Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
1.
J Neurosci Res ; 102(10): e25386, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39364619

ABSTRACT

Voltage-gated sodium channels, including NaV1.7, NaV1.8, and NaV1.9, play important roles in pain transmission and chronic pain development. However, the specific mechanisms of their action remain unclear, highlighting the need for in vivo stimulation studies of these channels. Optogenetics, a novel technique for targeting the activation or inhibition of specific neural circuits using light, offers a promising solution. In our previous study, we used optogenetics to selectively excite NaV1.7-expressing neurons in the dorsal root ganglion of mice to induce nocifensive behavior. Here, we further characterize the impact of nocifensive behavior by activation of NaV1.7, NaV1.8, or NaV1.9-expressing neurons. Using CRISPR/Cas9-mediated homologous recombination, NaV1.7-iCre, NaV1.8-iCre, or NaV1.9-iCre mice expressing iCre recombinase under the control of the endogenous NaV1.7, NaV1.8, or NaV1.9 gene promoter were produced. These mice were then bred with channelrhodopsin-2 (ChR2) Cre-reporter Ai32 mice to obtain NaV1.7-ChR2, NaV1.8-ChR2, or NaV1.9-ChR2 mice. Blue light exposure triggered paw withdrawal in all mice, with the strongest response in NaV1.8-ChR2 mice. These light sensitivity differences observed across NaV1.x-ChR2 mice may be dependent on ChR2 expression or reflect the inherent disparities in their pain transmission roles. In conclusion, we have generated noninvasive pain models, with optically activated peripheral nociceptors. We believe that studies using optogenetics will further elucidate the role of sodium channel subtypes in pain transmission.


Subject(s)
Ganglia, Spinal , NAV1.7 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel , NAV1.9 Voltage-Gated Sodium Channel , Optogenetics , Animals , Optogenetics/methods , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Mice , NAV1.9 Voltage-Gated Sodium Channel/genetics , Ganglia, Spinal/metabolism , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Mice, Transgenic , Male , Nociception/physiology
2.
J Gen Physiol ; 156(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39378238

ABSTRACT

While voltage-gated sodium channels Nav1.7 and Nav1.8 both contribute to electrogenesis in dorsal root ganglion (DRG) neurons, details of their interactions have remained unexplored. Here, we studied the functional contribution of Nav1.8 in DRG neurons using a dynamic clamp to express Nav1.7L848H, a gain-of-function Nav1.7 mutation that causes inherited erythromelalgia (IEM), a human genetic model of neuropathic pain, and demonstrate a profound functional interaction of Nav1.8 with Nav1.7 close to the threshold for AP generation. At the voltage threshold of -21.9 mV, we observed that Nav1.8 channel open-probability exceeded Nav1.7WT channel open-probability ninefold. Using a kinetic model of Nav1.8, we showed that a reduction of Nav1.8 current by even 25-50% increases rheobase and reduces firing probability in small DRG neurons expressing Nav1.7L848H. Nav1.8 subtraction also reduces the amplitudes of subthreshold membrane potential oscillations in these cells. Our results show that within DRG neurons that express peripheral sodium channel Nav1.7, the Nav1.8 channel amplifies excitability at a broad range of membrane voltages with a predominant effect close to the AP voltage threshold, while Nav1.7 plays a major role at voltages closer to resting membrane potential. Our data show that dynamic-clamp reduction of Nav1.8 conductance by 25-50% can reverse hyperexcitability of DRG neurons expressing a gain-of-function Nav1.7 mutation that causes pain in humans and suggests, more generally, that full inhibition of Nav1.8 may not be required for relief of pain due to DRG neuron hyperexcitability.


Subject(s)
Ganglia, Spinal , NAV1.7 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel , Neuralgia , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , Ganglia, Spinal/metabolism , Animals , Neuralgia/metabolism , Neuralgia/physiopathology , Neurons/metabolism , Neurons/physiology , Action Potentials , Mice , Humans , Rats
3.
eNeuro ; 11(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-39256039

ABSTRACT

Alteration of synaptic function in the dorsal horn (DH) has been implicated as a cellular substrate for the development of neuropathic pain, but certain details remain unclear. In particular, the lack of information on the types of synapses that undergo functional changes hinders the understanding of disease pathogenesis from a synaptic plasticity perspective. Here, we addressed this issue by using optogenetic and retrograde tracing ex vivo to selectively stimulate first-order nociceptors expressing Nav1.8 (NRsNav1.8) and record the responses of spinothalamic tract neurons in spinal lamina I (L1-STTNs). We found that spared nerve injury (SNI) increased excitatory postsynaptic currents (EPSCs) in L1-STTNs evoked by photostimulation of NRsNav1.8 (referred to as Nav1.8-STTN EPSCs). This effect was accompanied by a significant change in the failure rate and paired-pulse ratio of synaptic transmission from NRsNav1.8 to L1-STTN and in the frequency (not amplitude) of spontaneous EPSCs recorded in L1-STTNs. However, no change was observed in the ratio of AMPA to NMDA receptor-mediated components of Nav1.8-STTN EPSCs or in the amplitude of unitary EPSCs constituting Nav1.8-STTN EPSCs recorded with extracellular Ca2+ replaced by Sr2+ In addition, there was a small increase (approximately 10%) in the number of L1-STTNs showing immunoreactivity for phosphorylated extracellular signal-regulated kinases in mice after SNI compared with sham. Similarly, only a small percentage of L1-STTNs showed a lower action potential threshold after SNI. In conclusion, our results show that SNI induces presynaptic modulation at NRNav1.8 (consisting of both peptidergic and nonpeptidergic nociceptors) synapses on L1-STTNs forming the lateral spinothalamic tract.


Subject(s)
Excitatory Postsynaptic Potentials , NAV1.8 Voltage-Gated Sodium Channel , Nociceptors , Spinothalamic Tracts , Synaptic Transmission , Animals , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , Nociceptors/metabolism , Nociceptors/physiology , Spinothalamic Tracts/metabolism , Excitatory Postsynaptic Potentials/physiology , Male , Synaptic Transmission/physiology , Mice , Optogenetics , Mice, Inbred C57BL , Mice, Transgenic
4.
Sci Rep ; 14(1): 17360, 2024 07 29.
Article in English | MEDLINE | ID: mdl-39075089

ABSTRACT

Prostaglandin E2 (PGE2) is a major contributor to inflammatory pain hyperalgesia, however, the extent to which it modulates the activity of nociceptive axons is incompletely understood. We developed and characterized a microfluidic cell culture model to investigate sensitisation of the axons of dorsal root ganglia neurons. We show that application of PGE2 to fluidically isolated axons leads to sensitisation of their responses to depolarising stimuli. Interestingly the application of PGE2 to the DRG axons elicited a direct and persistent spiking activity propagated to the soma. Both the persistent activity and the membrane depolarisation in the axons are abolished by the EP4 receptor inhibitor and a blocker of cAMP synthesis. Further investigated into the mechanisms of the spiking activity showed that the PGE2 evoked depolarisation was inhibited by Nav1.8 sodium channel blockers but was refractory to the application of TTX or zatebradine. Interestingly, the depolarisation of axons was blocked by blocking ANO1 channels with T16Ainh-A01. We further show that PGE2-elicited axonal responses are altered by the changes in chloride gradient within the axons following treatment with bumetanide a Na-K-2Cl cotransporter NKCC1 inhibitor, but not by VU01240551 an inhibitor of potassium-chloride transporter KCC2. Our data demonstrate a novel role for PGE2/EP4/cAMP pathway which culminates in a sustained depolarisation of sensory axons mediated by a chloride current through ANO1 channels. Therefore, using a microfluidic culture model, we provide evidence for a potential dual function of PGE2 in inflammatory pain: it sensitises depolarisation-evoked responses in nociceptive axons and directly triggers action potentials by activating ANO1 and Nav1.8 channels.


Subject(s)
Anoctamin-1 , Axons , Dinoprostone , Ganglia, Spinal , NAV1.8 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Animals , Dinoprostone/pharmacology , Dinoprostone/metabolism , Axons/metabolism , Axons/drug effects , Axons/physiology , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Rats , Anoctamin-1/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Action Potentials/drug effects , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Rats, Sprague-Dawley , Cells, Cultured , Solute Carrier Family 12, Member 2/metabolism , Cyclic AMP/metabolism
5.
Bioorg Med Chem Lett ; 110: 129862, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38944398

ABSTRACT

Chronic pain is a common and challenging clinical problem that significantly impacts patients' quality of life. The sodium channel Nav1.8 plays a crucial role in the occurrence and development of chronic pain, making it one of the key targets for treating chronic pain. In this article, we combined virtual screening with cell membrane chromatography techniques to establish a novel method for rapid high-throughput screening of selective Nav1.8 inhibitors. Using this approach, we identified a small molecule compound 6, which not only demonstrated high affinity and inhibitory activity against Nav1.8 but also exhibited significant inhibitory effects on CFA-induced chronic inflammatory pain. Compared to the positive drug VX-150, compound 6 showed a more prolonged analgesic effect, making it a promising candidate as a Nav1.8 inhibitor with potential clinical applications. This discovery provides a new therapeutic option for the treatment of chronic pain.


Subject(s)
Analgesics , NAV1.8 Voltage-Gated Sodium Channel , Sulfonamides , Analgesics/pharmacology , Analgesics/chemistry , Analgesics/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfonamides/chemical synthesis , Animals , Humans , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Structure-Activity Relationship , Benzenesulfonamides , Molecular Structure , Mice , Dose-Response Relationship, Drug , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/chemical synthesis
6.
Int J Mol Sci ; 25(11)2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38892333

ABSTRACT

The sodium channel NaV1.8, encoded by the SCN10A gene, has recently emerged as a potential regulator of cardiac electrophysiology. We have previously shown that NaV1.8 contributes to arrhythmogenesis by inducing a persistent Na+ current (late Na+ current, INaL) in human atrial and ventricular cardiomyocytes (CM). We now aim to further investigate the contribution of NaV1.8 to human ventricular arrhythmogenesis at the CM-specific level using pharmacological inhibition as well as a genetic knockout (KO) of SCN10A in induced pluripotent stem cell CM (iPSC-CM). In functional voltage-clamp experiments, we demonstrate that INaL was significantly reduced in ventricular SCN10A-KO iPSC-CM and in control CM after a specific pharmacological inhibition of NaV1.8. In contrast, we did not find any effects on ventricular APD90. The frequency of spontaneous sarcoplasmic reticulum Ca2+ sparks and waves were reduced in SCN10A-KO iPSC-CM and control cells following the pharmacological inhibition of NaV1.8. We further analyzed potential triggers of arrhythmias and found reduced delayed afterdepolarizations (DAD) in SCN10A-KO iPSC-CM and after the specific inhibition of NaV1.8 in control cells. In conclusion, we show that NaV1.8-induced INaL primarily impacts arrhythmogenesis at a subcellular level, with minimal effects on systolic cellular Ca2+ release. The inhibition or knockout of NaV1.8 diminishes proarrhythmic triggers in ventricular CM. In conjunction with our previously published results, this work confirms NaV1.8 as a proarrhythmic target that may be useful in an anti-arrhythmic therapeutic strategy.


Subject(s)
Arrhythmias, Cardiac , Heart Ventricles , Induced Pluripotent Stem Cells , Myocytes, Cardiac , NAV1.8 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Heart Ventricles/metabolism , Heart Ventricles/cytology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/etiology , Action Potentials/drug effects
7.
Biochem Biophys Res Commun ; 721: 150126, 2024 08 20.
Article in English | MEDLINE | ID: mdl-38776832

ABSTRACT

Voltage-gated sodium channel subtypes, Nav1.7, Nav1.8, and Nav1.9 are predominantly expressed in peripheral sensory neurons. Recent genetic studies have revealed that they are involved in pathological pain processing and that the blockade of Nav1.7, Nav1.8, or Nav1.9 will become a promising pharmacotherapy especially for neuropathic pain. A growing number of drug discovery programs have targeted either of the subtypes to obtain a selective inhibitor which can provide pain relief without affecting the cardiovascular and central nervous systems, though none of them has been approved yet. Here we describe the in vitro characteristics of ANP-230, a novel sodium channel blocker under clinical development. Surprisingly, ANP-230 was shown to block three pain-related subtypes, human Nav1.7, Nav1.8, and Nav1.9 with similar potency, but had only low inhibitory activity to human cardiac Nav1.5 channel and rat central Nav channels. The voltage clamp experiments using different step pulse protocols revealed that ANP-230 had a "tonic block" mode of action without state- and use-dependency. In addition, ANP-230 caused a depolarizing shift of the activation curve and decelerated gating kinetics in human Nav1.7-stably expressing cells. The depolarizing shift of activation curve was commonly observed in human Nav1.8-stably expressing cells as well as rat dorsal root ganglion neurons. These data suggested a quite unique mechanism of Nav channel inhibition by ANP-230. Finally, ANP-230 reduced excitability of rat dorsal root ganglion neurons in a concentration dependent manner. Collectively, these promising results indicate that ANP-230 could be a potent drug for neuropathic pain.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel , NAV1.9 Voltage-Gated Sodium Channel , Sodium Channel Blockers , Humans , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , Animals , Rats , NAV1.9 Voltage-Gated Sodium Channel/metabolism , NAV1.9 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/genetics , Sodium Channel Blockers/pharmacology , HEK293 Cells , Voltage-Gated Sodium Channel Blockers/pharmacology , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/cytology
8.
Elife ; 122024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687187

ABSTRACT

Nociceptive sensory neurons convey pain-related signals to the CNS using action potentials. Loss-of-function mutations in the voltage-gated sodium channel NaV1.7 cause insensitivity to pain (presumably by reducing nociceptor excitability) but clinical trials seeking to treat pain by inhibiting NaV1.7 pharmacologically have struggled. This may reflect the variable contribution of NaV1.7 to nociceptor excitability. Contrary to claims that NaV1.7 is necessary for nociceptors to initiate action potentials, we show that nociceptors can achieve similar excitability using different combinations of NaV1.3, NaV1.7, and NaV1.8. Selectively blocking one of those NaV subtypes reduces nociceptor excitability only if the other subtypes are weakly expressed. For example, excitability relies on NaV1.8 in acutely dissociated nociceptors but responsibility shifts to NaV1.7 and NaV1.3 by the fourth day in culture. A similar shift in NaV dependence occurs in vivo after inflammation, impacting ability of the NaV1.7-selective inhibitor PF-05089771 to reduce pain in behavioral tests. Flexible use of different NaV subtypes exemplifies degeneracy - achieving similar function using different components - and compromises reliable modulation of nociceptor excitability by subtype-selective inhibitors. Identifying the dominant NaV subtype to predict drug efficacy is not trivial. Degeneracy at the cellular level must be considered when choosing drug targets at the molecular level.


Subject(s)
Analgesics , Benzenesulfonamides , Nociceptors , Phenyl Ethers , Animals , Analgesics/pharmacology , Nociceptors/metabolism , Nociceptors/drug effects , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/genetics , Mice , Action Potentials/drug effects , Pain/drug therapy , Humans , Sodium Channels/metabolism , Sodium Channels/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics
9.
Brain ; 147(9): 3157-3170, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38447953

ABSTRACT

Vincristine-induced peripheral neuropathy is a common side effect of vincristine treatment, which is accompanied by pain and can be dose-limiting. The molecular mechanisms that underlie vincristine-induced pain are not well understood. We have established an animal model to investigate pathophysiological mechanisms of vincristine-induced pain. Our previous studies have shown that the tetrodotoxin-sensitive voltage-gated sodium channel Nav1.6 in medium-diameter dorsal root ganglion (DRG) neurons contributes to the maintenance of vincristine-induced allodynia. In this study, we investigated the effects of vincristine administration on excitability in small-diameter DRG neurons and whether the tetrodotoxin-resistant (TTX-R) Nav1.8 channels contribute to mechanical allodynia. Current-clamp recordings demonstrated that small DRG neurons become hyper-excitable following vincristine treatment, with both reduced current threshold and increased firing frequency. Using voltage-clamp recordings in small DRG neurons, we now show an increase in TTX-R current density and a -7.3 mV hyperpolarizing shift in the half-maximal potential (V1/2) of activation of Nav1.8 channels in vincristine-treated animals, which likely contributes to the hyperexcitability that we observed in these neurons. Notably, vincristine treatment did not enhance excitability of small DRG neurons from Nav1.8 knockout mice, and the development of mechanical allodynia was delayed but not abrogated in these mice. Together, our data suggest that sodium channel Nav1.8 in small DRG neurons contributes to the development of vincristine-induced mechanical allodynia.


Subject(s)
Ganglia, Spinal , Hyperalgesia , NAV1.8 Voltage-Gated Sodium Channel , Neurons , Vincristine , Animals , Vincristine/toxicity , Vincristine/pharmacology , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Male , Mice, Knockout , Tetrodotoxin/pharmacology , Action Potentials/drug effects , Mice, Inbred C57BL , Antineoplastic Agents, Phytogenic/toxicity , Patch-Clamp Techniques
10.
Inflamm Res ; 73(4): 669-691, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38483556

ABSTRACT

OBJECTIVE AND DESIGN: Our aim was to determine an age-dependent role of Nav1.8 and ASIC3 in dorsal root ganglion (DRG) neurons in a rat pre-clinical model of long-term inflammatory pain. METHODS: We compared 6 and 24 months-old female Wistar rats after cutaneous inflammation. We used behavioral pain assessments over time, qPCR, quantitative immunohistochemistry, selective pharmacological manipulation, ELISA and in vitro treatment with cytokines. RESULTS: Older rats exhibited delayed recovery from mechanical allodynia and earlier onset of spontaneous pain than younger rats after inflammation. Moreover, the expression patterns of Nav1.8 and ASIC3 were time and age-dependent and ASIC3 levels remained elevated only in aged rats. In vivo, selective blockade of Nav1.8 with A803467 or of ASIC3 with APETx2 alleviated mechanical and cold allodynia and also spontaneous pain in both age groups with slightly different potency. Furthermore, in vitro IL-1ß up-regulated Nav1.8 expression in DRG neurons cultured from young but not old rats. We also found that while TNF-α up-regulated ASIC3 expression in both age groups, IL-6 and IL-1ß had this effect only on young and aged neurons, respectively. CONCLUSION: Inflammation-associated mechanical allodynia and spontaneous pain in the elderly can be more effectively treated by inhibiting ASIC3 than Nav1.8.


Subject(s)
Acid Sensing Ion Channels , Hyperalgesia , NAV1.8 Voltage-Gated Sodium Channel , Pain , Animals , Female , Rats , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Acid Sensing Ion Channels/pharmacology , Analgesics/therapeutic use , Ganglia, Spinal , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Inflammation/metabolism , Pain/drug therapy , Pain/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Sensory Receptor Cells/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism
11.
Nature ; 628(8008): 604-611, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538784

ABSTRACT

The immune system has a critical role in orchestrating tissue healing. As a result, regenerative strategies that control immune components have proved effective1,2. This is particularly relevant when immune dysregulation that results from conditions such as diabetes or advanced age impairs tissue healing following injury2,3. Nociceptive sensory neurons have a crucial role as immunoregulators and exert both protective and harmful effects depending on the context4-12. However, how neuro-immune interactions affect tissue repair and regeneration following acute injury is unclear. Here we show that ablation of the NaV1.8 nociceptor impairs skin wound repair and muscle regeneration after acute tissue injury. Nociceptor endings grow into injured skin and muscle tissues and signal to immune cells through the neuropeptide calcitonin gene-related peptide (CGRP) during the healing process. CGRP acts via receptor activity-modifying protein 1 (RAMP1) on neutrophils, monocytes and macrophages to inhibit recruitment, accelerate death, enhance efferocytosis and polarize macrophages towards a pro-repair phenotype. The effects of CGRP on neutrophils and macrophages are mediated via thrombospondin-1 release and its subsequent autocrine and/or paracrine effects. In mice without nociceptors and diabetic mice with peripheral neuropathies, delivery of an engineered version of CGRP accelerated wound healing and promoted muscle regeneration. Harnessing neuro-immune interactions has potential to treat non-healing tissues in which dysregulated neuro-immune interactions impair tissue healing.


Subject(s)
Calcitonin Gene-Related Peptide , Macrophages , Neutrophils , Nociceptors , Wound Healing , Animals , Mice , Autocrine Communication , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Efferocytosis , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , Muscle, Skeletal , NAV1.8 Voltage-Gated Sodium Channel/deficiency , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Nociceptors/metabolism , Paracrine Communication , Peripheral Nervous System Diseases/complications , Receptor Activity-Modifying Protein 1/metabolism , Regeneration/drug effects , Skin , Thrombospondin 1/metabolism , Wound Healing/drug effects , Wound Healing/immunology , Humans , Male , Female
12.
Int J Mol Sci ; 24(12)2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37373335

ABSTRACT

In heart failure and atrial fibrillation, a persistent Na+ current (INaL) exerts detrimental effects on cellular electrophysiology and can induce arrhythmias. We have recently shown that NaV1.8 contributes to arrhythmogenesis by inducing a INaL. Genome-wide association studies indicate that mutations in the SCN10A gene (NaV1.8) are associated with increased risk for arrhythmias, Brugada syndrome, and sudden cardiac death. However, the mediation of these NaV1.8-related effects, whether through cardiac ganglia or cardiomyocytes, is still a subject of controversial discussion. We used CRISPR/Cas9 technology to generate homozygous atrial SCN10A-KO-iPSC-CMs. Ruptured-patch whole-cell patch-clamp was used to measure the INaL and action potential duration. Ca2+ measurements (Fluo 4-AM) were performed to analyze proarrhythmogenic diastolic SR Ca2+ leak. The INaL was significantly reduced in atrial SCN10A KO CMs as well as after specific pharmacological inhibition of NaV1.8. No effects on atrial APD90 were detected in any groups. Both SCN10A KO and specific blockers of NaV1.8 led to decreased Ca2+ spark frequency and a significant reduction of arrhythmogenic Ca2+ waves. Our experiments demonstrate that NaV1.8 contributes to INaL formation in human atrial CMs and that NaV1.8 inhibition modulates proarrhythmogenic triggers in human atrial CMs and therefore NaV1.8 could be a new target for antiarrhythmic strategies.


Subject(s)
Atrial Fibrillation , Heart Failure , Humans , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Genome-Wide Association Study , Anti-Arrhythmia Agents/pharmacology , Myocytes, Cardiac/metabolism , Heart Failure/metabolism , Action Potentials , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism
13.
Philos Trans R Soc Lond B Biol Sci ; 378(1879): 20220175, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37122207

ABSTRACT

We illustrate use of induced pluripotent stem cells (iPSCs) as platforms for investigating cardiomyocyte phenotypes in a human family pedigree exemplified by novel heterozygous RYR2-A1855D and SCN10A-Q1362H variants occurring alone and in combination. The proband, a four-month-old boy, presented with polymorphic ventricular tachycardia. Genetic tests revealed double novel heterozygous RYR2-A1855D and SCN10A-Q1362H variants inherited from his father (F) and mother (M), respectively. His father showed ventricular premature beats; his mother was asymptomatic. Molecular biological characterizations demonstrated greater TNNT2 messenger RNA (mRNA) expression in the iPSCs-induced cardiomyocytes (iPS-CMs) than in the iPSCs. Cardiac troponin Ts became progressively organized but cytoplasmic RYR2 and SCN10A aggregations occurred in the iPS-CMs. Proband-specific iPS-CMs showed decreased RYR2 and SCN10A mRNA expression. The RYR2-A1855D variant resulted in premature spontaneous sarcoplasmic reticular Ca2+ transients, Ca2+ oscillations and increased action potential durations. SCN10A-Q1362H did not confer any specific phenotype. However, the combined heterozygous RYR2-A1855D and SCN10A-Q1362H variants in the proband iPS-CMs resulted in accentuated Ca2+ homeostasis disorders, action potential prolongation and susceptibility to early afterdepolarizations at high stimulus frequencies. These findings attribute the clinical phenotype in the proband to effects of the heterozygous RYR2 variant exacerbated by heterozygous SCN10A modification. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.


Subject(s)
Induced Pluripotent Stem Cells , Tachycardia, Ventricular , Humans , Infant , Male , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Homeostasis , Mutation , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/pharmacology , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism
14.
Nature ; 615(7952): 472-481, 2023 03.
Article in English | MEDLINE | ID: mdl-36859544

ABSTRACT

The meninges are densely innervated by nociceptive sensory neurons that mediate pain and headache1,2. Bacterial meningitis causes life-threatening infections of the meninges and central nervous system, affecting more than 2.5 million people a year3-5. How pain and neuroimmune interactions impact meningeal antibacterial host defences are unclear. Here we show that Nav1.8+ nociceptors signal to immune cells in the meninges through the neuropeptide calcitonin gene-related peptide (CGRP) during infection. This neuroimmune axis inhibits host defences and exacerbates bacterial meningitis. Nociceptor neuron ablation reduced meningeal and brain invasion by two bacterial pathogens: Streptococcus pneumoniae and Streptococcus agalactiae. S. pneumoniae activated nociceptors through its pore-forming toxin pneumolysin to release CGRP from nerve terminals. CGRP acted through receptor activity modifying protein 1 (RAMP1) on meningeal macrophages to polarize their transcriptional responses, suppressing macrophage chemokine expression, neutrophil recruitment and dural antimicrobial defences. Macrophage-specific RAMP1 deficiency or pharmacological blockade of RAMP1 enhanced immune responses and bacterial clearance in the meninges and brain. Therefore, bacteria hijack CGRP-RAMP1 signalling in meningeal macrophages to facilitate brain invasion. Targeting this neuroimmune axis in the meninges can enhance host defences and potentially produce treatments for bacterial meningitis.


Subject(s)
Brain , Meninges , Meningitis, Bacterial , Neuroimmunomodulation , Humans , Brain/immunology , Brain/microbiology , Calcitonin Gene-Related Peptide/metabolism , Meninges/immunology , Meninges/microbiology , Meninges/physiopathology , Pain/etiology , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Meningitis, Bacterial/complications , Meningitis, Bacterial/immunology , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/pathology , Streptococcus agalactiae/immunology , Streptococcus agalactiae/pathogenicity , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity , Nociceptors/metabolism , Receptor Activity-Modifying Protein 1/metabolism , Macrophages/immunology , Macrophages/metabolism
15.
Mol Pain ; 19: 17448069231170072, 2023.
Article in English | MEDLINE | ID: mdl-37002193

ABSTRACT

BACKGROUND: Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder, and its specific pathogenesis is still unclear. We have previously reported that TTX-resistant (TTX-R) sodium channels in colon-specific dorsal root ganglion (DRG) neurons were sensitized in a rat model of visceral hypersensitivity induced by neonatal colonic inflammation (NCI). However, the detailed molecular mechanism for activation of sodium channels remains unknown. This study was designed to examine roles for melatonin (MT) in sensitization of sodium channels in NCI rats. METHODS: Colorectal distention (CRD) in adult male rats as a measure of visceral hypersensitivity. Colon-specific dorsal root ganglion (DRG) neurons were labeled with DiI and acutely dissociated for measuring excitability and sodium channel current under whole-cell patch clamp configurations. Western blot and Immunofluorescence were employed to detect changes in expression of Nav1.8 and MT2. RESULTS: The results showed that rats exhibited visceral hypersensitivity after NCI treatment. Intrathecal application of melatonin significantly increased the threshold of CRD in NCI rats with a dose-dependent manner, but has no role in the control group. Whole-cell patch clamp recording showed that melatonin remarkably decreased the excitability and the density of TTX-R sodium channel in DRG neurons from NCI rats. The expression of MT2 receptor at the protein level was markedly lower in NCI rats. 8MP, an agonist of MT2 receptor, enhanced the distention threshold in NCI rats. Application of 8MP reversed the enhanced hypersensitivity of DRG neurons from NCI rats. 8MP also reduced TTX-R sodium current density and modulated dynamics of TTX-R sodium current activation. CONCLUSIONS: These data suggest that sensitization of sodium channels of colon DRG neurons in NCI rats is most likely mediated by MT2 receptor, thus identifying a potential target for treatment for chronic visceral pain in patients with IBS.


Subject(s)
Irritable Bowel Syndrome , Melatonin , Visceral Pain , Rats , Animals , Male , Irritable Bowel Syndrome/complications , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/metabolism , Rats, Sprague-Dawley , Melatonin/pharmacology , Melatonin/therapeutic use , Melatonin/metabolism , Visceral Pain/metabolism , Nociception , Receptor, Melatonin, MT2/metabolism , Ganglia, Spinal/metabolism , Tetrodotoxin , NAV1.8 Voltage-Gated Sodium Channel/metabolism
16.
Mol Psychiatry ; 28(1): 76-82, 2023 01.
Article in English | MEDLINE | ID: mdl-36224259

ABSTRACT

Pitt Hopkins Syndrome (PTHS) is a rare syndromic form of autism spectrum disorder (ASD) caused by autosomal dominant mutations in the Transcription Factor 4 (TCF4) gene. TCF4 is a basic helix-loop-helix transcription factor that is critical for neurodevelopment and brain function through its binding to cis-regulatory elements of target genes. One potential therapeutic strategy for PTHS is to identify dysregulated target genes and normalize their dysfunction. Here, we propose that SCN10A is an important target gene of TCF4 that is an applicable therapeutic approach for PTHS. Scn10a encodes the voltage-gated sodium channel Nav1.8 and is consistently shown to be upregulated in PTHS mouse models. In this perspective, we review prior literature and present novel data that suggests inhibiting Nav1.8 in PTHS mouse models is effective at normalizing neuron function, brain circuit activity and behavioral abnormalities and posit this therapeutic approach as a treatment for PTHS.


Subject(s)
Intellectual Disability , NAV1.8 Voltage-Gated Sodium Channel , Animals , Mice , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Facies , Hyperventilation/genetics , Intellectual Disability/drug therapy , Intellectual Disability/genetics , Intellectual Disability/metabolism , Transcription Factor 4/genetics , NAV1.8 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/metabolism
17.
Mol Pain ; 19: 17448069221150138, 2023.
Article in English | MEDLINE | ID: mdl-36550597

ABSTRACT

Polysorbate 80 is a non-ionic detergent derived from polyethoxylated sorbitan and oleic acid. It is widely used in pharmaceuticals, foods, and cosmetics as an emulsifier. Nav1.7 is a peripheral sodium channel that is highly expressed in sympathetic and sensory neurons, and it plays a critical role in determining the threshold of action potentials (APs). We found that 10 µg/mL polysorbate 80 either abolished APs or increased the threshold of the APs of dorsal root ganglions. We thus investigated whether polysorbate 80 inhibits Nav1.7 sodium current using a whole-cell patch-clamp recording technique. Polysorbate 80 decreased the Nav1.7 current in a concentration-dependent manner with a half-maximal inhibitory concentration (IC50) of 250.4 µg/mL at a holding potential of -120 mV. However, the IC50 was 1.1 µg/mL at a holding potential of -90 mV and was estimated to be 0.9 µg/mL at the resting potentials of neurons, where most channels are inactivated. The activation rate and the voltage dependency of activation of Nav1.7 were not changed by polysorbate 80. However, polysorbate 80 caused hyperpolarizing shifts in the voltage dependency of the steady-state fast inactivation curve. The blocking of Nav1.7 currents by polysorbate 80 was not reversible at a holding potential of -90 mV but was completely reversible at -120 mV, where the channels were mostly in the closed state. Polysorbate 80 also slowed recovery from inactivation and induced robust use-dependent inhibition, indicating that it is likely to bind to and stabilize the inactivated state. Our results indicate that polysorbate 80 inhibits Nav1.7 current in concentration-, state-, and use-dependent manners when used even below commercial concentrations. This suggests that polysorbate 80 may be helpful in pain medicine as an excipient. In addition, in vitro experiments using polysorbate 80 with neurons should be conducted with caution.


Subject(s)
Neurons , Polysorbates , Polysorbates/pharmacology , Polysorbates/metabolism , Neurons/metabolism , Sodium Channels/metabolism , Membrane Potentials/physiology , Action Potentials , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism
18.
eNeuro ; 9(2)2022.
Article in English | MEDLINE | ID: mdl-35131865

ABSTRACT

The antidiabetic drug metformin has been shown to reduce pain hypersensitivity in preclinical models of chronic pain and in neuropathic pain in humans. Multiple intracellular pathways have been described as metformin targets. Among them, metformin is an activator of the adenosine 5'-monophosphate protein kinase that can in turn modulate the activity of the E3 ubiquitin ligase NEDD4-2 and thus post-translational expression of voltage-gated sodium channels (NaVs). In this study, we found that the bulk of the effect of metformin on Na1.7 is dependent on NEDD4-2. In HEK cells, the expression of NaV1.7 at the membrane fraction, obtained by a biotinylation approach, is only reduced by metformin when cotransfected with NEDD4-2. Similarly, in voltage-clamp recordings, metformin significantly reduced NaV1.7 current density when cotransfected with NEDD4-2. In mouse dorsal root ganglion (DRG) neurons, without changing the biophysical properties of NaV1.7, metformin significantly decreased NaV1.7 current densities, but not in Nedd4L knock-out mice (SNS-Nedd4L-/-). In addition, metformin induced a significant reduction in NEDD4-2 phosphorylation at the serine-328 residue in DRG neurons, an inhibitory phosphorylation site of NEDD4-2. In current-clamp recordings, metformin reduced the number of action potentials elicited by DRG neurons from Nedd4Lfl/fl , with a partial decrease also present in SNS-Nedd4L-/- mice, suggesting that metformin can also change neuronal excitability in an NEDD4-2-independent manner. We suggest that NEDD4-2 is a critical player for the effect of metformin on the excitability of nociceptive neurons; this action may contribute to the relief of neuropathic pain.


Subject(s)
Metformin , Voltage-Gated Sodium Channels , Animals , Ganglia, Spinal/metabolism , Hypoglycemic Agents/pharmacology , Metformin/metabolism , Metformin/pharmacology , Mice , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Nedd4 Ubiquitin Protein Ligases/metabolism , Ubiquitin/metabolism , Ubiquitin/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Voltage-Gated Sodium Channels/metabolism
19.
Clin Exp Pharmacol Physiol ; 49(3): 350-359, 2022 03.
Article in English | MEDLINE | ID: mdl-34750860

ABSTRACT

It has been repeatedly proved that Nav1.8 tetrodotoxin (TTX)-resistant sodium currents are expressed in peripheral sensory neurons where they play important role in nociception. There are very few publications that show the presence of TTX-resistant sodium currents in central neurons. The aim of this study was to assess if functional Nav1.8 TTX-resistant sodium currents are expressed in prefrontal cortex pyramidal neurons. All recordings were performed in the presence of TTX in the extracellular solution to block TTX-sensitive sodium currents. The TTX-resistant sodium current recorded in this study was mainly carried by the Nav1.8 sodium channel isoform because the Nav1.9 current was inhibited by the -65 mV holding potential that we used throughout the study. Moreover, the sodium current that we recorded was inhibited by treatment with the selective Nav1.8 inhibitor A-803467. Confocal microscopy experiments confirmed the presence of the Nav1.8 α subunit in prefrontal cortex pyramidal neurons. Activation and steady state inactivation properties of TTX-resistant sodium currents were also assessed in this study and they were similar to activation and inactivation properties of TTX-resistant sodium currents expressed in dorsal root ganglia (DRG) neurons. Moreover, this study showed that carbamazepine (60 µM) inhibited the maximal amplitude of the TTX-resistant sodium current. Furthermore, we found that carbamazepine shifts steady state inactivation curve of TTX-resistant sodium currents toward hyperpolarization. This study suggests that the Nav1.8 TTX-resistant sodium channel is expressed not only in DRG neurons, but also in cortical neurons and may be molecular target for antiepileptic drugs such as carbamazepine.


Subject(s)
Gene Expression Regulation/drug effects , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Prefrontal Cortex/cytology , Pyramidal Cells/physiology , Sodium/metabolism , Tetrodotoxin/pharmacology , Action Potentials/drug effects , Animals , Anticonvulsants/pharmacology , Carbamazepine/pharmacology , Ion Channel Gating/drug effects , NAV1.8 Voltage-Gated Sodium Channel/genetics , Rats , Rats, Wistar
20.
Nat Commun ; 12(1): 6586, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34782600

ABSTRACT

An interplay between Ca2+/calmodulin-dependent protein kinase IIδc (CaMKIIδc) and late Na+ current (INaL) is known to induce arrhythmias in the failing heart. Here, we elucidate the role of the sodium channel isoform NaV1.8 for CaMKIIδc-dependent proarrhythmia. In a CRISPR-Cas9-generated human iPSC-cardiomyocyte homozygous knock-out of NaV1.8, we demonstrate that NaV1.8 contributes to INaL formation. In addition, we reveal a direct interaction between NaV1.8 and CaMKIIδc in cardiomyocytes isolated from patients with heart failure (HF). Using specific blockers of NaV1.8 and CaMKIIδc, we show that NaV1.8-driven INaL is CaMKIIδc-dependent and that NaV1.8-inhibtion reduces diastolic SR-Ca2+ leak in human failing cardiomyocytes. Moreover, increased mortality of CaMKIIδc-overexpressing HF mice is reduced when a NaV1.8 knock-out is introduced. Cellular and in vivo experiments reveal reduced ventricular arrhythmias without changes in HF progression. Our work therefore identifies a proarrhythmic CaMKIIδc downstream target which may constitute a prognostic and antiarrhythmic strategy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/metabolism , Homeostasis/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Animals , Arrhythmias, Cardiac/metabolism , CRISPR-Cas Systems , Heart Failure/pathology , Humans , Mice , Mice, Knockout , Mice, Transgenic , Molecular Medicine , Myocytes, Cardiac
SELECTION OF CITATIONS
SEARCH DETAIL