Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Auton Res ; 34(1): 191-201, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38064009

RESUMEN

PURPOSE: Guanfacine is an α2A-adrenergic receptor agonist, FDA-approved to treat attention-deficit hyperactivity disorder and high blood pressure, typically as an extended-release formulation up to 7 mg/day. In our dysautonomia clinic, we observed that off-label use of short-acting guanfacine at 1 mg/day facilitated symptom relief in two families with multiple members presenting with severe generalized anxiety. We also noted anecdotal improvements in associated dysautonomia symptoms such as hyperhidrosis, cognitive impairment, and palpitations. We postulated that a genetic deficit existed in these patients that might augment guanfacine susceptibility. METHODS: We used whole-exome sequencing to identify mutations in patients with shared generalized anxiety and dysautonomia symptoms. Guanfacine-induced changes in the function of voltage-gated Na+ channels were investigated using voltage-clamp electrophysiology. RESULTS: Whole-exome sequencing uncovered the p.I739V mutation in SCN9A in the proband of two nonrelated families. Moreover, guanfacine inhibited ionic currents evoked by wild-type and mutant NaV1.7 encoded by SCN9A, as well as other NaV channel subtypes to a varying degree. CONCLUSION: Our study provides further evidence for a possible pathophysiological role of NaV1.7 in anxiety and dysautonomia. Combined with off-target effects on NaV channel function, daily administration of 1 mg short-acting guanfacine may be sufficient to normalize NaV channel mutation-induced changes in sympathetic activity, perhaps aided by partial inhibition of NaV1.7 or other channel subtypes. In a broader context, expanding genetic and functional data about ion channel aberrations may enable the prospect of stratifying patients in which mutation-induced increased sympathetic tone normalization by guanfacine can support treatment strategies for anxiety and dysautonomia symptoms.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo , Guanfacina , Humanos , Guanfacina/uso terapéutico , Canal de Sodio Activado por Voltaje NAV1.7/genética , Mutación , Ansiedad/tratamiento farmacológico , Ansiedad/genética , Agonistas alfa-Adrenérgicos
2.
Proc Natl Acad Sci U S A ; 117(40): 24920-24928, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958636

RESUMEN

Australian funnel-web spiders are infamous for causing human fatalities, which are induced by venom peptides known as δ-hexatoxins (δ-HXTXs). Humans and other primates did not feature in the prey or predator spectrum during evolution of these spiders, and consequently the primate lethality of δ-HXTXs remains enigmatic. Funnel-web envenomations are mostly inflicted by male spiders that wander from their burrow in search of females during the mating season, which suggests a role for δ-HXTXs in self-defense since male spiders rarely feed during this period. Although 35 species of Australian funnel-web spiders have been described, only nine δ-HXTXs from four species have been characterized, resulting in a lack of understanding of the ecological roles and molecular evolution of δ-HXTXs. Here, by profiling venom-gland transcriptomes of 10 funnel-web species, we report 22 δ-HXTXs. Phylogenetic and evolutionary assessments reveal a remarkable sequence conservation of δ-HXTXs despite their deep evolutionary origin within funnel-web spiders, consistent with a defensive role. We demonstrate that δ-HXTX-Ar1a, the lethal toxin from the Sydney funnel-web spider Atrax robustus, induces pain in mice by inhibiting inactivation of voltage-gated sodium (NaV) channels involved in nociceptive signaling. δ-HXTX-Ar1a also inhibited inactivation of cockroach NaV channels and was insecticidal to sheep blowflies. Considering their algogenic effects in mice, potent insecticidal effects, and high levels of sequence conservation, we propose that the δ-HXTXs were repurposed from an initial insecticidal predatory function to a role in defending against nonhuman vertebrate predators by male spiders, with their lethal effects on humans being an unfortunate evolutionary coincidence.


Asunto(s)
Evolución Molecular , Neurotoxinas/genética , Poliaminas/química , Arañas/genética , Secuencia de Aminoácidos/genética , Animales , Australia , Secuencia Conservada/genética , Femenino , Humanos , Masculino , Ratones , Neurotoxinas/química , Neurotoxinas/metabolismo , Péptidos/genética , Filogenia , Poliaminas/metabolismo , Conducta Sexual Animal/fisiología , Venenos de Araña/genética , Arañas/patogenicidad , Transcriptoma/genética , Vertebrados/genética , Vertebrados/fisiología
3.
Nature ; 534(7608): 494-9, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27281198

RESUMEN

Voltage-gated sodium (Nav) channels initiate action potentials in most neurons, including primary afferent nerve fibres of the pain pathway. Local anaesthetics block pain through non-specific actions at all Nav channels, but the discovery of selective modulators would facilitate the analysis of individual subtypes of these channels and their contributions to chemical, mechanical, or thermal pain. Here we identify and characterize spider (Heteroscodra maculata) toxins that selectively activate the Nav1.1 subtype, the role of which in nociception and pain has not been elucidated. We use these probes to show that Nav1.1-expressing fibres are modality-specific nociceptors: their activation elicits robust pain behaviours without neurogenic inflammation and produces profound hypersensitivity to mechanical, but not thermal, stimuli. In the gut, high-threshold mechanosensitive fibres also express Nav1.1 and show enhanced toxin sensitivity in a mouse model of irritable bowel syndrome. Together, these findings establish an unexpected role for Nav1.1 channels in regulating the excitability of sensory nerve fibres that mediate mechanical pain.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Nocicepción/efectos de los fármacos , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Venenos de Araña/farmacología , Estrés Mecánico , Animales , Modelos Animales de Enfermedad , Femenino , Ganglios Sensoriales/citología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Síndrome del Colon Irritable/metabolismo , Masculino , Vaina de Mielina/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/química , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/metabolismo , Oocitos/metabolismo , Dolor/inducido químicamente , Dolor/metabolismo , Estructura Terciaria de Proteína , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Arañas/química , Especificidad por Sustrato/efectos de los fármacos , Temperatura
4.
Epilepsia ; 62(6): e82-e87, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33901312

RESUMEN

We identified nine patients from four unrelated families harboring three biallelic variants in SCN1B (NM_001037.5: c.136C>T; p.[Arg46Cys], c.178C>T; p.[Arg60Cys], and c.472G>A; p.[Val158Met]). All subjects presented with early infantile epileptic encephalopathy 52 (EIEE52), a rare, severe developmental and epileptic encephalopathy featuring infantile onset refractory seizures followed by developmental stagnation or regression. Because SCN1B influences neuronal excitability through modulation of voltage-gated sodium (NaV ) channel function, we examined the effects of human SCN1BR46C (ß1R46C ), SCN1BR60C (ß1R60C ), and SCN1BV158M (ß1V158M ) on the three predominant brain NaV channel subtypes NaV 1.1 (SCN1A), NaV 1.2 (SCN2A), and NaV 1.6 (SCN8A). We observed a shift toward more depolarizing potentials of conductance-voltage relationships (NaV 1.2/ß1R46C , NaV 1.2/ß1R60C , NaV 1.6/ß1R46C , NaV 1.6/ß1R60C , and NaV 1.6/ß1V158M ) and channel availability (NaV 1.1/ß1R46C , NaV 1.1/ß1V158M , NaV 1.2/ß1R46C , NaV 1.2/ß1R60C , and NaV 1.6/ß1V158M ), and detected a slower recovery from fast inactivation for NaV 1.1/ß1V158M . Combined with modeling data indicating perturbation-induced structural changes in ß1, these results suggest that the SCN1B variants reported here can disrupt normal NaV channel function in the brain, which may contribute to EIEE52.


Asunto(s)
Espasmos Infantiles/genética , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética , Canales de Sodio Activados por Voltaje/genética , Canales de Sodio Activados por Voltaje/metabolismo , Niño , Preescolar , Mapeo Cromosómico , ADN/genética , Epilepsia Refractaria/etiología , Electroencefalografía , Exoma , Femenino , Variación Genética , Humanos , Lactante , Masculino , Modelos Moleculares , Mutación Missense/genética , Linaje , Convulsiones/etiología
5.
Proc Natl Acad Sci U S A ; 115(17): 4495-4500, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29636418

RESUMEN

Gating pore currents through the voltage-sensing domains (VSDs) of the skeletal muscle voltage-gated sodium channel NaV1.4 underlie hypokalemic periodic paralysis (HypoPP) type 2. Gating modifier toxins target ion channels by modifying the function of the VSDs. We tested the hypothesis that these toxins could function as blockers of the pathogenic gating pore currents. We report that a crab spider toxin Hm-3 from Heriaeus melloteei can inhibit gating pore currents due to mutations affecting the second arginine residue in the S4 helix of VSD-I that we have found in patients with HypoPP and describe here. NMR studies show that Hm-3 partitions into micelles through a hydrophobic cluster formed by aromatic residues and reveal complex formation with VSD-I through electrostatic and hydrophobic interactions with the S3b helix and the S3-S4 extracellular loop. Our data identify VSD-I as a specific binding site for neurotoxins on sodium channels. Gating modifier toxins may constitute useful hits for the treatment of HypoPP.


Asunto(s)
Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.4/metabolismo , Neurotoxinas/toxicidad , Parálisis Periódica Hiperpotasémica/metabolismo , Estructura Secundaria de Proteína , Venenos de Araña/toxicidad , Sustitución de Aminoácidos , Animales , Femenino , Células HEK293 , Humanos , Activación del Canal Iónico , Canal de Sodio Activado por Voltaje NAV1.4/química , Canal de Sodio Activado por Voltaje NAV1.4/genética , Parálisis Periódica Hiperpotasémica/genética , Parálisis Periódica Hiperpotasémica/patología , Xenopus laevis
6.
Proc Natl Acad Sci U S A ; 114(26): 6836-6841, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28607094

RESUMEN

The Nav1.1 voltage-gated sodium channel is a critical contributor to excitability in the brain, where pathological loss of function leads to such disorders as epilepsy, Alzheimer's disease, and autism. This voltage-gated sodium (Nav) channel subtype also plays an important role in mechanical pain signaling by primary afferent somatosensory neurons. Therefore, pharmacologic modulation of Nav1.1 represents a potential strategy for treating excitability disorders of the brain and periphery. Inactivation is a complex aspect of Nav channel gating and consists of fast and slow components, each of which may involve a contribution from one or more voltage-sensing domains. Here, we exploit the Hm1a spider toxin, a Nav1.1-selective modulator, to better understand the relationship between these temporally distinct modes of inactivation and ask whether they can be distinguished pharmacologically. We show that Hm1a inhibits the gating movement of the domain IV voltage sensor (VSDIV), hindering both fast and slow inactivation and leading to an increase in Nav1.1 availability during high-frequency stimulation. In contrast, ICA-121431, a small-molecule Nav1.1 inhibitor, accelerates a subsequent VSDIV gating transition to accelerate entry into the slow inactivated state, resulting in use-dependent block. Further evidence for functional coupling between fast and slow inactivation is provided by a Nav1.1 mutant in which fast inactivation removal has complex effects on slow inactivation. Taken together, our data substantiate the key role of VSDIV in Nav channel fast and slow inactivation and demonstrate that these gating processes are sequential and coupled through VSDIV. These findings provide insight into a pharmacophore on VSDIV through which modulation of inactivation gating can inhibit or facilitate Nav1.1 function.


Asunto(s)
Acetamidas/farmacología , Activación del Canal Iónico/efectos de los fármacos , Mutación , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Venenos de Araña/farmacología , Arañas/química , Tiazoles/farmacología , Animales , Humanos , Activación del Canal Iónico/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Dominios Proteicos , Venenos de Araña/química , Xenopus laevis
7.
J Neurosci ; 37(20): 5204-5214, 2017 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-28450535

RESUMEN

Voltage-gated sodium (NaV) channels are responsible for the initiation and conduction of action potentials within primary afferents. The nine NaV channel isoforms recognized in mammals are often functionally divided into tetrodotoxin (TTX)-sensitive (TTX-s) channels (NaV1.1-NaV1.4, NaV1.6-NaV1.7) that are blocked by nanomolar concentrations and TTX-resistant (TTX-r) channels (NaV1.8 and NaV1.9) inhibited by millimolar concentrations, with NaV1.5 having an intermediate toxin sensitivity. For small-diameter primary afferent neurons, it is unclear to what extent different NaV channel isoforms are distributed along the peripheral and central branches of their bifurcated axons. To determine the relative contribution of TTX-s and TTX-r channels to action potential conduction in different axonal compartments, we investigated the effects of TTX on C-fiber-mediated compound action potentials (C-CAPs) of proximal and distal peripheral nerve segments and dorsal roots from mice and pigtail monkeys (Macaca nemestrina). In the dorsal roots and proximal peripheral nerves of mice and nonhuman primates, TTX reduced the C-CAP amplitude to 16% of the baseline. In contrast, >30% of the C-CAP was resistant to TTX in distal peripheral branches of monkeys and WT and NaV1.9-/- mice. In nerves from NaV1.8-/- mice, TTX-r C-CAPs could not be detected. These data indicate that NaV1.8 is the primary isoform underlying TTX-r conduction in distal axons of somatosensory C-fibers. Furthermore, there is a differential spatial distribution of NaV1.8 within C-fiber axons, being functionally more prominent in the most distal axons and terminal regions. The enrichment of NaV1.8 in distal axons may provide a useful target in the treatment of pain of peripheral origin.SIGNIFICANCE STATEMENT It is unclear whether individual sodium channel isoforms exert differential roles in action potential conduction along the axonal membrane of nociceptive, unmyelinated peripheral nerve fibers, but clarifying the role of sodium channel subtypes in different axonal segments may be useful for the development of novel analgesic strategies. Here, we provide evidence from mice and nonhuman primates that a substantial portion of the C-fiber compound action potential in distal peripheral nerves, but not proximal nerves or dorsal roots, is resistant to tetrodotoxin and that, in mice, this effect is mediated solely by voltage-gated sodium channel 1.8 (NaV1.8). The functional prominence of NaV1.8 within the axonal compartment immediately proximal to its termination may affect strategies targeting pain of peripheral origin.


Asunto(s)
Axones/fisiología , Canal de Sodio Activado por Voltaje NAV1.8/fisiología , Conducción Nerviosa/fisiología , Nervios Periféricos/fisiología , Piel/inervación , Tetrodotoxina/administración & dosificación , Vías Aferentes/efectos de los fármacos , Vías Aferentes/fisiología , Animales , Axones/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Macaca nemestrina , Masculino , Canal de Sodio Activado por Voltaje NAV1.8/efectos de los fármacos , Fibras Nerviosas Amielínicas , Conducción Nerviosa/efectos de los fármacos , Nervios Periféricos/efectos de los fármacos , Piel/efectos de los fármacos , Fenómenos Fisiológicos de la Piel/efectos de los fármacos , Bloqueadores del Canal de Sodio Activado por Voltaje/administración & dosificación
8.
J Physiol ; 596(10): 1863-1872, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29193176

RESUMEN

Voltage-gated sodium (NaV ) channel gating is a complex phenomenon which involves a distinct contribution of four integral voltage-sensing domains (VSDI, VSDII, VSDIII and VSDIV). Utilizing accrued pharmacological and structural insights, we build on an established chimera approach to introduce animal toxin sensitivity in each VSD of an acceptor channel by transferring in portable S3b-S4 motifs from the four VSDs of a toxin-susceptible donor channel (NaV 1.2). By doing so, we observe that in NaV 1.8, a relatively unexplored channel subtype with distinctly slow gating kinetics, VSDI-III participate in channel opening whereas VSDIV can regulate opening as well as fast inactivation. These results illustrate the effectiveness of a pharmacological approach to investigate the mechanism underlying gating of a mammalian NaV channel complex.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.8/fisiología , Toxinas Biológicas/farmacología , Animales , Humanos , Activación del Canal Iónico , Potenciales de la Membrana , Canal de Sodio Activado por Voltaje NAV1.8/efectos de los fármacos , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología
9.
Proc Natl Acad Sci U S A ; 112(8): E891-900, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25675485

RESUMEN

GABAA receptors shape synaptic transmission by modulating Cl(-) conductance across the cell membrane. Remarkably, animal toxins that specifically target GABAA receptors have not been identified. Here, we report the discovery of micrurotoxin1 (MmTX1) and MmTX2, two toxins present in Costa Rican coral snake venom that tightly bind to GABAA receptors at subnanomolar concentrations. Studies with recombinant and synthetic toxin variants on hippocampal neurons and cells expressing common receptor compositions suggest that MmTX1 and MmTX2 allosterically increase GABAA receptor susceptibility to agonist, thereby potentiating receptor opening as well as desensitization, possibly by interacting with the α(+)/ß(-) interface. Moreover, hippocampal neuron excitability measurements reveal toxin-induced transitory network inhibition, followed by an increase in spontaneous activity. In concert, toxin injections into mouse brain result in reduced basal activity between intense seizures. Altogether, we characterized two animal toxins that enhance GABAA receptor sensitivity to agonist, thereby establishing a previously unidentified class of tools to study this receptor family.


Asunto(s)
Venenos Elapídicos/farmacología , Elapidae/metabolismo , Péptidos/farmacología , Receptores de GABA-A/metabolismo , Secuencia de Aminoácidos , Animales , Venenos Elapídicos/química , Células HEK293 , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Activación del Canal Iónico/efectos de los fármacos , Cinética , Masculino , Ratones , Datos de Secuencia Molecular , Mutación/genética , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Péptidos/química , Unión Proteica/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Ratas , Receptores de GABA-A/química , Receptores de GABA-A/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Xenopus
10.
Biotechnol Bioeng ; 113(10): 2202-12, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27093300

RESUMEN

Disulfide-rich peptides isolated from cone snails are of great interest as drug leads due to their high specificity and potency toward therapeutically relevant ion channels and receptors. They commonly contain the inhibitor cystine knot (ICK) motif comprising three disulfide bonds forming a knotted core. Here we report the successful enzymatic backbone cyclization of an ICK-containing peptide κ-PVIIA, a 27-amino acid conopeptide from Conus purpurascens, using a mutated version of the bacterial transpeptidase, sortase A. Although a slight loss of activity was observed compared to native κ-PVIIA, cyclic κ-PVIIA is a functional peptide that inhibits the Shaker voltage-gated potassium (Kv) channel. Molecular modeling suggests that the decrease in potency may be related to the loss of crucial, but previously unidentified electrostatic interactions between the N-terminus of the peptide and the Shaker channel. This hypothesis was confirmed by testing an N-terminally acetylated κ-PVIIA, which shows a similar decrease in activity. We also investigated the conformational dynamics and hydrogen bond network of cyc-PVIIA, both of which are important factors to be considered for successful cyclization of peptides. We found that cyc-PVIIA has the same conformational dynamics, but different hydrogen bond network compared to those of κ-PVIIA. The ability to efficiently cyclize ICK peptides using sortase A will enable future protein engineering for this class of peptides and may help in the development of novel therapeutic molecules. Biotechnol. Bioeng. 2016;113: 2202-2212. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Aminoaciltransferasas/ultraestructura , Proteínas Bacterianas/ultraestructura , Conotoxinas/química , Caracol Conus/metabolismo , Cisteína Endopeptidasas/ultraestructura , Cistina/química , Modelos Moleculares , Canales de Potasio/ultraestructura , Aminoaciltransferasas/química , Animales , Proteínas Bacterianas/química , Sitios de Unión , Cisteína Endopeptidasas/química , Disulfuros/química , Activación Enzimática , Modelos Químicos , Péptidos/química , Canales de Potasio/química , Unión Proteica , Conformación Proteica , Pliegue de Proteína
11.
Proc Natl Acad Sci U S A ; 110(51): E5016-24, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297919

RESUMEN

Voltage-gated sodium (Nav) channels are embedded in a multicomponent membrane signaling complex that plays a crucial role in cellular excitability. Although the mechanism remains unclear, ß-subunits modify Nav channel function and cause debilitating disorders when mutated. While investigating whether ß-subunits also influence ligand interactions, we found that ß4 dramatically alters toxin binding to Nav1.2. To explore these observations further, we solved the crystal structure of the extracellular ß4 domain and identified (58)Cys as an exposed residue that, when mutated, eliminates the influence of ß4 on toxin pharmacology. Moreover, our results suggest the presence of a docking site that is maintained by a cysteine bridge buried within the hydrophobic core of ß4. Disrupting this bridge by introducing a ß1 mutation implicated in epilepsy repositions the (58)Cys-containing loop and disrupts ß4 modulation of Nav1.2. Overall, the principles emerging from this work (i) help explain tissue-dependent variations in Nav channel pharmacology; (ii) enable the mechanistic interpretation of ß-subunit-related disorders; and (iii) provide insights in designing molecules capable of correcting aberrant ß-subunit behavior.


Asunto(s)
Subunidad beta-4 de Canal de Sodio Activado por Voltaje/química , Sustitución de Aminoácidos , Animales , Cristalografía por Rayos X , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Humanos , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.2/química , Canal de Sodio Activado por Voltaje NAV1.2/genética , Canal de Sodio Activado por Voltaje NAV1.2/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/química , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/metabolismo , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/genética , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/metabolismo , Xenopus laevis
12.
Nature ; 456(7219): 202-8, 2008 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19005548

RESUMEN

Voltage-activated sodium (Na(v)) channels are crucial for the generation and propagation of nerve impulses, and as such are widely targeted by toxins and drugs. The four voltage sensors in Na(v) channels have distinct amino acid sequences, raising fundamental questions about their relative contributions to the function and pharmacology of the channel. Here we use four-fold symmetric voltage-activated potassium (K(v)) channels as reporters to examine the contributions of individual S3b-S4 paddle motifs within Na(v) channel voltage sensors to the kinetics of voltage sensor activation and to forming toxin receptors. Our results uncover binding sites for toxins from tarantula and scorpion venom on each of the four paddle motifs in Na(v) channels, and reveal how paddle-specific interactions can be used to reshape Na(v) channel activity. One paddle motif is unique in that it slows voltage sensor activation, and toxins selectively targeting this motif impede Na(v) channel inactivation. This reporter approach and the principles that emerge will be useful in developing new drugs for treating pain and Na(v) channelopathies.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Canales de Sodio/metabolismo , Secuencias de Aminoácidos , Animales , Modelos Moleculares , Mutagénesis , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Venenos de Escorpión/farmacología , Canales de Sodio/genética , Venenos de Araña/farmacología , Xenopus
13.
Handb Exp Pharmacol ; 221: 203-29, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24737238

RESUMEN

Voltage-gated sodium (Nav) channels are essential contributors to neuronal excitability, making them the most commonly targeted ion channel family by toxins found in animal venoms. These molecules can be used to probe the functional aspects of Nav channels on a molecular level and to explore their physiological role in normal and diseased tissues. This chapter summarizes our existing knowledge of the mechanisms by which animal toxins influence Nav channels as well as their potential application in designing therapeutic drugs.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Sodio/metabolismo , Toxinas Biológicas/farmacología , Canales de Sodio Activados por Voltaje/efectos de los fármacos , Animales , Humanos , Transducción de Señal/efectos de los fármacos , Canales de Sodio Activados por Voltaje/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(50): 20213-8, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22123950

RESUMEN

Palmitoylation is a common lipid modification known to regulate the functional properties of various proteins and is a vital step in the biosynthesis of voltage-activated sodium (Nav) channels. We discovered a mutation in an intracellular loop of rNav1.2a (G1079C), which results in a higher apparent affinity for externally applied PaurTx3 and ProTx-II, two voltage sensor toxins isolated from tarantula venom. To explore whether palmitoylation of the introduced cysteine underlies this observation, we compared channel susceptibility to a range of animal toxins in the absence and presence of 2-Br-palmitate, a palmitate analog that prevents palmitate incorporation into proteins, and found that palmitoylation contributes to the increased affinity of PaurTx3 and ProTx-II for G1079C. Further investigations with 2-Br-palmitate revealed that palmitoylation can regulate the gating and pharmacology of wild-type (wt) rNav1.2a. To identify rNav1.2a palmitoylation sites contributing to these phenomena, we substituted three endogenous cysteines predicted to be palmitoylated and found that the gating behavior of this triple cysteine mutant is similar to wt rNav1.2a treated with 2-Br-palmitate. As with chemically depalmitoylated rNav1.2a channels, this mutant also exhibits an increased susceptibility for PaurTx3. Additional mutagenesis experiments showed that palmitoylation of one cysteine in particular (C1182) primarily influences PaurTx3 sensitivity and may enhance the inactivation process of wt rNav1.2a. Overall, our results demonstrate that lipid modifications are capable of altering the gating and pharmacological properties of rNav1.2a.


Asunto(s)
Lipoilación , Proteínas del Tejido Nervioso/farmacología , Proteínas del Tejido Nervioso/fisiología , Canales de Sodio/farmacología , Canales de Sodio/fisiología , Animales , Colesterol/farmacología , Cisteína/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Lipoilación/efectos de los fármacos , Proteínas Mutantes/metabolismo , Mutación/genética , Canal de Sodio Activado por Voltaje NAV1.2 , Proteínas del Tejido Nervioso/genética , Ratas , Canales de Sodio/genética , Venenos de Araña/toxicidad , Xenopus
15.
Pharmacol Ther ; 245: 108416, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37061202

RESUMEN

Voltage-activated Na+ (NaV) channels are crucial contributors to rapid electrical signaling in the human body. As such, they are among the most targeted membrane proteins by clinical therapeutics and natural toxins. Several of the nine mammalian NaV channel subtypes play a documented role in pain or other sensory processes such as itch, touch, and smell. While causal relationships between these subtypes and biological function have been extensively described, the physiological role of NaV1.9 is less understood. Yet, mutations in NaV1.9 can cause striking disease phenotypes related to sensory perception such as loss or gain of pain and chronic itch. Here, we explore our current knowledge of the mechanisms by which NaV1.9 may contribute to pain and elaborate on the challenges associated with establishing links between experimental conditions and human disease. This review also discusses the lack of comprehensive insights into NaV1.9-specific pharmacology, an unfortunate situation since modulatory compounds may have tremendous potential in the clinic to treat pain or as precision tools to examine the extent of NaV1.9 participation in sensory perception processes.


Asunto(s)
Dolor , Toxinas Biológicas , Animales , Humanos , Mamíferos , Mutación , Dolor/tratamiento farmacológico , Transducción de Señal
16.
Biomed Pharmacother ; 165: 115173, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37453200

RESUMEN

Nav1.1 is an important pharmacological target as this voltage-gated sodium channel is involved in neurological and cardiac syndromes. Channel activators are actively sought to try to compensate for haploinsufficiency in several of these pathologies. Herein we used a natural source of new peptide compounds active on ion channels and screened for drugs capable to inhibit channel inactivation as a way to compensate for decreased channel function. We discovered that JzTx-34 is highly active on Nav1.1 and subsequently performed a full structure-activity relationship investigation to identify its pharmacophore. These experiments will help interpret the mechanism of action of this and formerly identified peptides as well as the future identification of new peptides. We also reveal structural determinants that make natural ICK peptides active against Nav1.1 challenging to synthesize. Altogether, the knowledge gained by this study will help facilitate the discovery and development of new compounds active on this critical ion channel target.


Asunto(s)
Péptidos , Canales de Sodio Activados por Voltaje , Humanos , Péptidos/farmacología , Péptidos/química , Relación Estructura-Actividad
17.
Br J Pharmacol ; 179(3): 473-486, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34411279

RESUMEN

BACKGROUND AND PURPOSE: Voltage-gated sodium (NaV ) channels are expressed de novo in carcinomas where their activity promotes invasiveness. Breast and colon cancer cells express the neonatal splice variant of NaV 1.5 (nNaV 1.5), which has several amino acid substitutions in the domain I voltage-sensor compared with its adult counterpart (aNaV 1.5). This study aimed to determine whether nNaV 1.5 channels could be distinguished pharmacologically from aNaV 1.5 channels. EXPERIMENTAL APPROACH: Cells expressing either nNaV 1.5 or aNaV 1.5 channels were exposed to low MW inhibitors, an antibody or natural toxins, and changes in electrophysiological parameters were measured. Stable expression in EBNA cells and transient expression in Xenopus laevis oocytes were used. Currents were recorded by whole-cell patch clamp and two-electrode voltage-clamp, respectively. KEY RESULTS: Several clinically used blockers of NaV channels (lidocaine, procaine, phenytoin, mexiletine, ranolazine, and riluzole) could not distinguish between nNaV 1.5 or aNaV 1.5 channels. However, two tarantula toxins (HaTx and ProTx-II) and a polyclonal antibody (NESOpAb) preferentially inhibited currents elicited by either nNaV 1.5 or aNaV 1.5 channels by binding to the spliced region of the channel. Furthermore, the amino acid residue at position 211 (aspartate in aNaV 1.5/lysine in nNaV 1.5), that is, the charge reversal in the spliced region of the channel, played a key role in the selectivity, especially in antibody binding. CONCLUSION AND IMPLICATIONS: We conclude that the cancer-related nNaV 1.5 channel can be distinguished pharmacologically from its nearest neighbour, aNaV 1.5 channels. Thus, it may be possible to design low MW compounds as antimetastatic drugs for non-toxic therapy of nNaV 1.5-expressing carcinomas.


Asunto(s)
Carcinoma , Venenos de Araña , Canales de Sodio Activados por Voltaje , Humanos , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Venenos de Araña/química , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Canales de Sodio Activados por Voltaje/metabolismo
18.
Nat Commun ; 13(1): 417, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35058427

RESUMEN

Photoactivatable drugs targeting ligand-gated ion channels open up new opportunities for light-guided therapeutic interventions. Photoactivable toxins targeting ion channels have the potential to control excitable cell activities with low invasiveness and high spatiotemporal precision. As proof-of-concept, we develop HwTxIV-Nvoc, a UV light-cleavable and photoactivatable peptide that targets voltage-gated sodium (NaV) channels and validate its activity in vitro in HEK293 cells, ex vivo in brain slices and in vivo on mice neuromuscular junctions. We find that HwTxIV-Nvoc enables precise spatiotemporal control of neuronal NaV channel function under all conditions tested. By creating multiple photoactivatable toxins, we demonstrate the broad applicability of this toxin-photoactivation technology.


Asunto(s)
Luz , Péptidos/toxicidad , Toxinas Biológicas/toxicidad , Canales de Sodio Activados por Voltaje/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/fisiología , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de la radiación , Ratones Endogámicos C57BL , Neuronas/fisiología , Neuronas/efectos de la radiación , Péptidos/síntesis química , Péptidos/química , Ingeniería de Proteínas , Factores de Tiempo , Rayos Ultravioleta , Pez Cebra
19.
Toxicol Lett ; 346: 16-22, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33878385

RESUMEN

The reef stonefish (Synanceia verrucosa) is a venomous fish which causes excruciatingly painful envenomations. While some research on the pathophysiology and functions of the venom have been conducted, there are still some gaps in the understanding of the venom effects due to the extreme lability of fish venom toxins and the lack of available testing platforms. Here we set out to assess new functions of the venom whilst also attempting to address some unclear pathophysiological effects from previous literature. Utilising a biolayer interferometry assay, our results highlight that the venom binds to the orthosteric site of the α-1 nicotinic acetylcholine receptor as well as the domain IV of voltage-gated Ca2+ (CaV1.2) channel mimotopes. Both these results add some clarity to the previously ambiguous literature. We further assessed the coagulotoxic effects of the venom using thromboelastography and Stago STA-R Max coagulation analyser assays. We reveal that the venom produced anticoagulant activity and significantly delayed time until clot formation of recalcified human plasma which is likely through the degradation of phospholipids. There was a difference between fresh and lyophilised venom activity toward the nicotinic acetylcholine receptor mimotopes and coagulation assays, whilst no difference was observed in the activity toward the domain IV of CaV1.2 mimotopes. This research adds further insights into the neglected area of fish venom whilst also highlighting the extreme labile nature of fish venom toxins.


Asunto(s)
Venenos de los Peces/toxicidad , Peces/fisiología , Receptores Nicotínicos/química , Animales , Sitios de Unión , Coagulación Sanguínea/efectos de los fármacos , Humanos , Plasma/química , Dominios Proteicos , Tromboelastografía
20.
Toxins (Basel) ; 13(8)2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34437420

RESUMEN

Bites from helodermatid lizards can cause pain, paresthesia, paralysis, and tachycardia, as well as other symptoms consistent with neurotoxicity. Furthermore, in vitro studies have shown that Heloderma horridum venom inhibits ion flux and blocks the electrical stimulation of skeletal muscles. Helodermatids have long been considered the only venomous lizards, but a large body of robust evidence has demonstrated venom to be a basal trait of Anguimorpha. This clade includes varanid lizards, whose bites have been reported to cause anticoagulation, pain, and occasionally paralysis and tachycardia. Despite the evolutionary novelty of these lizard venoms, their neuromuscular targets have yet to be identified, even for the iconic helodermatid lizards. Therefore, to fill this knowledge gap, the venoms of three Heloderma species (H. exasperatum, H. horridum and H. suspectum) and two Varanus species (V. salvadorii and V. varius) were investigated using Gallus gallus chick biventer cervicis nerve-muscle preparations and biolayer interferometry assays for binding to mammalian ion channels. Incubation with Heloderma venoms caused the reduction in nerve-mediated muscle twitches post initial response of avian skeletal muscle tissue preparation assays suggesting voltage-gated sodium (NaV) channel binding. Congruent with the flaccid paralysis inducing blockage of electrical stimulation in the skeletal muscle preparations, the biolayer interferometry tests with Heloderma suspectum venom revealed binding to the S3-S4 loop within voltage-sensing domain IV of the skeletal muscle channel subtype, NaV1.4. Consistent with tachycardia reported in clinical cases, the venom also bound to voltage-sensing domain IV of the cardiac smooth muscle calcium channel, CaV1.2. While Varanus varius venom did not have discernable effects in the avian tissue preparation assay at the concentration tested, in the biointerferometry assay both V. varius and V. salvadorii bound to voltage-sensing domain IV of both NaV1.4 and CaV1.2, similar to H. suspectum venom. The ability of varanid venoms to bind to mammalian ion channels but not to the avian tissue preparation suggests prey-selective actions, as did the differential potency within the Heloderma venoms for avian versus mammalian pathophysiological targets. This study thus presents the detailed characterization of Heloderma venom ion channel neurotoxicity and offers the first evidence of varanid lizard venom neurotoxicity. In addition, the data not only provide information useful to understanding the clinical effects produced by envenomations, but also reveal their utility as physiological probes, and underscore the potential utility of neglected venomous lineages in the drug design and development pipeline.


Asunto(s)
Canales de Calcio/metabolismo , Lagartos , Neurotoxinas/toxicidad , Canales de Sodio/metabolismo , Ponzoñas/toxicidad , Animales , Pollos , Técnicas In Vitro , Masculino , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/fisiología , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA