Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Ethnopharmacol ; 288: 114998, 2022 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-35063590

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Chronic pain management represents a serious healthcare problem worldwide. The use of opioid analgesics for pain has always been hampered by their side effects; in particular, the addictive liability associated with chronic use. Finding a morphine replacement has been a long-standing goal in the field of analgesia. In traditional Chinese medicine, processed Buthus martensii Karsch (BmK) scorpion has been used as a painkiller to treat chronic inflammatory arthritis and spondylitis, so called "Scorpio-analgesia". However, the molecular basis and the underline mechanism for the Scorpio-analgesia are still unclear. AIM OF THE STUDY: The study aims to investigate the molecular basis of "Scorpio analgesia" and identify novel analgesics from BmK scorpion. MATERIALS AND METHODS: In this study, the analgesic abilities were determined using formalin-, acetic acid- and complete Freund's adjuvant-induced pain models. The effect of BmK venom and processed BmK venom on Nav1.7 were detected by whole-cell voltage-clamp recordings on HEK293-hNav1.7 stable cell line. Action potentials in Dorsal root ganglion (DRG) neurons induced by Makatoxin-3-R58A were recorded in current-clamp mode. The content of Makatoxin-3 was detected using competitive enzyme-linked immunosorbent assay based on the Makatoxin-3 antibody. High performance liquid chromatography, western blot and circular dichroism spectroscopy were used to analysis the stability of Makatoxin-3. RESULTS: Here we demonstrate that Makatoxin-3, an α-like toxin in BmK scorpion venom targeting Nav1.7 is the critical component in Scorpio-analgesia. The analgesic effect of Makatoxin-3 could not be reversed by naloxone and is more potent than Nav1.7-selective inhibitors and non-steroidal anti-inflammatory drugs in inflammatory models. Moreover, a R58A mutant of Makatoxin-3 is capable of eliciting analgesia effect without inducing pain response. CONCLUSIONS: This study advances ion channel biology and proposes Nav1.7 agonists, rather than the presumed Nav1.7-only blockers, for non-narcotic relief of chronic pain.


Assuntos
Analgésicos/farmacologia , Inflamação/tratamento farmacológico , Dor/tratamento farmacológico , Venenos de Escorpião/farmacologia , Potenciais de Ação/efeitos dos fármacos , Analgésicos/isolamento & purificação , Animais , Modelos Animais de Doenças , Adjuvante de Freund , Gânglios Espinais/efeitos dos fármacos , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Dor/patologia , Agonistas do Canal de Sódio Disparado por Voltagem/isolamento & purificação , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia
2.
Neurosci Lett ; 753: 135844, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33775738

RESUMO

The two voltage gated sodium channels Nav1.7 and Nav1.8 are expressed in the peripheral nervous system and involved in various pain conditions including inflammatory and neuropathic pain. Rodent models bearing deletions or mutations of the corresponding genes, Scn9a and Scn10a, were created in order to understand the role of these channels in the pathophysiological mechanism underlying pain symptoms. This review summarizes the pain behavior profiles reported in Scn9a and Scn10a rodent models. The complete loss-of-function or knockout (KO) of Scn9a or Scn10a and the conditional KO (cKO) of Scn9a in specific cell populations were shown to decrease sensitivity to various pain stimuli. The Possum mutant mice bearing a dominant hypermorphic mutation in Scn10a revealed higher sensitivity to noxious stimuli. Several gain-of-function mutations were identified in patients with painful small fiber neuropathy. Future knowledge obtained from preclinical models bearing these mutations will allow understanding how these mutations affect pain. In addition, the review gives perspectives for creating models that better mimic patients' pain symptoms in view to developing novel analgesic strategies.


Assuntos
Analgésicos/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Dor/genética , Neuropatia de Pequenas Fibras/complicações , Analgésicos/uso terapêutico , Animais , Modelos Animais de Doenças , Mutação com Ganho de Função , Humanos , Mutação com Perda de Função , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Nociceptividade/efeitos dos fármacos , Dor/tratamento farmacológico , Ratos , Ratos Transgênicos , Neuropatia de Pequenas Fibras/tratamento farmacológico , Neuropatia de Pequenas Fibras/genética , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/uso terapêutico , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/uso terapêutico
3.
Sci Rep ; 10(1): 10730, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32612253

RESUMO

Voltage-gated Na+ (NaV) channels regulate homeostasis in bacteria and control membrane electrical excitability in mammals. Compared to their mammalian counterparts, bacterial NaV channels possess a simpler, fourfold symmetric structure and have facilitated studies of the structural basis of channel gating. However, the pharmacology of bacterial NaV remains largely unexplored. Here we systematically screened 39 NaV modulators on a bacterial channel (NaChBac) and characterized a selection of compounds on NaChBac and a mammalian channel (human NaV1.7). We found that while many compounds interact with both channels, they exhibit distinct functional effects. For example, the local anesthetics ambroxol and lidocaine block both NaV1.7 and NaChBac but affect activation and inactivation of the two channels to different extents. The voltage-sensing domain targeting toxin BDS-I increases NaV1.7 but decreases NaChBac peak currents. The pore binding toxins aconitine and veratridine block peak currents of NaV1.7 and shift activation (aconitine) and inactivation (veratridine) respectively. In NaChBac, they block the peak current by binding to the pore residue F224. Nonetheless, aconitine has no effect on activation or inactivation, while veratridine only modulates activation of NaChBac. The conservation and divergence in the pharmacology of bacterial and mammalian NaV channels provide insights into the molecular basis of channel gating and will facilitate organism-specific drug discovery.


Assuntos
Anestésicos Locais/farmacologia , Proteínas de Bactérias/metabolismo , Interações Medicamentosas , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Canais de Sódio/metabolismo , Toxinas Biológicas/farmacologia , Aconitina/farmacologia , Proteínas de Bactérias/química , Fenômenos Eletrofisiológicos , Células HEK293 , Humanos , Ativação do Canal Iônico , Canal de Sódio Disparado por Voltagem NAV1.7/química , Canais de Sódio/química , Veratridina/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia
4.
Toxins (Basel) ; 12(5)2020 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-32397386

RESUMO

Ciguatoxins (CTXs) are a group of neurotoxins responsible for the syndrome ciguatera fish poisoning (CFP) as a result of the consumption of contaminated fish. The presence of these toxins has been detected around the Pacific, Caribbean and Indian coasts. Recent reports indicate the emergence of CFP in other geographic areas, in particular in European coasts, of the Canary Islands (Spain) and Madeira (Portugal). A neuroblastoma cell line of murine origin (N2a) has been applied to assay different groups of neurotoxins, acting on voltage-gated sodium channel (VGSC) of excitable cells, N2a-MTT. The great potential of N2a-MTT as a sensitive tool for the CTXs screening is clearly recognized, notably because it allows the detection of these toxins at levels below recommended as security levels. However, the complexity of the matrix is a critical point on the application of N2a-MTT, which needs to be evaluated. The aim of this work is to provide recommendations for an implemented N2a-MTT method for CTXs determination in fish that avoids matrix effects, particularly those related to high lipid content.


Assuntos
Bioensaio , Ciguatoxinas/análise , Ciguatoxinas/farmacologia , Peixes/metabolismo , Neurônios/efeitos dos fármacos , Agonistas do Canal de Sódio Disparado por Voltagem/análise , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Potenciais da Membrana , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Ouabaína/farmacologia , Veratridina/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo
5.
Toxins (Basel) ; 12(4)2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32218140

RESUMO

Research in toxinology has created a pharmacological paradox. With an estimated 220,000 venomous animals worldwide, the study of peptidyl toxins provides a vast number of effector molecules. However, due to the complexity of the protein-protein interactions, there are fewer than ten venom-derived molecules on the market. Structural characterization and identification of post-translational modifications are essential to develop biological lead structures into pharmaceuticals. Utilizing advancements in mass spectrometry, we have created a high definition approach that fuses conventional high-resolution MS-MS with ion mobility spectrometry (HDMSE) to elucidate these primary structure characteristics. We investigated venom from ten species of "tiger" spider (Genus: Poecilotheria) and discovered they contain isobaric conformers originating from non-enzymatic Asp isomerization. One conformer pair conserved in five of ten species examined, denominated PcaTX-1a and PcaTX-1b, was found to be a 36-residue peptide with a cysteine knot, an amidated C-terminus, and isoAsp33Asp substitution. Although the isomerization of Asp has been implicated in many pathologies, this is the first characterization of Asp isomerization in a toxin and demonstrates the isomerized product's diminished physiological effects. This study establishes the value of a HDMSE approach to toxin screening and characterization.


Assuntos
Ácido Aspártico/química , Espectrometria de Mobilidade Iônica , Espectrometria de Massas , Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Neurotoxinas/farmacologia , Venenos de Aranha/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Descoberta de Drogas , Humanos , Isomerismo , Potenciais da Membrana , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Neurotoxinas/química , Ligação Proteica , Conformação Proteica , Processamento de Proteína Pós-Traducional , Venenos de Aranha/química , Relação Estrutura-Atividade , Agonistas do Canal de Sódio Disparado por Voltagem/química
6.
Mol Pharmacol ; 97(6): 377-383, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32193331

RESUMO

PF-05089771 is an aryl sulfonamide Nav1.7 channel blocker that binds to the inactivated state of Nav1.7 channels with high affinity but binds only weakly to channels in the resting state. Such aryl sulfonamide Nav1.7 channel blockers bind to the extracellular surface of the S1-S4 voltage-sensor segment of homologous Domain 4, whose movement is associated with inactivation. This binding site is different from that of classic sodium channel inhibitors like lidocaine, which also bind with higher affinity to the inactivated state than the resting state but bind at a site within the pore of the channel. The common dependence on gating state with distinct binding sites raises the possibility that inhibition by aryl sulfonamides and by classic local anesthetics might show an interaction mediated by their mutual state dependence. We tested this possibility by examining the state-dependent inhibition by PF-05089771 and lidocaine of human Nav1.7 channels expressed in human embryonic kidney 293 cells. At -80 mV, where a small fraction of channels are in an inactivated state under drug-free conditions, inhibition by PF-05089771 was both enhanced and speeded in the presence of lidocaine. The results suggest that lidocaine binding to the channel enhances PF-05089771 inhibition by altering the equilibrium between resting states (with D4S4 in the inner position) and inactivated states (with D4S4 in the outer position). The gating state-mediated interaction between the compounds illustrates a principle applicable to many state-dependent agents. SIGNIFICANCE STATEMENT: The results show that lidocaine enhances the degree and rate of inhibition of Nav1.7 channels by the aryl sulfonamide compound PF-05089771, consistent with state-dependent binding by lidocaine increasing the fraction of channels presenting a high-affinity binding site for PF-05089771 and suggesting that combinations of agents targeted to the pore-region binding site of lidocaine and the external binding site of aryl sulfonamides may have synergistic actions.


Assuntos
Lidocaína/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Éteres Fenílicos/farmacologia , Sulfonamidas/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Sinergismo Farmacológico , Células HEK293 , Humanos
7.
PLoS One ; 15(3): e0219106, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32134913

RESUMO

Dravet syndrome is caused by dominant loss-of-function mutations in SCN1A which cause reduced activity of Nav1.1 leading to lack of neuronal inhibition. On the other hand, gain-of-function mutations in SCN8A can lead to a severe epileptic encephalopathy subtype by over activating NaV1.6 channels. These observations suggest that Nav1.1 and Nav1.6 represent two opposing sides of the neuronal balance between inhibition and activation. Here, we hypothesize that Dravet syndrome may be treated by either enhancing Nav1.1 or reducing Nav1.6 activity. To test this hypothesis we generated and characterized a novel DS zebrafish model and tested new compounds that selectively activate or inhibit the human NaV1.1 or NaV1.6 channel respectively. We used CRISPR/Cas9 to generate two separate Scn1Lab knockout lines as an alternative to previous zebrafish models generated by random mutagenesis or morpholino oligomers. Using an optimized locomotor assay, spontaneous burst movements were detected that were unique to Scn1Lab knockouts and disappear when introducing human SCN1A mRNA. Besides the behavioral phenotype, Scn1Lab knockouts show sudden, electrical discharges in the brain that indicate epileptic seizures in zebrafish. Scn1Lab knockouts showed increased sensitivity to the GABA antagonist pentylenetetrazole and a reduction in whole organism GABA levels. Drug screenings further validated a Dravet syndrome phenotype. We tested the NaV1.1 activator AA43279 and two novel NaV1.6 inhibitors MV1369 and MV1312 in the Scn1Lab knockouts. Both type of compounds significantly reduced the number of spontaneous burst movements and seizure activity. Our results show that selective inhibition of NaV1.6 could be just as efficient as selective activation of NaV1.1 and these approaches could prove to be novel potential treatment strategies for Dravet syndrome and other (genetic) epilepsies. Compounds tested in zebrafish however, should always be further validated in other model systems for efficacy in mammals and to screen for potential side effects.


Assuntos
Epilepsias Mioclônicas/patologia , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Anticonvulsivantes/farmacologia , Modelos Animais de Doenças , Epilepsias Mioclônicas/metabolismo , Humanos , Locomoção/efeitos dos fármacos , Morfolinos/metabolismo , Mutagênese , Canal de Sódio Disparado por Voltagem NAV1.1/química , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Canal de Sódio Disparado por Voltagem NAV1.6/química , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pentilenotetrazol/farmacologia , Fenótipo , RNA Guia de Cinetoplastídeos/metabolismo , RNA Mensageiro/metabolismo , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética , Ácido gama-Aminobutírico/metabolismo
8.
Sci Rep ; 9(1): 16890, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31729429

RESUMO

Multiple voltage-gated Na+ (Nav) channelopathies can be ascribed to subtle changes in the Nav macromolecular complex. Fibroblast growth factor 14 (FGF14) is a functionally relevant component of the Nav1.6 channel complex, a causative link to spinocerebellar ataxia 27 (SCA27) and an emerging risk factor for neuropsychiatric disorders. Yet, how this protein:channel complex is regulated in the cell is still poorly understood. To search for key cellular pathways upstream of the FGF14:Nav1.6 complex, we have developed, miniaturized and optimized an in-cell assay in 384-well plates by stably reconstituting the FGF14:Nav1.6 complex using the split-luciferase complementation assay. We then conducted a high-throughput screening (HTS) of 267 FDA-approved compounds targeting known mediators of cellular signaling. Of the 65 hits initially detected, 24 were excluded based on counter-screening and cellular toxicity. Based on target analysis, potency and dose-response relationships, 5 compounds were subsequently repurchased for validation and confirmed as hits. Among those, the tyrosine kinase inhibitor lestaurtinib was highest ranked, exhibiting submicromolar inhibition of FGF14:Nav1.6 assembly. While providing evidence for a robust in-cell HTS platform that can be adapted to search for any channelopathy-associated regulatory proteins, these results lay the potential groundwork for repurposing cancer drugs for neuropsychopharmacology.


Assuntos
Antineoplásicos , Ensaios de Triagem em Larga Escala/métodos , Mapas de Interação de Proteínas/fisiologia , Agonistas do Canal de Sódio Disparado por Voltagem/isolamento & purificação , Bloqueadores do Canal de Sódio Disparado por Voltagem/isolamento & purificação , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Fatores de Crescimento de Fibroblastos/agonistas , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/química , Células HEK293 , Humanos , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/química , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Ligação Proteica , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo
9.
Neuron ; 104(2): 370-384.e5, 2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31537465

RESUMO

Sleep pressure increases during wake and dissipates during sleep, but the molecules and neurons that measure homeostatic sleep pressure remain poorly understood. We present a pharmacological assay in larval zebrafish that generates short-term increases in wakefulness followed by sustained rebound sleep after washout. The intensity of global neuronal activity during drug-induced wakefulness predicted the amount of subsequent rebound sleep. Whole-brain mapping with the neuronal activity marker phosphorylated extracellular signal-regulated kinase (pERK) identified preoptic Galanin (Galn)-expressing neurons as selectively active during rebound sleep, and the relative induction of galn transcripts was predictive of total rebound sleep time. Galn is required for sleep homeostasis, as galn mutants almost completely lacked rebound sleep following both pharmacologically induced neuronal activity and physical sleep deprivation. These results suggest that Galn plays a key role in responding to sleep pressure signals derived from neuronal activity and functions as an output arm of the vertebrate sleep homeostat.


Assuntos
Antagonistas GABAérgicos/farmacologia , Galanina/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Pentilenotetrazol/farmacologia , Privação do Sono/metabolismo , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos , 4-Aminopiridina/farmacologia , Aconitina/farmacologia , Animais , Cafeína/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Galanina/genética , Galanina/metabolismo , Homeostase , Mutação , Neurônios/metabolismo , Fosforilação , Bloqueadores dos Canais de Potássio/farmacologia , Área Pré-Óptica , Antagonistas de Receptores Purinérgicos P1/farmacologia , Sono/genética , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Vigília/genética , Peixe-Zebra
10.
Mol Pharm ; 16(9): 3948-3956, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31361498

RESUMO

Many oral mucosal conditions cause considerable and prolonged pain that to date has been difficult to alleviate via topical delivery, and the use of injection causes many patients dental anxiety and needle-prick pain. Therefore, developing a noninjectable drug delivery system as an alternative administration procedure may vastly improve the health and wellbeing of these patients. Recent advances in the development of mucoadhesive electrospun patches for the direct delivery of therapeutics to the oral mucosa offer a potential solution, but as yet, the release of local anesthetics from this system and their uptake by oral tissue have not been demonstrated. Here, we demonstrate the fabrication of lidocaine-loaded electrospun fiber patches, drug release, and subsequent uptake and permeation through the porcine buccal mucosa. Lidocaine HCl and lidocaine base were incorporated into the electrospun patches to evaluate the difference in drug permeation for the two drug compositions. Lidocaine released from the lidocaine HCl-containing electrospun patches was significantly quicker than from the lidocaine base patches, with double the amount of drug released from the lidocaine HCl patches in the first 15 min (0.16 ± 0.04 mg) compared to that from the lidocaine base patches (0.07 ± 0.01 mg). The permeation of lidocaine from the lidocaine HCl electrospun patches through ex vivo porcine buccal mucosa was also detected in 15 min, whereas permeation of lidocaine from the lidocaine base patch was not detected. Matrix-assisted laser desorption ionization-mass spectrometry imaging was used to investigate localization of lidocaine within the oral tissue. Lidocaine in the solution as well as from the mucoadhesive patch penetrated into the buccal mucosal tissue in a time-dependent manner and was detectable in the lamina propria after only 15 min. Moreover, the lidocaine released from lidocaine HCl electrospun patches retained biological activity, inhibiting veratridine-mediated opening of voltage-gated sodium channels in SH-SY5Y neuroblastoma cells. These data suggest that a mucoadhesive electrospun patch may be used as a vehicle for rapid uptake and sustained anesthetic drug delivery to treat or prevent oral pain.


Assuntos
Anestésicos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Lidocaína/farmacocinética , Mucosa Bucal/efeitos dos fármacos , Absorção pela Mucosa Oral/fisiologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacocinética , Administração Bucal , Anestésicos/administração & dosagem , Animais , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Dor Facial/tratamento farmacológico , Humanos , Lidocaína/administração & dosagem , Mucosa Bucal/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Suínos , Distribuição Tecidual , Veratridina/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/administração & dosagem
11.
Bioorg Med Chem Lett ; 29(6): 815-820, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30704812

RESUMO

The voltage-gated sodium channel, Nav1.1, is predominantly expressed in parvalbumin-positive fast spiking interneurons and has been genetically linked to Dravet syndrome. Starting from a high throughput screening hit isoxazole derivative 5, modifications of 5 via combinations of IonWorks and Q-patch assays successfully identified the nicotinamide derivative 4. Its increasing decay time constant (tau) of Nav1.1 currents at 0.03 µM along with significant selectivity against Nav1.2, Nav1.5, and Nav1.6 and acceptable brain exposure in mice was observed. Compound 4 is a promising Nav1.1 activator that can be used to analyze pathophysiological functions of the Nav1.1 channel towards treating various central nervous system diseases.


Assuntos
Descoberta de Drogas , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Pirrolidinas/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Células CHO , Cricetulus , Camundongos , Estrutura Molecular , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Niacinamida/síntese química , Pirrolidinas/síntese química , Pirrolidinas/química , Relação Estrutura-Atividade , Agonistas do Canal de Sódio Disparado por Voltagem/síntese química , Agonistas do Canal de Sódio Disparado por Voltagem/química
12.
Br J Pharmacol ; 175(14): 2926-2939, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29791744

RESUMO

BACKGROUND AND PURPOSE: Pharmacological agents that either inhibit or enhance flux of ions through voltage-gated sodium (Nav ) channels may provide opportunities for treatment of human health disorders. During studies to characterize agents that modulate Nav 1.3 function, we identified a compound that appears to exhibit both enhancement and inhibition of sodium ion conduction that appeared to be dependent on the gating state that the channel was in. The objective of the current study was to determine if these different modulatory effects are mediated by the same or distinct interactions with the channel. EXPERIMENTAL APPROACH: Electrophysiology and site-directed mutation were used to investigate the effects of PF-06526290 on Nav channel function. KEY RESULTS: PF-06526290 greatly slows inactivation of Nav channels in a subtype-independent manner. However, upon prolonged depolarization to induce inactivation, PF-06526290 becomes a Nav subtype-selective inhibitor. Mutation of the domain 4 voltage sensor modulates inhibition of Nav 1.3 or Nav 1.7 channels by PF-06526290 but has no effect on PF-06526290 mediated slowing of inactivation. CONCLUSIONS AND IMPLICATIONS: These findings suggest that distinct interactions may underlie the two modes of Nav channel modulation by PF-06526290 and that a single compound can affect sodium channel function in several ways.


Assuntos
Sulfonamidas/farmacologia , Tiazóis/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/fisiologia , Animais , Células CHO , Cricetulus , Gânglios Espinais , Células HEK293 , Humanos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia
13.
Neuropharmacology ; 127: 87-108, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28416444

RESUMO

Toxins and venom components that target voltage-gated sodium (NaV) channels have evolved numerous times due to the importance of this class of ion channels in the normal physiological function of peripheral and central neurons as well as cardiac and skeletal muscle. NaV channel activators in particular have been isolated from the venom of spiders, wasps, snakes, scorpions, cone snails and sea anemone and are also produced by plants, bacteria and algae. These compounds have provided key insight into the molecular structure, function and pathophysiological roles of NaV channels and are important tools due to their at times exquisite subtype-selectivity. We review the pharmacology of NaV channel activators with particular emphasis on mammalian isoforms and discuss putative applications for these compounds. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'


Assuntos
Peptídeos/farmacologia , Peçonhas/química , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Animais , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia
14.
Xenobiotica ; 47(6): 515-525, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27414285

RESUMO

1. A model of aconitine-induced bradycardia and hypotension, which is similar to aconitine poisoning in humans, was constructed in conscious rats by oral administration. 2. Blood pressure (BP) and heart rate (HR) of Sprague-Dawley rats were measured using a volume pressure recording (VPR) system. The pharmacokinetics of toxic doses of aconitine and its metabolites were analyzed using UPLC-MS/MS. 3. The HR was significantly decreased by 29% at 2 h after oral administration of 200 µg/kg aconitine. When the dose was increased to 400 µg/kg, systolic BP and diastolic BP were significantly decreased by 11% and 12% at 2 h after the administration, except when bradycardia occurred at 2 h and 4 h. The drug concentration-time curve showed a double-peak phenomenon in rats administered a 400 µg/kg dose. The AUC0-12 h value in the 400 µg/kg group significantly increased 0.8-fold compared to the 200 µg/kg group. Moreover, a high plasma concentration of 16-O-demethyaconitine was found in the rats that received two toxic doses. 4. In conclusion, bradycardia and hypotension are induced in conscious rats by a toxic dose of aconitine (400 µg/kg), and there was no significant difference in dose-normalized AUC0-12 h values between oral administrations of 200 µg/kg and that of 400 µg/kg. However, the dose-normalized Cmax and AUC0-12 h values in 200 µg/kg and 400 µg/kg groups were significantly smaller than those in 100 µg/kg group. The metabolites of aconitine, 16-O-demethyaconitine, and benzoylaconitine may also contribute to the hypotensive response.


Assuntos
Aconitina/efeitos adversos , Bradicardia/induzido quimicamente , Hipotensão/induzido quimicamente , Modelos Animais , Agonistas do Canal de Sódio Disparado por Voltagem/efeitos adversos , Aconitina/administração & dosagem , Aconitina/farmacologia , Administração Oral , Animais , Área Sob a Curva , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem , Agonistas do Canal de Sódio Disparado por Voltagem/administração & dosagem , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia
15.
Toxins (Basel) ; 8(10)2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27763551

RESUMO

Millions of years of evolution have fine-tuned the ability of venom peptides to rapidly incapacitate both prey and potential predators. Toxicofera reptiles are characterized by serous-secreting mandibular or maxillary glands with heightened levels of protein expression. These glands are the core anatomical components of the toxicoferan venom system, which exists in myriad points along an evolutionary continuum. Neofunctionalisation of toxins is facilitated by positive selection at functional hotspots on the ancestral protein and venom proteins have undergone dynamic diversification in helodermatid and varanid lizards as well as advanced snakes. A spectacular point on the venom system continuum is the long-glanded blue coral snake (Calliophis bivirgatus), a specialist feeder that preys on fast moving, venomous snakes which have both a high likelihood of prey escape but also represent significant danger to the predator itself. The maxillary venom glands of C. bivirgatus extend one quarter of the snake's body length and nestle within the rib cavity. Despite the snake's notoriety its venom has remained largely unstudied. Here we show that the venom uniquely produces spastic paralysis, in contrast to the flaccid paralysis typically produced by neurotoxic snake venoms. The toxin responsible, which we have called calliotoxin (δ-elapitoxin-Cb1a), is a three-finger toxin (3FTx). Calliotoxin shifts the voltage-dependence of NaV1.4 activation to more hyperpolarised potentials, inhibits inactivation, and produces large ramp currents, consistent with its profound effects on contractile force in an isolated skeletal muscle preparation. Voltage-gated sodium channels (NaV) are a particularly attractive pharmacological target as they are involved in almost all physiological processes including action potential generation and conduction. Accordingly, venom peptides that interfere with NaV function provide a key defensive and predatory advantage to a range of invertebrate venomous species including cone snails, scorpions, spiders, and anemones. Enhanced activation or delayed inactivation of sodium channels by toxins is associated with the extremely rapid onset of tetanic/excitatory paralysis in envenomed prey animals. A strong selection pressure exists for the evolution of such toxins where there is a high chance of prey escape. However, despite their prevalence in other venomous species, toxins causing delay of sodium channel inhibition have never previously been described in vertebrate venoms. Here we show that NaV modulators, convergent with those of invertebrates, have evolved in the venom of the long-glanded coral snake. Calliotoxin represents a functionally novel class of 3FTx and a structurally novel class of NaV toxins that will provide significant insights into the pharmacology and physiology of NaV. The toxin represents a remarkable case of functional convergence between invertebrate and vertebrate venom systems in response to similar selection pressures. These results underscore the dynamic evolution of the Toxicofera reptile system and reinforces the value of using evolution as a roadmap for biodiscovery.


Assuntos
Venenos Elapídicos/farmacologia , Elapidae , Canal de Sódio Disparado por Voltagem NAV1.4/fisiologia , Neurotoxinas/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Linhagem Celular Tumoral , Galinhas , Venenos Elapídicos/toxicidade , Células HEK293 , Humanos , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/inervação , Músculo Esquelético/fisiologia , Neurotoxinas/toxicidade , Agonistas do Canal de Sódio Disparado por Voltagem/toxicidade
16.
Mar Drugs ; 14(2)2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26891306

RESUMO

Voltage-gated sodium channels (VGSCs) are responsible for the generation of the action potential. Among nine classified VGSC subtypes (Nav1.1-Nav1.9), Nav1.7 is primarily expressed in the sensory neurons, contributing to the nociception transmission. Therefore Nav1.7 becomes a promising target for analgesic drug development. In this study, we compared the influence of an array of VGSC agonists including veratridine, BmK NT1, brevetoxin-2, deltamethrin and antillatoxin (ATX) on membrane depolarization which was detected by Fluorescence Imaging Plate Reader (FLIPR) membrane potential (FMP) blue dye. In HEK-293 cells heterologously expressing hNav1.7 α-subunit, ATX produced a robust membrane depolarization with an EC50 value of 7.8 ± 2.9 nM whereas veratridine, BmK NT1, and deltamethrin produced marginal response. Brevetoxin-2 was without effect on membrane potential change. The ATX response was completely inhibited by tetrodotoxin suggesting that the ATX response was solely derived from hNav1.7 activation, which was consistent with the results where ATX produced a negligible response in null HEK-293 cells. Six VGSC antagonists including lidocaine, lamotrigine, phenytoin, carbamazepine, riluzole, and 2-amino-6-trifluoromethylthiobenzothiazole all concentration-dependently inhibited ATX response with IC50 values comparable to that reported from patch-clamp experiments. Considered together, we demonstrate that ATX is a unique efficacious hNav1.7 activator which offers a useful probe to develop a rapid throughput screening assay to identify hNav1.7 antagonists.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Analgésicos/administração & dosagem , Analgésicos/farmacologia , Relação Dose-Resposta a Droga , Desenho de Fármacos , Descoberta de Drogas/métodos , Células HEK293 , Humanos , Concentração Inibidora 50 , Lipopeptídeos/farmacologia , Técnicas de Patch-Clamp , Peptídeos Cíclicos/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/administração & dosagem , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos
17.
Behav Brain Res ; 292: 316-22, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26099814

RESUMO

In this study, we investigated the anxiogenic-like effects of systemically administered veratrine in rat models of anxiety. In the light/dark test, veratrine (0.6 mg/kg, s.c.) significantly and dose-dependently decreased the time rats spent in and the number of entries into a light box 30 min after administration, suggesting that veratrine increases anxiety-like behaviors. These findings were also supported by results from the elevated-plus maze test and the tail-swing behavior test. In addition, veratrine (0.6 mg/kg, s.c.) significantly increased the plasma concentration of corticosterone, an endogenous biomarker for anxiety, compared to vehicle. On the basis of these results, we conclude that veratrine induces anxiogenic-like behaviors in rats. The anxiogenic-like behaviors induced by veratrine (0.6 mg/kg, s.c.) were completely abolished by co-treatment with the typical benzodiazepine anxiolytic diazepam (1 mg/kg, s.c.), when assessed in the elevated-plus maze test. Similar results were obtained with co-treatment with riluzole (10 mg/kg, p.o.), which directly affects the glutamatergic system and has recently been suggested to have anxiolytic-like effects. In conclusion, this study provides evidence that systemically administered veratrine induces anxiogenic-like behaviors in rats. We propose the veratrine model as a novel pathological animal model to explore possible candidate drugs for anxiolytics.


Assuntos
Ansiedade/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Veratrina/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Ansiolíticos/farmacologia , Diazepam/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Ratos Wistar
18.
Protein Cell ; 6(6): 443-52, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25903152

RESUMO

Voltage-gated sodium channels (VGSCs) in primary sensory neurons play a key role in transmitting pain signals to the central nervous system. BmK I, a site-3 sodium channel-specific toxin from scorpion Buthus martensi Karsch, induces pain behaviors in rats. However, the subtypes of VGSCs targeted by BmK I were not entirely clear. We therefore investigated the effects of BmK I on the current amplitude, gating and kinetic properties of Nav1.8, which is associated with neuronal hyperexcitability in DRG neurons. It was found that BmK I dose-dependently increased Nav1.8 current in small-sized (<25 µm) acutely dissociated DRG neurons, which correlated with its inhibition on both fast and slow inactivation. Moreover, voltage-dependent activation and steady-state inactivation curves of Nav1.8 were shifted in a hyperpolarized direction. Thus, BmK I reduced the threshold of neuronal excitability and increased action potential firing in DRG neurons. In conclusion, our data clearly demonstrated that BmK I modulated Nav1.8 remarkably, suggesting BmK I as a valuable probe for studying Nav1.8. And Nav1.8 is an important target related to BmK I-evoked pain.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Venenos de Escorpião/farmacologia , Escorpiões , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Compostos de Anilina/farmacologia , Animais , Tamanho Celular , Células Cultivadas , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Furanos/farmacologia , Gânglios Espinais/citologia , Cinética , Masculino , Ratos , Ratos Sprague-Dawley , Venenos de Escorpião/antagonistas & inibidores , Células Receptoras Sensoriais/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia
19.
Neuron ; 85(3): 590-601, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25619656

RESUMO

Neuronal dendritic spines have been speculated to function as independent computational units, yet evidence for active electrical computation in spines is scarce. Here we show that strictly local voltage-gated sodium channel (Nav) activation can occur during excitatory postsynaptic potentials in the spines of olfactory bulb granule cells, which we mimic and detect via combined two-photon uncaging of glutamate and calcium imaging in conjunction with whole-cell recordings. We find that local Nav activation boosts calcium entry into spines through high-voltage-activated calcium channels and accelerates postsynaptic somatic depolarization, without affecting NMDA receptor-mediated signaling. Hence, Nav-mediated boosting promotes rapid output from the reciprocal granule cell spine onto the lateral mitral cell dendrite and thus can speed up recurrent inhibition. This striking example of electrical compartmentalization both adds to the understanding of olfactory network processing and broadens the general view of spine function.


Assuntos
Espinhas Dendríticas/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Bulbo Olfatório/fisiologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/fisiologia , Animais , Espinhas Dendríticas/efeitos dos fármacos , Feminino , Masculino , Bulbo Olfatório/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar
20.
Eur J Pharmacol ; 746: 333-43, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25261036

RESUMO

Brugada syndrome (BrS), which causes arrhythmias that lead to sudden cardiac death, is linked to loss-of-function mutations that affect sodium channels. Here, we investigate the rescue effect of alpha-allocryptopine (All) from Chinese herbal medicine in a T353I mutation of SCN5A, which combines trafficking abnormalities with Brugada syndrome. SCN5A-T353I expressed in HEK293 cells showed a small peak current (I(peak)) of only 59.6% of WT and an observably sustained current (I(sus)). We found that All strongly enhanced the I(peak) of the T353I channel by enhancing the plasma membrane (PM) expression of Nav1.5 and rescued defective trafficking after co-incubation with HEK293 cells that carry mutation channel 24 h. It is also beneficial to increase the I(peak) of the T353I mutation by All by prolonging the closed-state inactivation (CSI) process and shortening the recovery from inactivation of the T353I mutation. Interestingly, the I(sus) of T353I was significantly inhibited by All, which reduces the occurrence of LQT syndrome 3 (LQT3). We provide evidence that All can rescue the trafficking deficiencies and restore the cellular electrophysiological characteristics of SCN5A-T353I. This feature of All may benefit patients with the BrS-associated Nav1.5 channel and might have other potential therapeutic effects.


Assuntos
Alcaloides de Berberina/farmacologia , Síndrome de Brugada/genética , Membrana Celular/efeitos dos fármacos , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Fármacos Neuromusculares/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Substituição de Aminoácidos , Antiarrítmicos/farmacologia , Síndrome de Brugada/metabolismo , Membrana Celular/metabolismo , Genes Reporter/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Microscopia Confocal , Mutagênese Sítio-Dirigida , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Técnicas de Patch-Clamp , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...