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











Base de dados
Intervalo de ano de publicação
1.
Biochem Pharmacol ; 181: 114148, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32663452

RESUMO

Chronic pain is a common and often debilitating condition. Existing treatments are either inefficacious or associated with a wide range of side effects. The progress on developing safer and more effective analgesics has been slow, in large part due to our limited understanding of the physiological mechanisms underlying pain in different diseases. Generation and propagation of action potentials is a central component of pain sensation and voltage-gated sodium channels (NaVs) play a critical role in this process. In particular, the NaV subtype 1.7, has emerged as a promising universal target for the treatment of pain. Recently, a spider venom peptide, µ-TRTX-Pn3a, was found to be a highly selective inhibitor of NaV1.7. Here, we report the first recombinant expression method for Pn3a in a bacterial host, which provides an inexpensive route to production. Furthermore, we have developed a method for bio-conjugation of our recombinantly produced Pn3a via sortase A-mediated ligation, providing avenues for further pre-clinical development. We demonstrate how heterologous expression in bacteria enables facile isotope labelling of Pn3a, which allowed us to study the membrane binding properties of the peptide by high-resolution solution-state nuclear magnetic resonance (NMR) spectroscopy using a recently developed lipid nanodisc system. The heteronuclear NMR data indicate that the C-terminal region of the peptide undergoes a conformational change upon lipid binding. The membrane binding properties of Pn3a are further validated using isothermal titration calorimetry (ITC), which revealed that Pn3a binds to zwitterionic planar lipid bilayers with thermodynamics that are largely driven by enthalpic contributions.


Assuntos
Membrana Celular/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Peptídeos/metabolismo , Proteínas Recombinantes/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Animais , Membrana Celular/química , Células HEK293 , Humanos , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Espectroscopia de Ressonância Magnética , Potenciais da Membrana/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Técnicas de Patch-Clamp , Peptídeos/química , Peptídeos/genética , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/farmacologia , Venenos de Aranha/química , Venenos de Aranha/metabolismo
2.
BMC Anesthesiol ; 20(1): 43, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32085741

RESUMO

BACKGROUND: Substantial clinical and preclinical evidence have indicated the association between amide-linked local anesthesia and the long-term outcomes of cancer patients. However, the potential effects of local anesthesia on cancer recurrence are inconclusive and the underlying mechanisms remain poorly understood. METHODS: We systematically examined the effects of three commonly used local anesthetics in melanoma cells and analyzed the underlying mechanisms focusing on small GTPases. RESULTS: Ropivacaine and lidocaine but not bupivacaine inhibited migration and proliferation, and induced apoptosis in melanoma cells. In addition, ropivacaine and lidocaine but not bupivacaine significantly augmented the in vitro efficacy of vemurafenib (a B-Raf inhibitor for melanoma with BRAF V600E mutation) and dacarbazine (a chemotherapeutic drug). Mechanistically, ropivacaine but not bupivacaine decreased the activities of Ras superfamily members with the dominant inhibitory effects on RhoA and Ras, independent of sodium channel blockade. Rescue studies using constitutively active Ras and Rho activator calpeptin demonstrated that ropivacaine inhibited migration mainly through RhoA whereas growth and survival were mainly inhibited through Ras in melanoma cells. We further detected a global reduction of downstream signaling of Ras and RhoA in ropivacaine-treated melanoma cells. CONCLUSION: Our study is the first to demonstrate the anti-melanoma activity of ropivacaine and lidocaine but not bupivacaine, via targeting small GTPases. Our findings provide preclinical evidence on how amide-linked local anesthetics could affect melanoma patients.


Assuntos
Anestésicos Locais/farmacologia , Melanoma/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Proteínas ras/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/efeitos dos fármacos , Bupivacaína/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Lidocaína/farmacologia , Melanoma/tratamento farmacológico , Ropivacaina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
3.
Chemistry ; 26(9): 2025-2033, 2020 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-31769085

RESUMO

A novel series of C12-keto-type saxitoxin (STX) derivatives bearing an unusual nonhydrated form of the ketone at C12 has been synthesized, and their NaV -inhibitory activity has been evaluated in a cell-based assay as well as whole-cell patch-clamp recording. Among these compounds, 11-benzylidene STX (3 a) showed potent inhibitory activity against neuroblastoma Neuro 2A in both cell-based and electrophysiological analyses, with EC50 and IC50 values of 8.5 and 30.7 nm, respectively. Interestingly, the compound showed potent inhibitory activity against tetrodotoxin-resistant subtype of NaV 1.5, with an IC50 value of 94.1 nm. Derivatives 3 a-d and 3 f showed low recovery rates from NaV 1.2 subtype (ca 45-79 %) compared to natural dcSTX (2), strongly suggesting an irreversible mode of interaction. We propose an interaction model for the C12-keto derivatives with NaV in which the enone moiety in the STX derivatives 3 works as Michael acceptor for the carboxylate of Asp1717 .


Assuntos
Saxitoxina/química , Bloqueadores dos Canais de Sódio/síntese química , Canais de Sódio Disparados por Voltagem/metabolismo , Potenciais de Ação/efeitos dos fármacos , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular Tumoral , Humanos , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Técnicas de Patch-Clamp , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Teoria Quântica , Saxitoxina/metabolismo , Saxitoxina/farmacologia , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/química , Tetrodotoxina/metabolismo , Canais de Sódio Disparados por Voltagem/química , Canais de Sódio Disparados por Voltagem/genética
4.
J Med Chem ; 62(21): 9618-9641, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31525968

RESUMO

Nonselective antagonists of voltage-gated sodium (NaV) channels have been long used for the treatment of epilepsies. The efficacy of these drugs is thought to be due to the block of sodium channels on excitatory neurons, primarily NaV1.6 and NaV1.2. However, these currently marketed drugs require high drug exposure and suffer from narrow therapeutic indices. Selective inhibition of NaV1.6, while sparing NaV1.1, is anticipated to provide a more effective and better tolerated treatment for epilepsies. In addition, block of NaV1.2 may complement the anticonvulsant activity of NaV1.6 inhibition. We discovered a novel series of aryl sulfonamides as CNS-penetrant, isoform-selective NaV1.6 inhibitors, which also displayed potent block of NaV1.2. Optimization focused on increasing selectivity over NaV1.1, improving metabolic stability, reducing active efflux, and addressing a pregnane X-receptor liability. We obtained compounds 30-32, which produced potent anticonvulsant activity in mouse seizure models, including a direct current maximal electroshock seizure assay.


Assuntos
Amidas/química , Sistema Nervoso Central/metabolismo , Epilepsia/tratamento farmacológico , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Cães , Células Hep G2 , Humanos , Células Madin Darby de Rim Canino , Camundongos , Modelos Moleculares , Canal de Sódio Disparado por Voltagem NAV1.6/química , Domínios Proteicos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/uso terapêutico , Relação Estrutura-Atividade
5.
Br J Pharmacol ; 174(9): 880-892, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28239845

RESUMO

BACKGROUND AND PURPOSE: Acetylcholine-induced epithelial Cl- secretion is generally thought to be mediated by epithelial muscarinic receptors and nicotinic receptors on secretomotor neurons. However, recent data have shown expression of nicotinic receptors by intestinal epithelium and the stimulation of Cl- secretion by nicotine, in the presence of the neurotoxin, tetrodotoxin. Here, we aimed to identify the transporters activated by epithelial nicotinic receptors and to clarify their role in cholinergic regulation of intestinal ion transport. EXPERIMENTAL APPROACH: Ussing chamber experiments were performed, using rat distal colon with intact epithelia. Epithelia were basolaterally depolarized to measure currents across the apical membrane. Apically permeabilized tissue was also used to measure currents across the basolateral membrane in the presence of tetrodotoxin. KEY RESULTS: Nicotine had no effect on currents through Cl- channels in the apical membrane or on currents through K+ channels in the apical or the basolateral membrane. Instead, nicotine stimulated the Na+ -K+ -pump as indicated by Na+ -dependency and sensitivity of the nicotine-induced current across the basolateral membrane to cardiac steroids. Effects of nicotine were inhibited by nicotinic receptor antagonists such as hexamethonium and mimicked by dimethyl-4-phenylpiperazinium, a chemically different nicotinic agonist. Simultaneous stimulation of epithelial muscarinic and nicotinic receptors led to a strong potentiation of transepithelial Cl- secretion. CONCLUSIONS AND IMPLICATIONS: These results suggest a novel concept for the cholinergic regulation of transepithelial ion transport by costimulation of muscarinic and nicotinic epithelial receptors and a unique role of nicotinic receptors controlling the activity of the Na+ -K+ -ATPase.


Assuntos
Colo/metabolismo , Epitélio/metabolismo , Receptores Nicotínicos/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Canais de Cloreto/metabolismo , Colo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Epitélio/efeitos dos fármacos , Feminino , Masculino , Nicotina/metabolismo , Nicotina/farmacologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Receptores Nicotínicos/fisiologia , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/fisiologia
6.
Curr Med Chem ; 23(22): 2289-332, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27183981

RESUMO

Voltage-gated sodium channels (VGSC) are responsible for the selective influx of sodium ions in excitable cells. A number of physiological phenomena such as muscle contraction, pain sensation, processing of neuronal information in the brain as well as neuronal regulation of peripheral tissues rely on the activity of these channels. On the other hand, abnormal activity of VGSC are implicated in several pathological processes (e.g., cardiac arrhythmias, epilepsy, and chronic pain) which in some cases (e.g., channelopathies such as myotonias) are linked to specific gene mutations. As a result, VGSC have never stopped attracting the attention of medicinal chemists and the quest for novel drugs to treat these ion channels-associated diseases continues. In this review, VGSC blocking agents reported in the last lustrum are scrutinised with the aim to give a medicinal chemistry perspective on the most interesting compounds classified on the basis of (i) potential therapeutic application, (ii) targeted VGSC isoforms, and (iii) chemical scaffolds. Finally, the clinical potential of selected drug candidates from each chemotype is evaluated by comparing their ligand efficiency metrics. Possible routes for improvement of these preclinical candidates are also discussed.


Assuntos
Bibliotecas de Moléculas Pequenas/química , Bloqueadores dos Canais de Sódio/química , Canais de Sódio Disparados por Voltagem/metabolismo , Arritmias Cardíacas/tratamento farmacológico , Descoberta de Drogas/tendências , Epilepsia/tratamento farmacológico , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/uso terapêutico , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/uso terapêutico , Canais de Sódio Disparados por Voltagem/química
7.
Biochimie ; 121: 326-35, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26747232

RESUMO

The toxin PnTx4(5-5) from the spider Phoneutria nigriventer is extremely toxic/lethal to insects but has no macroscopic behavioral effects observed in mice after intracerebral injection. Nevertheless, it was demonstrated that it inhibits the N-methyl-d-aspartate (NMDA) - subtype of glutamate receptors of cultured rat hippocampal neurons. PnTx4(5-5) has 63% identity to PnTx4(6-1), another insecticidal toxin from P. nigriventer, which can slow down the sodium current inactivation in insect central nervous system, but has no effect on Nav1.2 and Nav1.4 rat sodium channels. Here, we have cloned and heterologous expressed the toxin PnTx4(5-5) in Escherichia coli. The recombinant toxin rPnTx4(5-5) was tested on the sodium channel NavBg from the cockroach Blatella germanica and on mammalian sodium channels Nav1.2-1.6, all expressed in Xenopus leavis oocytes. We showed that the toxin has different affinity and mode of action on insect and mammalian sodium channels. The most remarkable effect was on NavBg, where rPnTx4(5-5) strongly slowed down channel inactivation (EC50 = 212.5 nM), and at 1 µM caused an increase on current peak amplitude of 105.2 ± 3.1%. Interestingly, the toxin also inhibited sodium current on all the mammalian channels tested, with the higher current inhibition on Nav1.3 (38.43 ± 8.04%, IC50 = 1.5 µM). Analysis of activation curves on Nav1.3 and Nav1.5 showed that the toxin shifts channel activation to more depolarized potentials, which can explain the sodium current inhibition. Furthermore, the toxin also slightly slowed down sodium inactivation on Nav1.3 and Nav1.6 channels. As far as we know, this is the first araneomorph toxin described which can shift the sodium channel activation to more depolarized potentials and also slows down channel inactivation.


Assuntos
Escherichia coli/metabolismo , Neurotoxinas/toxicidade , Canais de Sódio/efeitos dos fármacos , Venenos de Aranha/toxicidade , Animais , Baratas , Escherichia coli/genética , Neurotoxinas/genética , Neurotoxinas/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/toxicidade , Canais de Sódio/metabolismo , Venenos de Aranha/química , Aranhas/genética
8.
Biochem Pharmacol ; 85(10): 1542-54, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23473802

RESUMO

One of the most potent insecticidal venom peptides described to date is Aps III from the venom of the trapdoor spider Apomastus schlingeri. Aps III is highly neurotoxic to lepidopteran crop pests, making it a promising candidate for bioinsecticide development. However, its disulfide-connectivity, three-dimensional structure, and mode of action have not been determined. Here we show that recombinant Aps III (rAps III) is an atypical knottin peptide; three of the disulfide bridges form a classical inhibitor cystine knot motif while the fourth disulfide acts as a molecular staple that restricts the flexibility of an unusually large ß hairpin loop that often houses the pharmacophore in this class of toxins. We demonstrate that the irreversible paralysis induced in insects by rAps III results from a potent block of insect voltage-gated sodium channels. Channel block by rAps III is voltage-independent insofar as it occurs without significant alteration in the voltage-dependence of channel activation or steady-state inactivation. Thus, rAps III appears to be a pore blocker that plugs the outer vestibule of insect voltage-gated sodium channels. This mechanism of action contrasts strikingly with virtually all other sodium channel modulators isolated from spider venoms that act as gating modifiers by interacting with one or more of the four voltage-sensing domains of the channel.


Assuntos
Miniproteínas Nó de Cistina/química , Dípteros/efeitos dos fármacos , Proteínas de Insetos/química , Neurotoxinas/química , Periplaneta/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/química , Venenos de Aranha/química , Canais de Sódio Disparados por Voltagem/química , Sequência de Aminoácidos , Animais , Miniproteínas Nó de Cistina/metabolismo , Miniproteínas Nó de Cistina/farmacologia , Dípteros/metabolismo , Dissulfetos/química , Escherichia coli/genética , Proteínas de Insetos/antagonistas & inibidores , Proteínas de Insetos/metabolismo , Cinética , Potenciais da Membrana/efeitos dos fármacos , Modelos Moleculares , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotoxinas/metabolismo , Neurotoxinas/farmacologia , Técnicas de Patch-Clamp , Periplaneta/metabolismo , Cultura Primária de Células , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Venenos de Aranha/metabolismo , Venenos de Aranha/farmacologia , Aranhas/química , Aranhas/fisiologia , Canais de Sódio Disparados por Voltagem/metabolismo
9.
Biochem J ; 451(1): 81-90, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23356888

RESUMO

Heart failure is one of the most prevalent causes of death in the western world. Sea anemone contains a myriad of short peptide neurotoxins affecting many pharmacological targets, several of which possess cardiotonic activity. In the present study we describe the isolation and characterization of AdE-1 (ion channel modifier), a novel cardiotonic peptide from the sea anemone Aiptasia diaphana, which differs from other cnidarian toxins. Although AdE-1 has the same cysteine residue arrangement as sea anemone type 1 and 2 Na(+) channel toxins, its sequence contains many substitutions in conserved and essential sites and its overall homology to other toxins identified to date is low (<36%). Physiologically, AdE-1 increases the amplitude of cardiomyocyte contraction and slows the late phase of the twitch relaxation velocity with no induction of spontaneous twitching. It increases action potential duration of cardiomyocytes with no effect on its threshold and on the cell's resting potential. Similar to other sea anemone Na(+) channel toxins such as Av2 (Anemonia viridis toxin II), AdE-1 markedly inhibits Na(+) current inactivation with no significant effect on current activation, suggesting a similar mechanism of action. However, its effects on twitch relaxation velocity, action potential amplitude and on the time to peak suggest that this novel toxin affects cardiomyocyte function via a more complex mechanism. Additionally, Av2's characteristic delayed and early after-depolarizations were not observed. Despite its structural differences, AdE-1 physiologic effectiveness is comparable with Av2 with a similar ED(50) value to blowfly larvae. This finding raises questions regarding the extent of the universality of structure-function in sea anemone Na(+) channel toxins.


Assuntos
Venenos de Cnidários , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Peptídeos , Anêmonas-do-Mar , Bloqueadores dos Canais de Sódio , Animais , Células Cultivadas , Venenos de Cnidários/química , Venenos de Cnidários/genética , Venenos de Cnidários/metabolismo , Venenos de Cnidários/farmacologia , Masculino , Miócitos Cardíacos/patologia , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Anêmonas-do-Mar/química , Anêmonas-do-Mar/genética , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia
10.
Toxicon ; 60(4): 478-91, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22543187

RESUMO

Voltage-gated sodium (Na(V)) channels play a central role in the propagation of action potentials in excitable cells in both humans and insects. Many venomous animals have therefore evolved toxins that modulate the activity of Na(V) channels in order to subdue their prey and deter predators. Spider venoms in particular are rich in Na(V) channel modulators, with one-third of all known ion channel toxins from spider venoms acting on Na(V) channels. Here we review the landscape of spider-venom peptides that have so far been described to target vertebrate or invertebrate Na(V) channels. These peptides fall into 12 distinct families based on their primary structure and cysteine scaffold. Some of these peptides have become useful pharmacological tools, while others have potential as therapeutic leads because they target specific Na(V) channel subtypes that are considered to be important analgesic targets. Spider venoms are conservatively predicted to contain more than 10 million bioactive peptides and so far only 0.01% of this diversity been characterised. Thus, it is likely that future research will reveal additional structural classes of spider-venom peptides that target Na(V) channels.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Peptídeos/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Venenos de Aranha/farmacologia , Sequência de Aminoácidos , Animais , Descoberta de Drogas , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/metabolismo , Venenos de Aranha/química , Venenos de Aranha/metabolismo , Aranhas/fisiologia
11.
J Pept Sci ; 18(7): 442-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22605564

RESUMO

Protoxin II is biologically active peptide containing the inhibitory cystine knot motif. A synthetic version of the toxin was generated with standard Fmoc solid phase peptide synthesis. If N-methylmorpholine was used as a base during synthesis of the linear protoxin II, it was found that a significant amount of racemization (approximately 50%) was observed during the process of cysteine residue coupling. This racemization could be suppressed by substituting N-methylmorpholine with 2,4,6-collidine. The crude linear toxin was then air oxidized and purified. Electrophysiological assessment of the synthesized protoxin II confirmed its previously described interactions with voltage-gated sodium channels. Eight other naturally occurring inhibitory knot peptides were also synthesized using this same methodology. The inhibitory potencies of these synthesized toxins on Nav1.7 and Nav1.2 channels are summarized.


Assuntos
Cisteína/química , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Peptídeos/síntese química , Peptídeos/metabolismo , Bloqueadores dos Canais de Sódio/síntese química , Bloqueadores dos Canais de Sódio/metabolismo , Técnicas de Síntese em Fase Sólida , Venenos de Aranha/síntese química , Venenos de Aranha/metabolismo , Linhagem Celular , Humanos , Morfolinas/química , Oxirredução , Peptídeos/química , Bloqueadores dos Canais de Sódio/química , Venenos de Aranha/química , Estereoisomerismo , Especificidade por Substrato
12.
Br J Pharmacol ; 166(4): 1490-502, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22300168

RESUMO

BACKGROUND AND PURPOSE: Terfenadine has been reported to cause cardiac death. Hence, we investigated its pro-arrhythmic potential in various in vitro models. EXPERIMENTAL APPROACH: Pro-arrhythmic effects of terfenadine were investigated in rabbit isolated hearts and left ventricular wedge preparations. Also, using whole-cell patch-clamp recording, we examined its effect on the human ether-à-go-go-related gene (hERG) current in HEK293 cells transfected with hERG and on the I(Na) current in rabbit ventricular cells and human atrial myocytes. KEY RESULTS: Terfenadine concentration- and use-dependently inhibited I(Na) in rabbit myocytes and in human atrial myocytes and also inhibited the hERG. In both the rabbit left ventricular wedge and heart preparations, terfenadine at 1 µM only slightly prolonged the QT- and JT-intervals but at 10 µM, it caused a marked widening of the QRS complex, cardiac wavelength shortening, incidences of in-excitability and non-TdP-like ventricular tachycardia/fibrillation (VT/VF) without prolongation of the QT/JT-interval. At 10 µM terfenadine elicited a lower incidence of early afterdepolarizations versus non- Torsades de Pointes (TdP)-like VT/VF (100% incidence), and did not induce TdPs. Although the concentration of terfenadine in the tissue-bath was low, it accumulated within the heart tissue. CONCLUSION AND IMPLICATIONS: Our data suggest that: (i) the induction of non-TdP-like VT/VF, which is caused by slowing of conduction via blockade of I(Na) (like Class Ic flecainide), may constitute a more important risk for terfenadine-induced cardiac death; (ii) although terfenadine is a potent hERG blocker, the risk for non-TdP-like VT/VF exceeds the risk for TdPs; and (iii) cardiac wavelength (λ) could serve as a biomarker to predict terfenadine-induced VT/VF.


Assuntos
Antagonistas não Sedativos dos Receptores H1 da Histamina/farmacologia , Síndrome do QT Longo/induzido quimicamente , Miócitos Cardíacos/efeitos dos fármacos , Taquicardia Ventricular/etiologia , Terfenadina/farmacologia , Torsades de Pointes/induzido quimicamente , Fibrilação Ventricular/etiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Antiarrítmicos/efeitos adversos , Antiarrítmicos/metabolismo , Antiarrítmicos/farmacologia , Apêndice Atrial/citologia , Apêndice Atrial/efeitos dos fármacos , Apêndice Atrial/metabolismo , Transporte Biológico , Células Cultivadas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Células HEK293 , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Antagonistas não Sedativos dos Receptores H1 da Histamina/efeitos adversos , Antagonistas não Sedativos dos Receptores H1 da Histamina/metabolismo , Humanos , Técnicas In Vitro , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/fisiopatologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Concentração Osmolar , Coelhos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Bloqueadores dos Canais de Sódio/efeitos adversos , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Terfenadina/efeitos adversos , Terfenadina/metabolismo , Torsades de Pointes/metabolismo , Torsades de Pointes/fisiopatologia
13.
Biophys J ; 102(3): 483-8, 2012 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-22325270

RESUMO

Polypeptide toxins isolated from the venom of cone snails, known as µ-conotoxins, block voltage-gated sodium channels by physically occluding the ion-conducting pathway. Using molecular dynamics, we show that one subtype of µ-conotoxins, PIIIA, effectively blocks the bacterial voltage-gated sodium channel Na(V)Ab, whose crystal structure has recently been elucidated. The spherically shaped toxin, carrying a net charge of +6 e with six basic residues protruding from its surface, is attracted by the negatively charged residues on the vestibular wall and the selectivity filter of the channel. The side chain of each of these six arginine and lysine residues can wedge into the selectivity filter, whereas the side chains of other basic residues form electrostatic complexes with two acidic residues on the channel. We construct the profile of potential of mean force for the unbinding of PIIIA from the channel, and predict that PIIIA blocks the bacterial sodium channel with subnanomolar affinity.


Assuntos
Proteínas de Bactérias/metabolismo , Conotoxinas/metabolismo , Conotoxinas/farmacologia , Simulação de Dinâmica Molecular , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Arcobacter , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Conotoxinas/química , Ligação Proteica , Conformação Proteica , Bloqueadores dos Canais de Sódio/química , Canais de Sódio/química , Especificidade por Substrato
14.
Neurogastroenterol Motil ; 23(11): 1007-12, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21848627

RESUMO

BACKGROUND: Disturbance in fluid secretion, driven by chloride secretion, might play a role in constipation. However, disturbed chloride secretion in those patients has yet to be evaluated. Therefore, the aim of this study was to compare chloride secretion in rectal biopsies of children with functional constipation (FC) to those without constipation. METHODS: To measure changes in short circuit current (I(sc) in µA cm(-2)) reflecting chloride secretion, intestinal biopsies from children with constipation, to either exclude or diagnose Hirschsprung's disease, and from children without constipation (controls) undergoing colonoscopy for screening of familial adenomatous polyposis, juvenile polyps or inflammatory bowel disease (IBD), were compared and studied in Ussing chambers. Following electrogenic sodium absorption blockade by amiloride, chloride secretory responses to calcium-linked (histamine, carbachol) and cAMP-linked (IBMX/forskolin) secretagogues were assessed. KEY RESULTS: Ninety-six patients (46 FC) participated; nine FC patients (n = 1 congenital syndrome and n = 8 technical problems) and 13 controls (n = 6 IBD; n = 7 technical problems) were excluded. No significant difference was found in mean (±SE) basal chloride currents between children with FC and controls (9.6 ± 1.1 vs 9.2 ± 0.8; P = 0.75, respectively). Responses to calcium-linked chloride secretagogues (histamine and carbachol) were significantly higher in controls (33.0 ± 3.0 vs 24.5 ± 2.3; P = 0.03 and 33.6 ± 3.4 vs 26.4 ± 2.7; P = 0.05 following histamine and carbachol, respectively). CONCLUSIONS & INFERENCES: Calcium-linked chloride secretion is disturbed in children with FC. Whether this defect occurs at the level of histamine receptors, components of receptor-linked signal transduction pathways or basolateral Ca(2+) -sensitive K(+) channels enhancing the electrical driving force for apical chloride secretion, remains to be explored.


Assuntos
Cloretos/metabolismo , Constipação Intestinal/metabolismo , Reto/metabolismo , 1-Metil-3-Isobutilxantina/metabolismo , Amilorida/metabolismo , Biópsia , Carbacol/metabolismo , Criança , Agonistas Colinérgicos/metabolismo , Colforsina/metabolismo , Constipação Intestinal/diagnóstico , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Defecação , Feminino , Doença de Hirschsprung/diagnóstico , Doença de Hirschsprung/fisiopatologia , Histamina/metabolismo , Agonistas dos Receptores Histamínicos/metabolismo , Humanos , Masculino , Inibidores de Fosfodiesterase/metabolismo , Reto/cirurgia , Bloqueadores dos Canais de Sódio/metabolismo
15.
Proc Natl Acad Sci U S A ; 108(25): 10260-5, 2011 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-21646513

RESUMO

Loss of cystic fibrosis transmembrane conductance regulator (CFTR) anion channel function causes cystic fibrosis (CF) lung disease. CFTR is expressed in airway epithelia, but how CF alters electrolyte transport across airway epithelia has remained uncertain. Recent studies of a porcine model showed that in vivo, excised, and cultured CFTR(-/-) and CFTR(ΔF508/ΔF508) airway epithelia lacked anion conductance, and they did not hyperabsorb Na(+). Therefore, we asked whether Cl(-) and Na(+) conductances were altered in human CF airway epithelia. We studied differentiated primary cultures of tracheal/bronchial epithelia and found that transepithelial conductance (Gt) under basal conditions and the cAMP-stimulated increase in Gt were markedly attenuated in CF epithelia compared with non-CF epithelia. These data reflect loss of the CFTR anion conductance. In CF and non-CF epithelia, the Na(+) channel inhibitor amiloride produced similar reductions in Gt and Na(+) absorption, indicating that Na(+) conductance in CF epithelia did not exceed that in non-CF epithelia. Consistent with previous reports, adding amiloride caused greater reductions in transepithelial voltage and short-circuit current in CF epithelia than in non-CF epithelia; these changes are attributed to loss of a Cl(-) conductance. These results indicate that Na(+) conductance was not increased in these cultured CF tracheal/bronchial epithelia and point to loss of anion transport as key to airway epithelial dysfunction in CF.


Assuntos
Cloretos/metabolismo , Fibrose Cística/fisiopatologia , Epitélio/metabolismo , Mucosa Respiratória/metabolismo , Sódio/metabolismo , Amilorida/metabolismo , Animais , Ânions/metabolismo , Células Cultivadas , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Humanos , Transporte de Íons/fisiologia , Mucosa Respiratória/anatomia & histologia , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/metabolismo , Suínos
16.
Am J Respir Cell Mol Biol ; 45(5): 1059-68, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21562317

RESUMO

Alveolar edema and decreased inspired Po(2) decrease the oxygen supply to alveolar epithelia, impairing ß(2)-adrenergic receptor (ß2AR) signaling and alveolar reabsorption. ß2AR agonists potently stimulate alveolar reabsorption. Thus, hypoxia impairs a major defense mechanism that provides protection from alveolar edema. Because in vivo data on the combined effects of prolonged hypoxia and ß2AR agonist treatment on ß2AR signaling are sparse, we tested whether in vivo hypoxia augments the inactivation of ß2AR during prolonged stimulation. Rats were exposed to normoxia (N) and hypoxia (8% O(2); H), and were also treated with terbutaline (T; 2.5 mg/kg, intraperitoneal, twice daily) or saline (S) for 4 days. ß2AR signaling was studied in alveolar epithelial (ATII) cells and in whole-lung tissue from treated rats. The terbutaline-stimulated formation of cyclic adenosine monophosphate was decreased by approximately 40% in whole lung and in ATII cells of NT, HS, and HT. The effects were not additive. The ß2AR number was increased in HS, but decreased in NT and HT. Treatment increased the G-protein-coupled receptor kinase 2 protein in the plasma membranes of ATII cells, but did not affect G proteins. In vivo hypoxia significantly decreased total and amiloride-sensitive alveolar fluid reabsorption, which was prevented by acute alveolar treatment and 4 days of systemic terbutaline treatment. The αENaC (subunit of epithelial Na channels) protein in plasma membranes was increased in HT, without effects on mRNA. These results indicate that prolonged alveolar hypoxia and treatment with terbutaline impaired ß2AR signaling in alveolar epithelia and in whole lungs, and this signaling was not further impaired by hypoxia. Despite impaired ß2AR signaling, treatment with terbutaline for 4 days prevented the inhibition of alveolar reabsorption caused by in vivo hypoxia.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Hipóxia/tratamento farmacológico , Pulmão/efeitos dos fármacos , Receptores Adrenérgicos beta 2/metabolismo , Terbutalina/farmacologia , Absorção , Agonistas de Receptores Adrenérgicos beta 2/metabolismo , Amilorida/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , AMP Cíclico/biossíntese , Canais Epiteliais de Sódio/efeitos dos fármacos , Canais Epiteliais de Sódio/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/biossíntese , Hipóxia/metabolismo , Pulmão/metabolismo , Masculino , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Ratos , Ratos Sprague-Dawley , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia
17.
Mol Pharmacol ; 80(2): 247-57, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21536754

RESUMO

Brilliant blue G (BBG), best known as an antagonist of P2X7 receptors, was found to inhibit voltage-gated sodium currents in N1E-115 neuroblastoma cells. Sodium currents elicited from a holding potential of -60 mV were blocked with an IC(50) of 2 µM. Block was enhanced in a use-dependent manner at higher stimulation rates. The voltage-dependence of inactivation was shifted in the hyperpolarizing direction, and recovery from inactivation was slowed by BBG. The most dramatic effect of BBG was to slow recovery from inactivation after long depolarizations, with 3 µM BBG increasing half-time for recovery (measured at -120 mV) from 24 to 854 ms after a 10-s step to 0 mV. These results were mimicked by a kinetic model in which BBG binds weakly to resting channels (K(d) = 170 µM) but tightly to fast-inactivated channels (K(d) = 5 µM) and even more tightly (K(d) = 0.2 µM) to slow-inactivated channels. In contrast to BBG, the structurally related food-coloring dye Brilliant Blue FCF had very little effect at concentrations up to 30 µM. These results show that BBG inhibits voltage-gated sodium channels at micromolar concentrations. Although BBG inhibition of sodium channels is less potent than inhibition of P2X7 receptors, there may be significant inhibition of sodium channels at BBG concentrations achieved in spinal cord or brain during experimental treatment of spinal cord injury or Huntington's disease. Considered as a sodium channel blocker, BBG is remarkably potent, acting with more than 10-fold greater potency than lacosamide, another blocker thought to bind to slow-inactivated channels.


Assuntos
Ativação do Canal Iônico/fisiologia , Neurônios/metabolismo , Corantes de Rosanilina/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/metabolismo , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Receptores Purinérgicos P2X7/metabolismo , Corantes de Rosanilina/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia
18.
Insect Biochem Mol Biol ; 41(1): 36-41, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20888415

RESUMO

A new class of sodium channel blocker insecticides (SCBIs), which include indoxacarb, its active metabolite, DCJW, and metaflumizone, preferably block inactivated states of both insect and mammalian sodium channels in a manner similar to that by which local anesthetic (LA) drugs block mammalian sodium channels. A recent study showed that two residues in the cockroach sodium channel, F1817 and Y1824, corresponding to two key LA-interacting residues identified in mammalian sodium channels are not important for the action of SCBIs on insect sodium channels, suggesting unique interactions of SCBIs with insect sodium channels. However, the mechanism of action of LAs on insect sodium channels has not been investigated. In this study, we examined the effects of lidocaine on a cockroach sodium channel variant, BgNa(v)1-1a, and determined whether F1817 and Y1824 are also critical for the action of LAs on insect sodium channels. Lidocaine blocked BgNa(v)1-1a channels in the resting state with potency similar to that observed in mammalian sodium channels. Lidocaine also stabilized both fast-inactivated and slow-inactivated states of BgNa(v)1-1a channels, and caused a limited degree of use- and frequency-dependent block, major characteristics of LA action on mammalian sodium channels. Alanine substitutions of F1817 and Y1824 reduced the sensitivity of the BgNa(v)1-1a channel to the use-dependent block by lidocaine, but not to tonic blocking and inactivation stabilizing effects of lidocaine. Thus, similar to those on mammalian sodium channels, F1817 and Y1824 are important for the action of lidocaine on cockroach sodium channels. Our results suggest that the receptor sites for lidocaine and SCBIs are different on insect sodium channels.


Assuntos
Anestésicos Locais/farmacologia , Baratas , Proteínas de Insetos/genética , Ativação do Canal Iônico/efeitos dos fármacos , Lidocaína/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/genética , Substituição de Aminoácidos , Anestésicos Locais/metabolismo , Animais , Baratas/efeitos dos fármacos , Baratas/metabolismo , Eletrofisiologia , Feminino , Proteínas de Insetos/antagonistas & inibidores , Proteínas de Insetos/metabolismo , Inseticidas/metabolismo , Inseticidas/farmacologia , Ativação do Canal Iônico/fisiologia , Cinética , Lidocaína/metabolismo , Oócitos/metabolismo , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/metabolismo , Xenopus laevis
19.
Acta Physiol (Oxf) ; 201(1): 97-107, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20969730

RESUMO

AIMS: As little is currently known about acid-sensing ion channels (ASICs) in intestinal epithelial cells, the aims of the present study were to investigate the expression and function of ASICs in intestinal epithelial cells, particularly their physiological role in the acid-stimulated duodenal mucosal bicarbonate secretion (DMBS). METHODS: RT-PCR and digital Ca²(+) imaging were used to determine the expression and function of ASICs in HT29 cells and SCBN cells, intestinal epithelial crypt cell lines. The acid-stimulated DMBS was measured in C57 black mice in vivo to study the role of ASICs in this physiological process. RESULTS: ASIC1a mRNA expression was detected in the duodenal mucosa stripped from mice and epithelial cell lines, in which cytoplasmic free Ca²(+) ([Ca²(+) ](cyt)) in response to extracellular acidosis was also increased. In Ca²(+) -containing solutions, acidosis (pH 6.0-5.0) raised [Ca²(+) ](cyt) in both HT29 cells and SCBN cells in a similar pH-dependent manner. Acidosis-induced increase in [Ca²(+) ](cyt) was markedly inhibited by amiloride (an ASICs blocker), SK&F96365 (a blocker for non-selective cation channels), or in Ca²(+) -free solutions; but was abolished by amiloride in Ca²(+) -free solutions. However, acidosis-induced increase in [Ca²(+) ](cyt) was slightly affected by U73122 (a PLC inhibitor), or nifedipine (a voltage-gated Ca²(+) channel blocker). After acidosis raised [Ca²(+) ](cyt) , stimulation of purinergic receptors with ATP further increased [Ca²(+) ](cyt) , but acidosis-induced increase in [Ca²(+) ](cyt) was not altered by suramin. Moreover, acid-stimulated murine DMBS was significantly attenuated by amiloride. CONCLUSION: Therefore, ASICs are functionally expressed in intestinal epithelial cells, and may play a role in acid-stimulated DMBS through a Ca²(+) signalling pathway.


Assuntos
Bicarbonatos/metabolismo , Duodeno/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/citologia , Proteínas do Tecido Nervoso/metabolismo , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Amilorida/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Células Epiteliais/citologia , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Transdução de Sinais/fisiologia , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/genética , Suramina/metabolismo
20.
Mol Pharmacol ; 78(6): 1124-34, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20855463

RESUMO

The voltage-gated sodium channel Na(v)1.7 plays a crucial role in pain, and drugs that inhibit hNa(v)1.7 may have tremendous therapeutic potential. ProTx-II and huwentoxin-IV (HWTX-IV), cystine knot peptides from tarantula venoms, preferentially block hNa(v)1.7. Understanding the interactions of these toxins with sodium channels could aid the development of novel pain therapeutics. Whereas both ProTx-II and HWTX-IV have been proposed to preferentially block hNa(v)1.7 activation by trapping the domain II voltage-sensor in the resting configuration, we show that specific residues in the voltage-sensor paddle of domain II play substantially different roles in determining the affinities of these toxins to hNa(v)1.7. The mutation E818C increases ProTx-II's and HWTX-IV's IC(50) for block of hNa(v)1.7 currents by 4- and 400-fold, respectively. In contrast, the mutation F813G decreases ProTx-II affinity by 9-fold but has no effect on HWTX-IV affinity. It is noteworthy that we also show that ProTx-II, but not HWTX-IV, preferentially interacts with hNa(v)1.7 to impede fast inactivation by trapping the domain IV voltage-sensor in the resting configuration. Mutations E1589Q and T1590K in domain IV each decreased ProTx-II's IC(50) for impairment of fast inactivation by ~6-fold. In contrast mutations D1586A and F1592A in domain-IV increased ProTx-II's IC(50) for impairment of fast inactivation by ~4-fold. Our results show that whereas ProTx-II and HWTX-IV binding determinants on domain-II may overlap, domain II plays a much more crucial role for HWTX-IV, and contrary to what has been proposed to be a guiding principle of sodium channel pharmacology, molecules do not have to exclusively target the domain IV voltage-sensor to influence sodium channel inactivation.


Assuntos
Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/metabolismo , Venenos de Aranha/metabolismo , Sequência de Aminoácidos , Animais , Feminino , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.7 , Bloqueadores dos Canais de Sódio/farmacologia , Venenos de Aranha/farmacologia , Aranhas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA