bioactive surface that interacts with receptor site-4. The disclosure of this hidden surface at both beta-toxins may be viewed as an exercise in "reverse evolution," providing a clue as to their evolution from a smaller ancestor of similar scaffold.


Asunto(s)
Secuencia de Aminoácidos , Evolución Molecular , Neuronas/metabolismo , Venenos de Escorpión/metabolismo , Eliminación de Secuencia , Canales de Sodio/metabolismo , Animales , Células Cultivadas , Locusta migratoria , Neuronas/citología , Unión Proteica/genética , Ratas , Ratas Wistar , Venenos de Escorpión/genética , Venenos de Escorpión/farmacología , Propiedades de Superficie
20.
Biochem J ; 406(1): 41-8, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17492942

RESUMEN

Av3 is a short peptide toxin from the sea anemone Anemonia viridis shown to be active on crustaceans and inactive on mammals. It inhibits inactivation of Na(v)s (voltage-gated Na+ channels) like the structurally dissimilar scorpion alpha-toxins and type I sea anemone toxins that bind to receptor site-3. To examine the potency and mode of interaction of Av3 with insect Na(v)s, we established a system for its expression, mutagenized it throughout, and analysed it in toxicity, binding and electrophysiological assays. The recombinant Av3 was found to be highly toxic to blowfly larvae (ED50=2.65+/-0.46 pmol/100 mg), to compete well with the site-3 toxin LqhalphaIT (from the scorpion Leiurus quinquestriatus) on binding to cockroach neuronal membranes (K(i)=21.4+/-7.1 nM), and to inhibit the inactivation of Drosophila melanogaster channel, DmNa(v)1, but not that of mammalian Na(v)s expressed in Xenopus oocytes. Moreover, like other site-3 toxins, the activity of Av3 was synergically enhanced by ligands of receptor site-4 (e.g. scorpion beta-toxins). The bioactive surface of Av3 was found to consist mainly of aromatic residues and did not resemble any of the bioactive surfaces of other site-3 toxins. These analyses have portrayed a toxin that might interact with receptor site-3 in a different fashion compared with other ligands of this site. This assumption was corroborated by a D1701R mutation in DmNa(v)1, which has been shown to abolish the activity of all other site-3 ligands, except Av3. All in all, the present study provides further evidence for the heterogeneity of receptor site-3, and raises Av3 as a unique model for design of selective anti-insect compounds.


Asunto(s)
Venenos de Cnidarios/química , Venenos de Cnidarios/farmacología , Insectos/efectos de los fármacos , Activación del Canal Iónico , Receptores de Superficie Celular/metabolismo , Anémonas de Mar/metabolismo , Canales de Sodio/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Arginina/genética , Ácido Aspártico/genética , Dicroismo Circular , Venenos de Cnidarios/metabolismo , Cucarachas/efectos de los fármacos , Drosophila melanogaster/metabolismo , Insectos/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Larva/efectos de los fármacos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Mutación/genética , Oocitos/efectos de los fármacos , Proteínas Recombinantes/química , Anémonas de Mar/química , Especificidad de la Especie , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA