Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 103(4): 221-229, 2023 04.
Article in English | MEDLINE | ID: mdl-36635052

ABSTRACT

Sodium channel inhibitors used as local anesthetics, antiarrhythmics, or antiepileptics typically have the property of use-dependent inhibition, whereby inhibition is enhanced by repetitive channel activation. For targeting pain, Nav1.8 channels are an attractive target because they are prominent in primary pain-sensing neurons, with little or no expression in most other kinds of neurons, and a number of Nav1.8-targeted compounds have been developed. We examined the characteristics of Nav1.8 inhibition by one of the most potent Nav1.8 inhibitors so far described, A-887826, and found that when studied with physiologic resting potentials and physiologic temperatures, inhibition had strong "reverse use dependence", whereby inhibition was relieved by repetitive short depolarizations. This effect was much stronger with A-887826 than with A-803467, another Nav1.8 inhibitor. The use-dependent relief from inhibition was seen in both human Nav1.8 channels studied in a cell line and in native Nav1.8 channels in mouse dorsal root ganglion (DRG) neurons. In native Nav1.8 channels, substantial relief of inhibition occurred during repetitive stimulation by action potential waveforms at 5 Hz, suggesting that the phenomenon is likely important under physiologic conditions. SIGNIFICANCE STATEMENT: Nav1.8 sodium channels are expressed in primary pain-sensing neurons and are a prime current target for new drugs for pain. This work shows that one of the most potent Nav1.8 inhibitors, A-887826, has the unusual property that inhibition is relieved by repeated short depolarizations. This "reverse use dependence" may reduce inhibition during physiological firing and should be selected against in drug development.


Subject(s)
Morpholines , NAV1.8 Voltage-Gated Sodium Channel , Neurons , Niacinamide , Pain , Animals , Humans , Mice , Ganglia, Spinal , Membrane Potentials , Morpholines/pharmacology , Morpholines/therapeutic use , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Neurons/drug effects , Neurons/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Pain/drug therapy , Pain/metabolism , Rats, Sprague-Dawley , Rats
2.
Toxins (Basel) ; 13(5)2021 05 17.
Article in English | MEDLINE | ID: mdl-34067828

ABSTRACT

The primary studies have shown that scorpion analgesic peptide N58A has a significant effect on voltage-gated sodium channels (VGSCs) and plays an important role in neuropathic pain. The purpose of this study was to investigate the analgesic effect of N58A on trigeminal neuralgia (TN) and its possible mechanism. The results showed that N58A could significantly increase the threshold of mechanical pain and thermal pain and inhibit the spontaneous asymmetric scratching behavior of rats. Western blotting results showed that N58A could significantly reduce the protein phosphorylation level of ERK1/2, P38, JNK, and ERK5/CREB pathways and the expression of Nav1.8 and Nav1.9 proteins in a dose-dependent manner. The changes in current and kinetic characteristics of Nav1.8 and Nav1.9 channels in TG neurons were detected by the whole-cell patch clamp technique. The results showed that N58A significantly decreased the current density of Nav1.8 and Nav1.9 in model rats, and shifted the activation curve to hyperpolarization and the inactivation curve to depolarization. In conclusion, the analgesic effect of N58A on the chronic constriction injury of the infraorbital (IoN-CCI) model rats may be closely related to the regulation of the MAPK pathway and Nav1.8 and Nav1.9 sodium channels.


Subject(s)
Analgesics/pharmacology , Peptides/pharmacology , Scorpion Venoms/chemistry , Trigeminal Neuralgia/drug therapy , Analgesics/administration & dosage , Analgesics/isolation & purification , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , MAP Kinase Signaling System/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.9 Voltage-Gated Sodium Channel/drug effects , NAV1.9 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Patch-Clamp Techniques , Peptides/administration & dosage , Peptides/isolation & purification , Rats , Rats, Sprague-Dawley , Scorpions , Tetrodotoxin/pharmacology
3.
J Neurosci ; 41(30): 6371-6387, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34131037

ABSTRACT

The nonpsychoactive phytocannabinoid cannabidiol (CBD) has been shown to have analgesic effects in animal studies but little is known about its mechanism of action. We examined the effects of CBD on intrinsic excitability of primary pain-sensing neurons. Studying acutely dissociated capsaicin-sensitive mouse DRG neurons at 37°C, we found that CBD effectively inhibited repetitive action potential firing, from 15-20 action potentials evoked by 1 s current injections in control to 1-3 action potentials with 2 µm CBD. Reduction of repetitive firing was accompanied by a reduction of action potential height, widening of action potentials, reduction of the afterhyperpolarization, and increased propensity to enter depolarization block. Voltage-clamp experiments showed that CBD inhibited both TTX-sensitive and TTX-resistant (TTX-R) sodium currents in a use-dependent manner. CBD showed strong state-dependent inhibition of TTX-R channels, with fast binding to inactivated channels during depolarizations and slow unbinding on repolarization. CBD alteration of channel availability at various voltages suggested that CBD binds especially tightly [Kd (dissociation constant), ∼150 nm] to the slow inactivated state of TTX-R channels, which can be substantially occupied at voltages as negative as -40 mV. Remarkably, CBD was more potent in inhibiting TTX-R channels and inhibiting action potential firing than the local anesthetic bupivacaine. We conclude that CBD might produce some of its analgesic effects by direct effects on neuronal excitability, with tight binding to the slow inactivated state of Nav1.8 channels contributing to effective inhibition of repetitive firing by modest depolarizations.SIGNIFICANCE STATEMENT Cannabidiol (CBD) has been shown to inhibit pain in various rodent models, but the mechanism of this effect is unknown. We describe the ability of CBD to inhibit repetitive action potential firing in primary nociceptive neurons from mouse dorsal root ganglia and analyze the effects on voltage-dependent sodium channels. We find that CBD interacts with TTX-resistant sodium channels in a state-dependent manner suggesting particularly tight binding to slow inactivated states of Nav1.8 channels, which dominate the overall inactivation of Nav1.8 channels for small maintained depolarizations from the resting potential. The results suggest that CBD can exert analgesic effects in part by directly inhibiting repetitive firing of primary nociceptors and suggest a strategy of identifying compounds that bind selectively to slow inactivated states of Nav1.8 channels for developing effective analgesics.


Subject(s)
Analgesics/pharmacology , Cannabidiol/pharmacology , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Nociceptors/drug effects , Action Potentials/drug effects , Animals , Cells, Cultured , Female , Ganglia, Spinal , Male , Mice , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Nociceptors/metabolism
4.
Basic Res Cardiol ; 115(2): 20, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32078054

ABSTRACT

Pharmacologic approaches for the treatment of atrial arrhythmias are limited due to side effects and low efficacy. Thus, the identification of new antiarrhythmic targets is of clinical interest. Recent genome studies suggested an involvement of SCN10A sodium channels (NaV1.8) in atrial electrophysiology. This study investigated the role and involvement of NaV1.8 (SCN10A) in arrhythmia generation in the human atria and in mice lacking NaV1.8. NaV1.8 mRNA and protein were detected in human atrial myocardium at a significant higher level compared to ventricular myocardium. Expression of NaV1.8 and NaV1.5 did not differ between myocardium from patients with atrial fibrillation and sinus rhythm. To determine the electrophysiological role of NaV1.8, we investigated isolated human atrial cardiomyocytes from patients with sinus rhythm stimulated with isoproterenol. Inhibition of NaV1.8 by A-803467 or PF-01247324 showed no effects on the human atrial action potential. However, we found that NaV1.8 significantly contributes to late Na+ current and consequently to an increased proarrhythmogenic diastolic sarcoplasmic reticulum Ca2+ leak in human atrial cardiomyocytes. Selective pharmacological inhibition of NaV1.8 potently reduced late Na+ current, proarrhythmic diastolic Ca2+ release, delayed afterdepolarizations as well as spontaneous action potentials. These findings could be confirmed in murine atrial cardiomyocytes from wild-type mice and also compared to SCN10A-/- mice (genetic ablation of NaV1.8). Pharmacological NaV1.8 inhibition showed no effects in SCN10A-/- mice. Importantly, in vivo experiments in SCN10A-/- mice showed that genetic ablation of NaV1.8 protects against atrial fibrillation induction. This study demonstrates that NaV1.8 is expressed in the murine and human atria and contributes to late Na+ current generation and cellular arrhythmogenesis. Blocking NaV1.8 selectively counteracts this pathomechanism and protects against atrial arrhythmias. Thus, our translational study reveals a new selective therapeutic target for treating atrial arrhythmias.


Subject(s)
Aniline Compounds/pharmacology , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/prevention & control , Furans/pharmacology , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Picolinic Acids/pharmacology , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Aged , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Mice, Knockout , Middle Aged , Myocytes, Cardiac/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism
5.
ACS Chem Neurosci ; 10(12): 4834-4846, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31697467

ABSTRACT

Naringenin (2S)-5,7-dihydroxy-2-(4-hydroxyphenyl)-3,4-dihydro-2H-1-benzopyran-4-one is a natural flavonoid found in fruits from the citrus family. Because (2S)-naringenin is known to racemize, its bioactivity might be related to one or both enantiomers. Computational studies predicted that (2R)-naringenin may act on voltage-gated ion channels, particularly the N-type calcium channel (CaV2.2) and the NaV1.7 sodium channel-both of which are key for pain signaling. Here we set out to identify the possible mechanism of action of naringenin. Naringenin inhibited depolarization-evoked Ca2+ influx in acetylcholine-, ATP-, and capsaicin-responding rat dorsal root ganglion (DRG) neurons. This was corroborated in electrophysiological recordings from DRG neurons. Pharmacological dissection of each of the voltage-gated Ca2+ channels subtypes could not pinpoint any selectivity of naringenin. Instead, naringenin inhibited NaV1.8-dependent and tetrodotoxin (TTX)-resistant while sparing tetrodotoxin sensitive (TTX-S) voltage-gated Na+ channels as evidenced by the lack of further inhibition by the NaV1.8 blocker A-803467. The effects of the natural flavonoid were validated ex vivo in spinal cord slices where naringenin decreased both the frequency and amplitude of sEPSC recorded in neurons within the substantia gelatinosa. The antinociceptive potential of naringenin was evaluated in male and female mice. Naringenin had no effect on the nociceptive thresholds evoked by heat. Naringenin's reversed allodynia was in mouse models of postsurgical and neuropathic pain. Here, driven by a call by the National Center for Complementary and Integrative Health's strategic plan to advance fundamental research into basic biological mechanisms of the action of natural products, we advance the antinociceptive potential of the flavonoid naringenin.


Subject(s)
Analgesics/pharmacology , Flavanones/pharmacology , Ganglia, Spinal/cytology , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Nociception/drug effects , Sensory Receptor Cells/drug effects , Sodium Channel Blockers/pharmacology , Sodium/metabolism , Analgesics/chemistry , Analgesics/therapeutic use , Animals , Calcium Channels/drug effects , Calcium Signaling/drug effects , Excitatory Postsynaptic Potentials/drug effects , Female , Flavanones/chemistry , Flavanones/metabolism , Flavanones/therapeutic use , Hyperalgesia/drug therapy , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neuralgia/drug therapy , Pain, Postoperative/drug therapy , Protein Conformation , Protein Interaction Mapping , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/classification , Sensory Receptor Cells/metabolism , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/therapeutic use , Specific Pathogen-Free Organisms , Structure-Activity Relationship
6.
Respir Physiol Neurobiol ; 270: 103267, 2019 12.
Article in English | MEDLINE | ID: mdl-31398537

ABSTRACT

Cough in respiratory diseases is attributed to the activation of airway C-fibers by inflammation. Inflammatory mediators can act on multiple receptors expressed in airway C-fibers, nonetheless, the action potential initiation in C-fibers depends on a limited number of voltage-gated sodium channel (NaV1) subtypes. We have recently demonstrated that NaV1.8 substantially contributes to the action potential initiation in the airway C-fiber subtype implicated in cough. We therefore hypothesized that the NaV1.8 blocker A-803467 inhibits cough. We evaluated the cough evoked by the inhalation of C-fiber activator capsaicin in awake guinea pigs. Compared to vehicle, intraperitoneal or inhaled A-803467 caused 30-50% inhibition of cough at the doses that did not alter respiratory rate. We conclude that the NaV1.8 blocker A-803467 inhibits cough in a manner consistent with its action on the C-fiber nerve terminals in the airways. Targeting voltage-gated sodium channels mediating action potential initiation in airway C-fibers may offer a means of cough inhibition that is independent of the stimulus.


Subject(s)
Aniline Compounds/therapeutic use , Antitussive Agents/therapeutic use , Cough/drug therapy , Furans/therapeutic use , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Sodium Channel Blockers/therapeutic use , Action Potentials/drug effects , Administration, Inhalation , Aniline Compounds/adverse effects , Animals , Antitussive Agents/adverse effects , Bronchi/innervation , Dose-Response Relationship, Drug , Furans/adverse effects , Guinea Pigs , Injections, Intraperitoneal , Lung/innervation , Male , Nerve Fibers, Unmyelinated/drug effects , Presynaptic Terminals/drug effects , Sodium Channel Blockers/adverse effects
7.
J Biol Chem ; 294(18): 7324-7334, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30804211

ABSTRACT

The voltage-gated sodium channel Nav1.8 is preferentially expressed in peripheral nociceptive neurons and contributes to inflammatory and neuropathic pain. Therefore, Nav1.8 has emerged as one of the most promising analgesic targets for pain relief. Using large-scale screening of various animal-derived toxins and venoms for Nav1.8 inhibitors, here we identified µ-EPTX-Na1a, a 62-residue three-finger peptide from the venom of the Chinese cobra (Naja atra), as a potent inhibitor of Nav1.8, exhibiting high selectivity over other voltage-gated sodium channel subtypes. Using whole-cell voltage-clamp recordings, we observed that purified µ-EPTX-Na1a blocked the Nav1.8 current. This blockade was associated with a depolarizing shift of activation and repolarizing shift of inactivation, a mechanism distinct from that of any other gating modifier toxin identified to date. In rodent models of inflammatory and neuropathic pain, µ-EPTX-Na1a alleviated nociceptive behaviors more potently than did morphine, indicating that µ-EPTX-Na1a has a potent analgesic effect. µ-EPTX-Na1a displayed no evident cytotoxicity and cardiotoxicity and produced no obvious adverse responses in mice even at a dose 30-fold higher than that producing a significant analgesic effect. Our study establishes µ-EPTX-Na1a as a promising lead for the development of Nav1.8-targeting analgesics to manage pain.


Subject(s)
Elapid Venoms/chemistry , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Sodium Channel Blockers/pharmacology , Analgesics/adverse effects , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Mice , Naja naja , Neuralgia/drug therapy , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
8.
Cardiovasc Drugs Ther ; 33(6): 649-660, 2019 12.
Article in English | MEDLINE | ID: mdl-31916131

ABSTRACT

PURPOSE: Several studies have indicated a potential role for SCN10A/NaV1.8 in modulating cardiac electrophysiology and arrhythmia susceptibility. However, by which mechanism SCN10A/NaV1.8 impacts on cardiac electrical function is still a matter of debate. To address this, we here investigated the functional relevance of NaV1.8 in atrial and ventricular cardiomyocytes (CMs), focusing on the contribution of NaV1.8 to the peak and late sodium current (INa) under normal conditions in different species. METHODS: The effects of the NaV1.8 blocker A-803467 were investigated through patch-clamp analysis in freshly isolated rabbit left ventricular CMs, human left atrial CMs and human-induced pluripotent stem cell-derived CMs (hiPSC-CMs). RESULTS: A-803467 treatment caused a slight shortening of the action potential duration (APD) in rabbit CMs and hiPSC-CMs, while it had no effect on APD in human atrial cells. Resting membrane potential, action potential (AP) amplitude, and AP upstroke velocity were unaffected by A-803467 application. Similarly, INa density was unchanged after exposure to A-803467 and NaV1.8-based late INa was undetectable in all cell types analysed. Finally, low to absent expression levels of SCN10A were observed in human atrial tissue, rabbit ventricular tissue and hiPSC-CMs. CONCLUSION: We here demonstrate the absence of functional NaV1.8 channels in non-diseased atrial and ventricular CMs. Hence, the association of SCN10A variants with cardiac electrophysiology observed in, e.g. genome wide association studies, is likely the result of indirect effects on SCN5A expression and/or NaV1.8 activity in cell types other than CMs.


Subject(s)
Atrial Appendage/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , NAV1.8 Voltage-Gated Sodium Channel/deficiency , Action Potentials , Animals , Atrial Appendage/cytology , Atrial Appendage/drug effects , Cell Line , Heart Ventricles/cytology , Heart Ventricles/drug effects , Humans , Induced Pluripotent Stem Cells/metabolism , Kinetics , Male , Myocytes, Cardiac/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/genetics , Rabbits , Species Specificity , Voltage-Gated Sodium Channel Blockers/pharmacology
9.
Neurosci Lett ; 696: 67-73, 2019 03 23.
Article in English | MEDLINE | ID: mdl-30528877

ABSTRACT

Fenamates are N-substituted anthranilic acid derivatives, clinically used as nonsteroidal anti-inflammatory drugs (NSAIDs) in fever, pain and inflammation treatments. Previous studies have shown that they are also modulators of diverse ion channels, exhibiting either activation or inhibitory effects. However, the effects of fenamates on sodium channel subtypes are still unknown. In this study, fenamates, including mefenamic acid, flufenamic acid and tolfenamic acid, were examined by whole-cell patch clamp techniques on the sodium channels hNav1.7 and hNav1.8, which are closely associated with pain. The results showed that the mefenamic acid, flufenamic acid, and tolfenamic acid inhibited the peak currents of hNav1.7 and hNav1.8 in CHO cells stably expressing hNav1.7 and hNav1.8. However, much lighter inhibition effects of hNav1.8 were registered in the experimental system. Furthermore, the mefenamic acid, flufenamic acid and tolfenamic acid significantly affected the inactivation processes of hNav1.7 and hNav1.8 with I-V curves left-shifted to hyperpolarized direction. These data indicate that the inhibition effects of Nav1.7 and Nav1.8 by mefenamic acid, flufenamic acid and tolfenamic acid might contribute to their analgesic activity in addition to their inhibition of cyclooxygenase. These findings provide a basis for further studies in the discovery of other potential targets for NSAIDs.


Subject(s)
Fenamates/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Pain/drug therapy , Analgesics/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus/metabolism , Humans , ortho-Aminobenzoates/pharmacology
10.
Cardiovasc Res ; 114(13): 1728-1737, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29931291

ABSTRACT

Aims: In heart failure (HF), enhanced persistent Na+ current (INaL) exerts detrimental effects on cellular electrophysiology and can induce arrhythmias. However, the underlying regulatory mechanisms remain unclear. Our aim was to potentially investigate the regulation and electrophysiological contribution of neuronal sodium channel NaV1.8 in failing human heart and eventually to reveal a novel anti-arrhythmic therapy. Methods and results: By western blot, we found that NaV1.8 protein expression is significantly up-regulated, while of the predominant cardiac isoform NaV1.5 is inversely reduced in human HF. Furthermore, to investigate the relation of NaV1.8 regulation with the cellular proarrhythmic events, we performed comprehensive electrophysiology recordings and explore the effect of NaV1.8 on INaL, action potential duration (APD), Ca2+ spark frequency, and arrhythmia induction in human failing cardiomyocytes. NaV1.8 inhibition with the specific blockers A-803467 and PF-01247324 decreased INaL, abbreviated APD and reduced cellular-spontaneous Ca2+-release and proarrhythmic events in human failing cardiomyocytes. Consistently, in mouse cardiomyocytes stressed with isoproterenol, pharmacologic inhibition and genetically knockout of NaV1.8 (SCN10A-/-), were associated with reduced INaL and abbreviated APD. Conclusion: We provide first evidence of differential regulation of NaV1.8 and NaV1.5 in the failing human myocardium and their contribution to arrhythmogenesis due to generation of INaL. We propose inhibition of NaV1.8 thus constitutes a promising novel approach for selective anti-arrhythmic therapy in HF.


Subject(s)
Arrhythmias, Cardiac/etiology , Heart Failure/complications , Heart Rate/drug effects , Myocytes, Cardiac/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Action Potentials , Aged , Animals , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Calcium Signaling , Case-Control Studies , Female , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Male , Mice, Knockout , Middle Aged , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/genetics , Time Factors , Up-Regulation , Voltage-Gated Sodium Channel Blockers/pharmacology
11.
J Physiol ; 596(10): 1863-1872, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29193176

ABSTRACT

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


Subject(s)
NAV1.8 Voltage-Gated Sodium Channel/physiology , Toxins, Biological/pharmacology , Animals , Humans , Ion Channel Gating , Membrane Potentials , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Voltage-Gated Sodium Channel Blockers/pharmacology
12.
Naunyn Schmiedebergs Arch Pharmacol ; 390(12): 1255-1270, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28905186

ABSTRACT

Tricyclic antidepressants (TCAs) and duloxetine are used to treat neuropathic pain. However, the mechanisms underlying their analgesic effects remain unclear. Although many investigators have shown inhibitory effects of antidepressants on voltage-gated sodium channels (Nav) as a possible mechanism of analgesia, to our knowledge, no one has compared effects on the diverse variety of sodium channel α subunits. We investigated the effects of antidepressants on sodium currents in Xenopus oocytes expressing Nav1.2, Nav1.3, Nav1.6, Nav1.7, and Nav1.8 with a ß1 subunit by using whole-cell, two-electrode, voltage clamp techniques. We also studied the role of the ß3 subunit on the effect of antidepressants on Nav1.3. All antidepressants inhibited sodium currents in an inactivated state induced by all five α subunits with ß1. The inhibitory effects were more potent for Nav1.3, Nav1.7, and Nav1.8, which are distributed in dorsal root ganglia, than Nav1.2 and Nav1.6, which are distributed primarily in the central nervous system. The effect of amitriptyline on Nav1.7 with ß1 was most potent with a half-maximal inhibitory concentration (IC50) 4.6 µmol/L. IC50 for amitriptyline on Nav1.3 coexpressed with ß1 was lowered from 8.4 to 4.5 µmol/L by coexpression with ß3. Antidepressants predominantly inhibited the sodium channels expressed in dorsal root ganglia, and amitriptyline has the most potent inhibitory effect. This is the first evidence, to our knowledge, showing the diverse effects of antidepressants on various α subunits. Moreover, the ß3 subunit appears important for inhibition of Nav1.3. These findings may aid better understanding of the mechanisms underlying the pain relieving effects of antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , NAV1.3 Voltage-Gated Sodium Channel/drug effects , NAV1.7 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Neurons/drug effects , Neurons/metabolism , Sodium Channel Blockers/pharmacology , Amitriptyline/pharmacology , Animals , Antidepressive Agents, Tricyclic/pharmacology , Duloxetine Hydrochloride/pharmacology , Ganglia, Spinal/drug effects , NAV1.2 Voltage-Gated Sodium Channel/drug effects , NAV1.6 Voltage-Gated Sodium Channel/drug effects , Oocytes , Patch-Clamp Techniques , Rats , Xenopus
13.
Sci Rep ; 7(1): 7574, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28790377

ABSTRACT

The analgesic studies on Stauntonia brachyanthera, a traditional Chinese folk medicine used to treat headache, pains and inflammatory diseases in local areas, showed that the EtOH extracts (EESB) and the characteristic ingredient YM11 could significantly inhibit the acetic acid-induced writhing responses by 43.1% and 78.95%, and decrease the xylene-induced ear edemas by 48.9% and 21.4%, respectively. EESB could significantly increase pain threshold of mice in hot-plate test, but the effect of YM11 was not obviously. Further study in formalin test showed the inhibitory effect of YM11 in 2nd phase was more significant than that in 1st phase, revealed the peripheral analgesic activity of YM11. The ELISA and Western Blot analysis suggested that the analgesic mechanisms of YM11 were related to the inhibitions of the expressions of TNF-α, IL-1ß and IL-6, and down-regulations of Nav1.8 protein in the left side of L4-6 DRG regulated by MAPKs, in which the levels of p-ERK, p-JNK and p-p38 were all decreased. In addition, the electrophysiological experiments indicated YM11 could reduce the Nav1.8 currents by 46.01% in small-diameter DRG neurons. Therefore, the analgesic activity of S. brachyanthera might be based on the regulation of inflammatory mediators and the directly control of the sodium channel prompt.


Subject(s)
Analgesics/pharmacology , Inflammation Mediators/antagonists & inhibitors , NAV1.8 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Plant Extracts/pharmacology , Ranunculales/chemistry , Analgesics/isolation & purification , Animals , Ganglia, Spinal/drug effects , Mice , Neurons/drug effects , Plant Extracts/isolation & purification , Rats
14.
Pulm Pharmacol Ther ; 47: 38-41, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28522215

ABSTRACT

Pathological cough is thought to be secondary to inappropriate activation of vagal sensory nerves. Sensory nerves can be activated by a large number of disparate stimuli. The most relevant stimuli to block for effective anti-tussive therapy likely depend on the specific underlying pathology that is leading to the coughing. Blocking voltage-gated sodium channels (NaV) will prevent action potential initiation and conduction and therefore prevent sensory communication between the airways and brainstem. In so doing, they would be expected to inhibit evoked cough independently of the nature of the stimulus and underlying pathology. There are nine subtypes of NaVs each with distinct pore-forming alpha subunits referred to NaV1.1-1.9. Among these channels, based on functional and genetic analysis of cough causing vagal afferent nerve subtypes, we hypothesize that targeting NaV1.7 and NaV1.8 is a rational strategy forward for the effective treatment of pathological cough.


Subject(s)
Antitussive Agents/therapeutic use , Cough/drug therapy , Voltage-Gated Sodium Channel Blockers/therapeutic use , Animals , Antitussive Agents/pharmacology , Cough/physiopathology , Humans , NAV1.7 Voltage-Gated Sodium Channel/drug effects , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Vagus Nerve/metabolism , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/drug effects , Voltage-Gated Sodium Channels/metabolism
15.
J Neurosci ; 37(20): 5204-5214, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28450535

ABSTRACT

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


Subject(s)
Axons/physiology , NAV1.8 Voltage-Gated Sodium Channel/physiology , Neural Conduction/physiology , Peripheral Nerves/physiology , Skin/innervation , Tetrodotoxin/administration & dosage , Afferent Pathways/drug effects , Afferent Pathways/physiology , Animals , Axons/drug effects , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Macaca nemestrina , Male , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Nerve Fibers, Unmyelinated , Neural Conduction/drug effects , Peripheral Nerves/drug effects , Skin/drug effects , Skin Physiological Phenomena/drug effects , Voltage-Gated Sodium Channel Blockers/administration & dosage
16.
Mediators Inflamm ; 2015: 124942, 2015.
Article in English | MEDLINE | ID: mdl-26504355

ABSTRACT

Tumor necrosis factor- (TNF-) α is a proinflammatory cytokine involved in the development and maintenance of inflammatory and neuropathic pain. Its effects are mediated by two receptors, TNF receptor-1 (TNFR-1) and TNF receptor-2 (TNFR-2). These receptors play a crucial role in the sensitization of voltage-gated sodium channels (VGSCs), a key mechanism in the pathogenesis of chronic pain. Using the whole-cell patch-clamp technique, we examined the influence of TNFR-1 and TNFR-2 on VGSCs and TTX-resistant NaV1.8 channels in isolated rat dorsal root ganglion neurons by using selective TNFR agonists. The TNFR-1 agonist R32W (10 pg/mL) caused an increase in the VGSC current (I(Na(V))) by 27.2 ± 5.1%, while the TNFR-2 agonist D145 (10 pg/mL) increased the current by 44.9 ± 2.6%. This effect was dose dependent. Treating isolated NaV1.8 with R32W (100 pg/mL) resulted in an increase in I(NaV(1.8)) by 18.9 ± 1.6%, while treatment with D145 (100 pg/mL) increased the current by 14.5 ± 3.7%. Based on the current-voltage relationship, 10 pg of R32W or D145 led to an increase in I(Na(V)) in a bell-shaped, voltage-dependent manner with a maximum effect at -30 mV. The effects of TNFR activation on VGSCs promote excitation in primary afferent neurons and this might explain the sensitization mechanisms associated with neuropathic and inflammatory pain.


Subject(s)
Ganglia, Spinal/metabolism , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Neurons/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/chemistry , Voltage-Gated Sodium Channels/metabolism , Animals , Cytokines/metabolism , Dose-Response Relationship, Drug , Inflammation , Male , Neuralgia/drug therapy , Neurons, Afferent/drug effects , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type II/agonists
17.
Eur J Pharmacol ; 764: 395-403, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26187311

ABSTRACT

Antidepressant drugs of the SSRI family are used as a third-line treatment for neuropathic pain. In contrast MAOi antidepressants, that also increase extracellular serotonin bioavailability have little or no effects on this condition. In addition to their action of the serotonin transporter, some SSRI have been shown to inhibit voltage gated sodium channels. Here we investigated the potential inhibition of SSRIs and MAOi antidepressants on Nav1.7 or Nav1.8, which are expressed in sensory neurons and play an important role in pain sensation. We used the whole-cell patch-clamp technique on HEK293 cells expressing either Nav1.7 or Nav1.8, and evaluated the effects of the SSRIs fluoxetine, paroxetine, and citalopram as well as one MAOi antidepressants on the electrophysiological properties of the Na(+) channels. Paroxetine exhibited the greatest affinity for Na(+) channels. In ascending order of affinity for Nav1.7 were paroxetine (IC50=10 µM), followed by fluoxetine (IC50=66 µM), then citalopram (IC50=174 µM). In ascending order of affinity for Nav1.8 were paroxetine (IC50=9 µM), followed by fluoxetine (IC50=49 µM), then citalopram (IC50=100 µM). Paroxetine and fluoxetine accelerated the onset of slow-inactivation and delayed the time-course of recovery from inactivation for both channels. Paroxetine and fluoxetine also had a prominent effect on the frequency-dependent inhibition, with a greater effect on Nav1.7. In contrast to SSRIs, MAOi did not affect Na(+) channels currents. These results suggest that, in certain conditions, the analgesic effect of SSRIs may in part be due to their interactions with Na(+) channels.


Subject(s)
Analgesics/pharmacology , Antidepressive Agents/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Sodium Channel Blockers/pharmacology , Citalopram/pharmacology , Dose-Response Relationship, Drug , Fluoxetine/pharmacology , HEK293 Cells , Humans , Membrane Potentials , Moclobemide/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Paroxetine/pharmacology , Patch-Clamp Techniques , Time Factors , Transfection
18.
Br J Pharmacol ; 172(10): 2654-70, 2015 May.
Article in English | MEDLINE | ID: mdl-25625641

ABSTRACT

BACKGROUND AND PURPOSE: NaV 1.8 ion channels have been highlighted as important molecular targets for the design of low MW blockers for the treatment of chronic pain. Here, we describe the effects of PF-01247324, a new generation, selective, orally bioavailable Nav 1.8 channel blocker of novel chemotype. EXPERIMENTAL APPROACH: The inhibition of Nav 1.8 channels by PF-01247324 was studied using in vitro patch-clamp electrophysiology and the oral bioavailability and antinociceptive effects demonstrated using in vivo rodent models of inflammatory and neuropathic pain. KEY RESULTS: PF-01247324 inhibited native tetrodotoxin-resistant (TTX-R) currents in human dorsal root ganglion (DRG) neurons (IC50 : 331 nM) and in recombinantly expressed h Nav 1.8 channels (IC50 : 196 nM), with 50-fold selectivity over recombinantly expressed TTX-R hNav 1.5 channels (IC50 : ∼10 µM) and 65-100-fold selectivity over TTX-sensitive (TTX-S) channels (IC50 : ∼10-18 µM). Native TTX-R currents in small-diameter rodent DRG neurons were inhibited with an IC50 448 nM, and the block of both human recombinant Nav 1.8 channels and TTX-R from rat DRG neurons was both frequency and state dependent. In vitro current clamp showed that PF-01247324 reduced excitability in both rat and human DRG neurons and also altered the waveform of the action potential. In vivo experiments n rodents demonstrated efficacy in both inflammatory and neuropathic pain models. CONCLUSIONS AND IMPLICATIONS: Using PF-01247324, we have confirmed a role for Nav 1.8 channels in both inflammatory and neuropathic pain. We have also demonstrated a key role for Nav 1.8 channels in action potential upstroke and repetitive firing of rat and human DRG neurons.


Subject(s)
Nociception/drug effects , Picolinic Acids/pharmacology , Sensory Receptor Cells/drug effects , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Administration, Oral , Animals , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Membrane Potentials/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Pain Measurement/drug effects , Picolinic Acids/administration & dosage , Picolinic Acids/pharmacokinetics , Rats , Tetrodotoxin/antagonists & inhibitors , Tetrodotoxin/pharmacology
20.
Anesthesiology ; 121(3): 620-31, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24809977

ABSTRACT

BACKGROUND: The neurosteroids allopregnanolone and pregnanolone are potent positive modulators of γ-aminobutyric acid type A receptors. Antinociceptive effects of allopregnanolone have attracted much attention because recent reports have indicated the potential of allopregnanolone as a therapeutic agent for refractory pain. However, the analgesic mechanisms of allopregnanolone are still unclear. Voltage-gated sodium channels (Nav) are thought to play important roles in inflammatory and neuropathic pain, but there have been few investigations on the effects of allopregnanolone on sodium channels. METHODS: Using voltage-clamp techniques, the effects of allopregnanolone sulfate (APAS) and pregnanolone sulfate (PAS) on sodium current were examined in Xenopus oocytes expressing Nav1.2, Nav1.6, Nav1.7, and Nav1.8 α subunits. RESULTS: APAS suppressed sodium currents of Nav1.2, Nav1.6, and Nav1.7 at a holding potential causing half-maximal current in a concentration-dependent manner, whereas it markedly enhanced sodium current of Nav1.8 at a holding potential causing maximal current. Half-maximal inhibitory concentration values for Nav1.2, Nav1.6, and Nav1.7 were 12 ± 4 (n = 6), 41 ± 2 (n = 7), and 131 ± 15 (n = 5) µmol/l (mean ± SEM), respectively. The effects of PAS were lower than those of APAS. From gating analysis, two compounds increased inactivation of all α subunits, while they showed different actions on activation of each α subunit. Moreover, two compounds showed a use-dependent block on Nav1.2, Nav1.6, and Nav1.7. CONCLUSION: APAS and PAS have diverse effects on sodium currents in oocytes expressing four α subunits. APAS inhibited the sodium currents of Nav1.2 most strongly.


Subject(s)
Pregnanolone/pharmacology , Voltage-Gated Sodium Channels/drug effects , Animals , Female , NAV1.2 Voltage-Gated Sodium Channel/drug effects , NAV1.6 Voltage-Gated Sodium Channel/drug effects , NAV1.7 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Receptors, GABA-A/drug effects , Voltage-Gated Sodium Channels/physiology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...