260 results on '"Nav1.9"'
Search Results
2. Small Fiber Neuropathy in Burning Mouth Syndrome: A Systematic Review.
- Author
-
Kouri, Maria, Adamo, Daniela, Vardas, Emmanouil, Georgaki, Maria, Canfora, Federica, Mignogna, Michele Davide, and Nikitakis, Nikolaos
- Subjects
- *
BURNING mouth syndrome , *PAIN threshold , *OROFACIAL pain , *NERVE fibers , *GENE expression , *BLINKING (Physiology) - Abstract
Burning mouth syndrome (BMS) is a chronic idiopathic orofacial pain disorder, characterized by persistent burning sensations and pain without clear pathological causes. Recent research suggests that small fiber neuropathy (SFN) may play a significant role in the neuropathic pain and sensory disturbances associated with BMS. Following PRISMA guidelines, this systematic review aims to evaluate and synthesize current evidence supporting SFN's involvement in BMS. The protocol is registered in PROSPERO (CRD42024555839). The results show eight studies reported reductions in nerve fiber density in tongue biopsies (ranging from 30% to 60%), along with morphological changes indicative of small fiber damage. Additionally, an increase in TRPV1-positive, NGF-positive, and P2X3-positive fibers, overexpression of Nav1.7, and slight underexpression of Nav1.9 mRNA were observed in BMS patients. Quantitative Sensory Testing in seven studies revealed sensory abnormalities such as reduced cool detection and cold pain thresholds. Blink reflex and corneal confocal microscopy also indicated peripheral and central small fiber damage, along with increased artemin mRNA expression. The evidence strongly supports SFN as a key factor in the pathophysiology of BMS, particularly due to reductions in nerve fiber density and altered sensory thresholds. However, variability across studies highlights the need for larger, standardized research to establish causal relationships and guide therapeutic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Familial Episodic Pain Syndrome: A Japanese Family Harboring the Novel Variant c.2431C>T (p.Leu811Phe) in SCN11A
- Author
-
Nagao, Chioko, Okuda, Hiroko, Bekker, Gert-Jan, Noguchi, Atsuko, Takahashi, Tsutomu, Koizumi, Akio, Youssefian, Shohab, Tezuka, Tohru, and Akioka, Shinji
- Published
- 2024
- Full Text
- View/download PDF
4. Visceral Nociception in Gastrointestinal Disease
- Author
-
Higham, James, Gupta, Rohit, Bulmer, David C., Brierley, Stuart M., editor, and Spencer, Nick J., editor
- Published
- 2023
- Full Text
- View/download PDF
5. Inhibition of the Human Neuronal Sodium Channel Nav1.9 by Arachidonyl-2-Chloroethylamide, An Analogue of Anandamide in a hNav1.9/rNav1.4 Chimera, An Experimental and in Silico Study.
- Author
-
Marchese-Rojas, Mario, Islas, Ángel A., Mancilla-Simbro, Claudia, Millan-PerezPeña, Lourdes, León, Jorge S., and Salinas-Stefanon, Eduardo M.
- Subjects
- *
SODIUM channels , *ANANDAMIDE , *MONTE Carlo method , *ANESTHETICS , *BINDING sites , *SYNTHETIC marijuana , *NEURALGIA - Abstract
• hNa v 1.9_C4 chimera in CHOK-1 cells encompasses the extracellular and transmembrane domain of hNa v 1.9. • Synthetic cannabinoid ACEA blocked hNa v 1.9_C4+ß1 currents causing a hyperpolarizing shift in inactivation. • hNa v 1.9_C4 channels display window currents, significantly reduced by ACEA. • Computationally, ACEA binds the local anaesthetic binding site. • The local anaesthetic profile of ACEA may be involved in cannabinoid analgesia. Cannabinoids regulate analgesia, which has aroused much interest in identifying new pharmacological therapies in the management of refractory pain. Voltage-gated Na+ channels (Na v s) play an important role in inflammatory and neuropathic pain. In particular, Na v 1.9 is involved in nociception and the understanding of its pharmacology has lagged behind because it is difficult to express in heterologous systems. Here, we utilized the chimeric channel hNa v 1.9_C4, that comprises the extracellular and transmembrane domains of hNa v 1.9, co-expressed with the ß1 subunit on CHO-K1 cells to characterize the electrophysiological effects of ACEA, a synthetic surrogate of the endogenous cannabinoid anandamide. ACEA induced a tonic block, decelerated the fast inactivation, markedly shifted steady-state inactivation in the hyperpolarized direction, decreasing the window current and showed use-dependent block, with a high affinity for the inactivated state (k i = 0.84 µM). Thus, we argue that ACEA possess a local anaesthetic-like profile. To provide a mechanistic understanding of its mode of action at the molecular level, we combined induced fit docking with Monte Carlo simulations and electrostatic complementarity. In agreement with the experimental evidence, our computer simulations revealed that ACEA binds Tyr1599 of the local anaesthetics binding site of the hNa v 1.9, contacting residues that bind cannabinol (CBD) in the Na v Ms channel. ACEA adopted a conformation remarkably similar to the crystallographic conformation of anandamide on a non-homologous protein, obstructing the Na+ permeation pathway below the selectivity filter to occupy a highly conserved binding pocket at the intracellular side. These results describe a mechanism of action, possibly involved in cannabinoid analgesia. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Isolation and transfection of myenteric neurons from mice for patch-clamp applications.
- Author
-
Kuehs, Samuel, Teege, Laura, Hellberg, Ann-Katrin, Stanke, Christina, Haag, Natja, Kurth, Ingo, Blum, Robert, Nau, Carla, and Leipold, Enrico
- Subjects
ENTERIC nervous system ,SUBMUCOUS plexus ,NEURAL circuitry ,CENTRAL nervous system ,NEURONS ,SODIUM channels - Abstract
The enteric nervous system (ENS) is a complex neuronal network organized in ganglionated plexuses that extend along the entire length of the gastrointestinal tract. Largely independent of the central nervous system, the ENS coordinates motility and peristalsis of the digestive tract, regulates secretion and absorption, and is involved in immunological processes. Electrophysiological methods such as the patch-clamp technique are particularly suitable to study the function of neurons as well as the biophysical parameters of the underlying ion channels under both physiological and pathophysiological conditions. However, application of the patch-clamp method to ENS neurons remained difficult because they are embedded in substantial tissue layers that limit access to and targeted manipulation of these cells. Here, we present a robust step-by-step protocol that involves isolation of ENS neurons from adult mice, culturing of the cells, their transfection with plasmid DNA, and subsequent electrophysiological characterization of individual neurons in current-clamp and voltage-clamp recordings. With this protocol, ENS neurons can be prepared, transfected, and electrophysiologically characterized within 72 h. Using isolated ENS neurons, we demonstrate the feasibility of the approach by functional overexpression of recombinant voltage-gated NaV1.9 mutant channels associated with hereditary sensory and autonomic neuropathy type 7 (HSAN-7), a disorder characterized by congenital analgesia and severe constipation that can require parenteral nutrition. Although our focus is on the electrophysiological evaluation of isolated ENS neurons, the presented methodology is also useful to analyze molecules other than sodium channels or to apply alternative downstream assays including calcium imaging, proteomic and nucleic acid approaches, or immunochemistry. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Isolation and transfection of myenteric neurons from mice for patch-clamp applications
- Author
-
Samuel Kuehs, Laura Teege, Ann-Katrin Hellberg, Christina Stanke, Natja Haag, Ingo Kurth, Robert Blum, Carla Nau, and Enrico Leipold
- Subjects
single cell isolation ,sodium channels ,NaV1.9 ,patch-clamp ,electrophysiology ,myenteric neurons ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The enteric nervous system (ENS) is a complex neuronal network organized in ganglionated plexuses that extend along the entire length of the gastrointestinal tract. Largely independent of the central nervous system, the ENS coordinates motility and peristalsis of the digestive tract, regulates secretion and absorption, and is involved in immunological processes. Electrophysiological methods such as the patch-clamp technique are particularly suitable to study the function of neurons as well as the biophysical parameters of the underlying ion channels under both physiological and pathophysiological conditions. However, application of the patch-clamp method to ENS neurons remained difficult because they are embedded in substantial tissue layers that limit access to and targeted manipulation of these cells. Here, we present a robust step-by-step protocol that involves isolation of ENS neurons from adult mice, culturing of the cells, their transfection with plasmid DNA, and subsequent electrophysiological characterization of individual neurons in current-clamp and voltage-clamp recordings. With this protocol, ENS neurons can be prepared, transfected, and electrophysiologically characterized within 72 h. Using isolated ENS neurons, we demonstrate the feasibility of the approach by functional overexpression of recombinant voltage-gated NaV1.9 mutant channels associated with hereditary sensory and autonomic neuropathy type 7 (HSAN-7), a disorder characterized by congenital analgesia and severe constipation that can require parenteral nutrition. Although our focus is on the electrophysiological evaluation of isolated ENS neurons, the presented methodology is also useful to analyze molecules other than sodium channels or to apply alternative downstream assays including calcium imaging, proteomic and nucleic acid approaches, or immunochemistry.
- Published
- 2022
- Full Text
- View/download PDF
8. Pathological changes of the sural nerve in patients with familial episodic pain syndrome.
- Author
-
Zheng, Yilei, Huang, Pengcheng, Li, Shumeng, Jiang, Kaiyan, Zhou, Binbin, Fang, Xin, Zhou, Meihong, Hong, Daojun, and Zhu, Min
- Abstract
Background: Familial episodic pain syndrome type 3 (FEPS3) is an inherited disorder characterized by the early-childhood onset of severe episodic pain that primarily affects the distal extremities. As skin biopsy has revealed a reduction in intraepidermal nerve fiber density and degeneration of the unmyelinated axons, it remains unclear whether FEPS3 patients have pathological changes in the peripheral nerve. Methods: The clinical features of patients with FEPS3 were summarized in a large autosomal dominant family. Sural nerve biopsies were conducted in two patients. Whole exome sequencing (WES) was performed in the index patient. Sanger sequencing was used to analyze family co-segregation. Results: Fourteen members exhibited typical and uniform clinical phenotypes characterized by length-dependent and age-dependent severe episodic pain affecting the distal extremities, which can be relieved with anti-inflammatory medicine. The WES revealed a heterozygous mutation c.665G > A (p.R222H) in the SCN11A gene, which was co-segregated with the clinical phenotype in this family. A sural biopsy in patient V:1, who was experiencing episodic pain at 16 years old, showed normal structure, while the sural nerve in patient IV:1, whose pain attack had completely diminished at 42 years old, displayed a decrease of the density of unmyelinated axons with the axonal degeneration. Conclusions: The clinical phenotype of FEPS3 showed distinctive characteristics that likely arise from dysfunctional nociceptive neurons that lack detectable pathological alterations in the nerve fibers. Nevertheless, long-term dysfunction of the Nav1.9 channel may cause degeneration of the unmyelinated fibers in FEPS3 patient with pain remission. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. Scorpion Neurotoxin Syb-prII-1 Exerts Analgesic Effect through Nav1.8 Channel and MAPKs Pathway.
- Author
-
Bai, Fei, Song, Yongbo, Cao, Yi, Ban, Mengqi, Zhang, Zhenyu, Sun, Yang, Feng, Yuan, and Li, Chunli
- Subjects
- *
MITOGEN-activated protein kinases , *CONOTOXINS , *NEUROTOXIC agents , *SCORPIONS , *TRIGEMINAL neuralgia , *SCORPION venom , *SODIUM channels - Abstract
Trigeminal neuralgia (TN) is a common type of peripheral neuralgia in clinical practice, which is usually difficult to cure. Common analgesic drugs are difficult for achieving the desired analgesic effect. Syb-prII-1 is a β-type scorpion neurotoxin isolated from the scorpion venom of Buthus martensi Karsch (BmK). It has an important influence on the voltage-gated sodium channel (VGSCs), especially closely related to Nav1.8 and Nav1.9. To explore whether Syb-prII-1 has a good analgesic effect on TN, we established the Sprague Dawley (SD) rats' chronic constriction injury of the infraorbital nerve (IoN-CCI) model. Behavioral, electrophysiological, Western blot, and other methods were used to verify the model. It was found that Syb-prII-1 could significantly relieve the pain behavior of IoN-CCI rats. After Syb-prII-1 was given, the phosphorylation level of the mitogen-activated protein kinases (MAPKs) pathway showed a dose-dependent decrease after IoN-CCI injury. Moreover, Syb-prII-1(4.0 mg/kg) could significantly change the steady-state activation and inactivation curves of Nav1.8. The steady-state activation and inactivation curves of Nav1.9 were similar to those of Nav1.8, but there was no significant difference. It was speculated that it might play an auxiliary role. The binding mode, critical residues, and specific interaction type of Syb-prII-1 and VSD2rNav1.8 were clarified with computational simulation methods. Our results indicated that Syb-prII-1 could provide a potential treatment for TN by acting on the Nav1.8 target. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. Familial episodic limb pain in kindreds with novel Nav1.9 mutations
- Author
-
Kabata, Risako and Kabata, Risako
- Published
- 2024
11. Protein arginine methyltransferase 7 modulates neuronal excitability by interacting with NaV1.9.
- Author
-
Ma, Tingbin, Li, Lulu, Chen, Rui, Yang, Luyao, Sun, Hao, Du, Shiyue, Xu, Xuan, Cao, Zhijian, Zhang, Xianwei, Zhang, Luoying, Shi, Xiaoliu, and Liu, Jing Yu
- Subjects
- *
PROTEIN arginine methyltransferases , *DORSAL root ganglia , *ACTION potentials , *CD4 antigen , *METHYLTRANSFERASES , *PAIN management , *RESEARCH , *PAIN , *NEURONS , *ANIMAL experimentation , *RESEARCH methodology , *SENSORY ganglia , *EVALUATION research , *COMPARATIVE studies , *TRANSFERASES , *MEMBRANE transport proteins , *MICE - Abstract
Abstract: Human NaV1.9 (hNaV1.9), encoded by SCN11A, is preferentially expressed in nociceptors, and its mutations have been linked to pain disorders. NaV1.9 could be a promising drug target for pain relief. However, the modulation of NaV1.9 activity has remained elusive. Here, we identified a new candidate NaV1.9-interacting partner, protein arginine methyltransferase 7 (PRMT7). Whole-cell voltage-clamp recordings showed that coelectroporation of human SCN11A and PRMT7 in dorsal root ganglion (DRG) neurons of Scn11a-/- mice increased the hNaV1.9 current density. By contrast, a PRMT7 inhibitor (DS-437) reduced mNaV1.9 currents in Scn11a+/+ mice. Using the reporter molecule CD4, we observed an increased distribution of hLoop1 on the cell surface of PRMT7-overexpressing HKE293T cells. Furthermore, we found that PRMT7 mainly binds to residues 563 to 566 within the first intracellular loop of hNaV1.9 (hLoop1) and methylates hLoop1 at arginine residue 519. Moreover, overexpression of PRMT7 increased the number of action potential fired in DRG neurons of Scn11a+/+ mice but not Scn11a-/- mice. However, DS-437 significantly inhibited the action potential frequency of DRG neurons and relieved pain hypersensitivity in Scn11aA796G/A796G mice. In summary, our observations revealed that PRMT7 modulates neuronal excitability by regulating NaV1.9 currents, which may provide a potential method for pain treatment. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
12. A Novel Spider Toxin Inhibits Fast Inactivation of the Nav1.9 Channel by Binding to Domain III and Domain IV Voltage Sensors.
- Author
-
Peng, Shuijiao, Chen, Minzhi, Xiao, Zhen, Xiao, Xin, Luo, Sen, Liang, Songping, Zhou, Xi, and Liu, Zhonghua
- Subjects
SODIUM channels ,TOXINS ,SPIDER venom ,SITE-specific mutagenesis ,SPIDERS ,VOLTAGE - Abstract
Venomous animals have evolved to produce peptide toxins that modulate the activity of voltage-gated sodium (Na
v ) channels. These specific modulators are powerful probes for investigating the structural and functional features of Nav channels. Here, we report the isolation and characterization of δ-theraphotoxin-Gr4b (Gr4b), a novel peptide toxin from the venom of the spider Grammostola rosea. Gr4b contains 37-amino acid residues with six cysteines forming three disulfide bonds. Patch-clamp analysis confirmed that Gr4b markedly slows the fast inactivation of Nav 1.9 and inhibits the currents of Nav 1.4 and Nav 1.7, but does not affect Nav 1.8. It was also found that Gr4b significantly shifts the steady-state activation and inactivation curves of Nav 1.9 to the depolarization direction and increases the window current, which is consistent with the change in the ramp current. Furthermore, analysis of Nav 1.9/Nav 1.8 chimeric channels revealed that Gr4b preferentially binds to the voltage-sensor of domain III (DIII VSD) and has additional interactions with the DIV VSD. The site-directed mutagenesis analysis indicated that N1139 and L1143 in DIII S3-S4 linker participate in toxin binding. In sum, this study reports a novel spider peptide toxin that may slow the fast inactivation of Nav 1.9 by binding to the new neurotoxin receptor site-DIII VSD. Taken together, these findings provide insight into the functional role of the Nav channel DIII VSD in fast inactivation and activation. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
13. A Novel Spider Toxin Inhibits Fast Inactivation of the Nav1.9 Channel by Binding to Domain III and Domain IV Voltage Sensors
- Author
-
Shuijiao Peng, Minzhi Chen, Zhen Xiao, Xin Xiao, Sen Luo, Songping Liang, Xi Zhou, and Zhonghua Liu
- Subjects
Nav1.9 ,fast inactivation ,domain III voltage-sensor ,spider peptide toxin ,neurotoxin receptor site ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Venomous animals have evolved to produce peptide toxins that modulate the activity of voltage-gated sodium (Nav) channels. These specific modulators are powerful probes for investigating the structural and functional features of Nav channels. Here, we report the isolation and characterization of δ-theraphotoxin-Gr4b (Gr4b), a novel peptide toxin from the venom of the spider Grammostola rosea. Gr4b contains 37-amino acid residues with six cysteines forming three disulfide bonds. Patch-clamp analysis confirmed that Gr4b markedly slows the fast inactivation of Nav1.9 and inhibits the currents of Nav1.4 and Nav1.7, but does not affect Nav1.8. It was also found that Gr4b significantly shifts the steady-state activation and inactivation curves of Nav1.9 to the depolarization direction and increases the window current, which is consistent with the change in the ramp current. Furthermore, analysis of Nav1.9/Nav1.8 chimeric channels revealed that Gr4b preferentially binds to the voltage-sensor of domain III (DIII VSD) and has additional interactions with the DIV VSD. The site-directed mutagenesis analysis indicated that N1139 and L1143 in DIII S3-S4 linker participate in toxin binding. In sum, this study reports a novel spider peptide toxin that may slow the fast inactivation of Nav1.9 by binding to the new neurotoxin receptor site-DIII VSD. Taken together, these findings provide insight into the functional role of the Nav channel DIII VSD in fast inactivation and activation.
- Published
- 2021
- Full Text
- View/download PDF
14. Translational Model Systems for Complex Sodium Channel Pathophysiology in Pain
- Author
-
Schrenk-Siemens, Katrin, Rösseler, Corinna, Lampert, Angelika, Barrett, James E., Editor-in-Chief, Flockerzi, Veit, Series Editor, Frohman, Michael A., Series Editor, Geppetti, Pierangelo, Series Editor, Hofmann, Franz B., Series Editor, Michel, Martin C., Series Editor, Page, Clive P, Series Editor, Rosenthal, Walter, Series Editor, Wang, KeWei, Series Editor, and Chahine, Mohamed, editor
- Published
- 2018
- Full Text
- View/download PDF
15. Estrogen-dependent depressor response of melatonin via baroreflex afferent function and intensification of PKC-mediated Nav1.9 activation
- Author
-
Wu, Di, Zhao, Dan, Huang, Di, Sun, Xun, Li, Ke-xin, Feng, Yan, Yan, Qiu-xin, Li, Xin-yu, Cui, Chang-peng, Li, Hu-die, and Li, Bai-yan
- Published
- 2022
- Full Text
- View/download PDF
16. Mechanical allodynia triggered by cold exposure in mice with the Scn11a p.R222S mutation: a novel model of drug therapy for neuropathic pain related to NaV1.9.
- Author
-
Matsubara, Yosuke, Okuda, Hiroko, Harada, Kouji H., Youssefian, Shohab, and Koizumi, Akio
- Subjects
PAIN management ,DRUG therapy ,ALLODYNIA ,MICE ,SODIUM channels ,BRUGADA syndrome - Abstract
Mutations within the SCN11A gene which encodes the voltage-gated sodium channel Na
V 1.9 mainly expressed in small fiber sensory neurons have been associated with neuropathic disorders; however, suitable medications have not been fully investigated. To develop drug therapies against NaV 1.9-related neuropathic pain, we aimed to establish a novel model using mice carrying the Scn11a p.R222S mutation initially identified in patients with familial episodic limb pain that is characterized by paroxysmal pain induced by fatigue or bad weather conditions. We investigated the influence of cold exposure (4 °C, overnight) on the behavioral and biochemical phenotypes of Scn11a p.R222S mutant (R222S) and wild type C57BL/6N (WT) mice. We also tested the effects of acetaminophen (125, 250 mg/kg, perorally, p.o.) and traditional Japanese medicine, goshajinkigan (0.5 or 1.0 g/kg, p.o.), which are analgesic drugs prescribed to patients with neuropathic pain, in this model of cold-induced mechanical allodynia in R222S mice. Cold-exposed R222S mice exhibited enhanced mechanical allodynia and thermal hypersensitivity compared with WT mice. The decrease of the mechanical withdrawal threshold in R222S mice was reversible 24 h after housing at room temperature. There was no significant change in the levels of interleukin-1β, interleukin-6, tumor necrosis factor-α, or interferon-γ in the plasma or spinal cords of WT and R222S mice after cold exposure. Both acetaminophen (250 mg/kg) and goshajinkigan (1.0 g/kg) significantly attenuated mechanical allodynia in R222S mice. The model of cold-induced mechanical allodynia in mice with the Scn11a p.R222S mutation is novel and useful for evaluating analgesic drugs for intractable neuropathies related to NaV 1.9. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
17. Painful and painless mutations of SCN9A and SCN11A voltage-gated sodium channels.
- Author
-
Baker, Mark D. and Nassar, Mohammed A.
- Subjects
- *
SODIUM channels , *GAIN-of-function mutations , *DORSAL root ganglia , *CENTRAL nervous system , *MYOCARDIUM , *SENSORY neurons , *CHRONIC pain - Abstract
Chronic pain is a global problem affecting up to 20% of the world's population and has a significant economic, social and personal cost to society. Sensory neurons of the dorsal root ganglia (DRG) detect noxious stimuli and transmit this sensory information to regions of the central nervous system (CNS) where activity is perceived as pain. DRG neurons express multiple voltage-gated sodium channels that underlie their excitability. Research over the last 20 years has provided valuable insights into the critical roles that two channels, NaV1.7 and NaV1.9, play in pain signalling in man. Gain of function mutations in NaV1.7 cause painful conditions while loss of function mutations cause complete insensitivity to pain. Only gain of function mutations have been reported for NaV1.9. However, while most NaV1.9 mutations lead to painful conditions, a few are reported to cause insensitivity to pain. The critical roles these channels play in pain along with their low expression in the CNS and heart muscle suggest they are valid targets for novel analgesic drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
18. N58A Exerts Analgesic Effect on Trigeminal Neuralgia by Regulating the MAPK Pathway and Tetrodotoxin-Resistant Sodium Channel
- Author
-
Chun-Li Li, Ran Yang, Yang Sun, Yuan Feng, and Yong-Bo Song
- Subjects
trigeminal neuralgia ,N58A ,Nav1.8 ,Nav1.9 ,MAPK ,analgesic effect ,Medicine - 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.
- Published
- 2021
- Full Text
- View/download PDF
19. SCN11A mRNA levels in female bipolar disorder PBMCs as tentative biomarker for distinct patient sub‐phenotypes.
- Author
-
Voinsky, Irena, McCarthy, Michael J., Shekhtman, Tatyana, Kelsoe, John R., and Gurwitz, David
- Subjects
- *
BIPOLAR disorder , *SODIUM channels , *MESSENGER RNA , *PREFRONTAL cortex , *NEUROBEHAVIORAL disorders , *SELF-poisoning - Abstract
Bipolar disorder (BD) is a complex neuropsychiatric disorder characterized by recurrent mania and depression episodes and requiring lifelong treatment with mood stabilizing drugs. Several lines of evidence, including with BD patient iPSC‐derived neurons, suggest that neuronal hyperexcitability may underlie the key clinical symptoms of BD. Indeed, higher mRNA levels of SCN11A, coding for the voltage‐gated sodium channel NaV1.9 implicated in nociception, were detected in iPSC‐derived neurons from BD patients, and were normalized by in vitro lithium. Here we studied SCN11A expression in peripheral blood mononuclear cells (PBMCs) from well‐phenotyped female BD patients and controls and evaluated their association with several clinical sub‐phenotypes. We observed higher mRNA levels of SCN11A in PBMCs from female BD patients with no records of alcohol dependence (p =.0050), no records of psychosis (p =.0097), or no records of suicide attempts (p =.0409). A trend was observed for higher SCN11A expression (FD = 1.91; p =.052) in BD PBMCs compared with controls. Datamining of published postmortem gene expression datasets indicated higher SCN11A expression in dorsolateral prefrontal cortex and orbitofrontal cortex tissues from BD patients compared with controls. Higher phenotype‐associated expression levels in PBMC from BD patients were also observed for ID2 (alcohol dependence, suicide attempts) and HDGFRP3 (seasonal BD pattern). Our findings suggest that higher PBMC SCN11A expression levels may be associated with certain behavioral BD sub‐phenotypes, including lack of alcohol dependence and psychosis, among BD patients. The NaV1.9 voltage‐gated sodium channel thus deserves consideration as a tentative phenotype modifier in BD. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
20. Characterization of Synthetic Tf2 as a NaV1.3 Selective Pharmacological Probe
- Author
-
Mathilde R. Israel, Thomas S. Dash, Stefanie N. Bothe, Samuel D. Robinson, Jennifer R. Deuis, David J. Craik, Angelika Lampert, Irina Vetter, and Thomas Durek
- Subjects
Tf2 ,sodium channel ,NaV1.3 ,NaV1.9 ,scorpion ,toxin ,Biology (General) ,QH301-705.5 - Abstract
NaV1.3 is a subtype of the voltage-gated sodium channel family. It has been implicated in the pathogenesis of neuropathic pain, although the contribution of this channel to neuronal excitability is not well understood. Tf2, a β-scorpion toxin previously identified from the venom of Tityus fasciolatus, has been reported to selectively activate NaV1.3. Here, we describe the activity of synthetic Tf2 and assess its suitability as a pharmacological probe for NaV1.3. As described for the native toxin, synthetic Tf2 (1 µM) caused early channel opening, decreased the peak current, and shifted the voltage dependence of NaV1.3 activation in the hyperpolarizing direction by −11.3 mV, with no activity at NaV1.1, NaV1.2, and NaV1.4-NaV1.8. Additional activity was found at NaV1.9, tested using the hNav1.9_C4 chimera, where Tf2 (1 µM) shifted the voltage dependence of activation by −6.3 mV. In an attempt to convert Tf2 into an NaV1.3 inhibitor, we synthetized the analogue Tf2[S14R], a mutation previously described to remove the excitatory activity of related β-scorpion toxins. Indeed, Tf2[S14R](10 µM) had reduced excitatory activity at NaV1.3, although it still caused a small −5.8 mV shift in the voltage dependence of activation. Intraplantar injection of Tf2 (1 µM) in mice caused spontaneous flinching and swelling, which was not reduced by the NaV1.1/1.3 inhibitor ICA-121431 nor in NaV1.9-/- mice, suggesting off-target activity. In addition, despite a loss of excitatory activity, intraplantar injection of Tf2[S14R](10 µM) still caused swelling, providing strong evidence that Tf2 has additional off-target activity at one or more non-neuronal targets. Therefore, due to activity at NaV1.9 and other yet to be identified target(s), the use of Tf2 as a selective pharmacological probe may be limited.
- Published
- 2020
- Full Text
- View/download PDF
21. Electrophysiological and Pharmacological Analyses of Nav1.9 Voltage-Gated Sodium Channel by Establishing a Heterologous Expression System
- Author
-
Xi Zhou, Zhen Xiao, Yan Xu, Yunxiao Zhang, Dongfang Tang, Xinzhou Wu, Cheng Tang, Minzhi Chen, Xiaoliu Shi, Ping Chen, Songping Liang, and Zhonghua Liu
- Subjects
electrophysiology ,pharmacology ,sodium channel ,Nav1.9 ,Nav1.9 mutants ,histamine ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Nav1. 9 voltage-gated sodium channel is preferentially expressed in peripheral nociceptive neurons. Recent progresses have proved its role in pain sensation, but our understanding of Nav1.9, in general, has lagged behind because of limitations in heterologous expression in mammal cells. In this work, functional expression of human Nav1.9 (hNav1.9) was achieved by fusing GFP to the C-terminal of hNav1.9 in ND7/23 cells, which has been proved to be a reliable method to the electrophysiological and pharmacological studies of hNav1.9. By using the hNav1.9 expression system, we investigated the electrophysiological properties of four mutations of hNav1.9 (K419N, A582T, A842P, and F1689L), whose electrophysiological functions have not been determined yet. The four mutations significantly caused positive shift of the steady-state fast inactivation and therefore increased hNav1.9 activity, consistent with the phenotype of painful peripheral neuropathy. Meanwhile, the effects of inflammatory mediators on hNav1.9 were also investigated. Impressively, histamine was found for the first time to enhance hNav1.9 activity, indicating its vital role in hNav1.9 modulating inflammatory pain. Taken together, our research provided a useful platform for hNav1.9 studies and new insight into mechanism of hNav1.9 linking to pain.
- Published
- 2017
- Full Text
- View/download PDF
22. Noxious Mechanosensation
- Author
-
Delmas, Patrick, Korogod, Sergiy Mikhailovych, Coste, Bertrand, and Wood, John N., book editor
- Published
- 2020
- Full Text
- View/download PDF
23. NaV1.9 Potentiates Oxidized Phospholipid-Induced TRP Responses Only under Inflammatory Conditions.
- Author
-
Martin, Corinna, Stoffer, Carolin, Mohammadi, Milad, Hugo, Julian, Leipold, Enrico, Oehler, Beatrice, Rittner, Heike L., and Blum, Robert
- Subjects
PHOSPHOLIPIDS ,PHOSPHOCHOLINE ,INFLAMMATORY mediators - Abstract
Oxidized phospholipids (OxPL) like oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3- phosphocholine (OxPAPC) were recently identified as novel proalgesic targets in acute and chronic inflammatory pain. These endogenous chemical irritants are generated in inflamed tissue and mediate their pain-inducing function by activating the transient receptor potential channels TRPA1 and TRPV1 expressed in sensory neurons. Notably, prototypical therapeutics interfering with OxPL were shown to inhibit TRP channel activation and pain behavior. Here, we asked how OxPL excite primary sensory neurons of dorsal root ganglia (DRG neurons from mice of either sex). Acute stimulation of sensory neurons with the prototypical OxPL 1-palmitoyl-2-glutaryl-sn-glycero-3- phosphocholine (PGPC) evoked repetitive calcium spikes in small-diameter neurons. As Na
V 1.9, a voltage-gated sodium channel involved in nociceptor excitability, was previously shown to be essential for the generation of calcium spikes in motoneurons, we asked if this channel is also important for OxPL mediated calcium spike and action potential generation in nociceptors. In wild-type and NaV 1.9-deficient neurons, the action potential firing rate and the calcium spike frequency to an acute PGPC stimulus was similar. When preincubated with inflammatory mediators, both, the action potential firing rate and the calciumspike frequency weremarkedly increased in response to an acute PGPC stimulus. However, this potentiating effect was completely lost in NaV 1.9-deficient small-diameter neurons. After treatment with inflammatory mediators, the resting membrane potential of NaV 1.9 KO neurons was slightly more negative than that of wild-type control neurons. This suggests that NaV 1.9 channels are active under this condition and therefore increases the ease with which action potentials are elicited after OxPL stimulation. In summary, our data suggest that NaV 1.9 has a switch function to potentiate the receptor potentials induced by OxPL under inflammatory conditions. Since human NaV 1.9 has been shown to mediate painful and painless channelopathies, this study provides new insights into the mechanism by which NaV 1.9 amplifies stimuli of endogenous irritants under inflammatory conditions. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
24. Understanding the physiological role of NaV1.9: Challenges and opportunities for pain modulation.
- Author
-
Brackx, Wayra, Collaço, Rita de Cássia, Theys, Margaux, Vander Cruyssen, Jolien, and Bosmans, Frank
- Subjects
- *
ITCHING , *SENSORY perception , *MEMBRANE proteins , *SENSORIMOTOR integration , *PAIN clinics , *HUMAN body - Abstract
Voltage-activated Na+ (Na V) channels are crucial contributors to rapid electrical signaling in the human body. As such, they are among the most targeted membrane proteins by clinical therapeutics and natural toxins. Several of the nine mammalian Na V channel subtypes play a documented role in pain or other sensory processes such as itch, touch, and smell. While causal relationships between these subtypes and biological function have been extensively described, the physiological role of Na V 1.9 is less understood. Yet, mutations in Na V 1.9 can cause striking disease phenotypes related to sensory perception such as loss or gain of pain and chronic itch. Here, we explore our current knowledge of the mechanisms by which Na V 1.9 may contribute to pain and elaborate on the challenges associated with establishing links between experimental conditions and human disease. This review also discusses the lack of comprehensive insights into Na V 1.9-specific pharmacology, an unfortunate situation since modulatory compounds may have tremendous potential in the clinic to treat pain or as precision tools to examine the extent of Na V 1.9 participation in sensory perception processes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. Modulations of Nav1.8 and Nav1.9 Channels in Monosodium Urate–Induced Gouty Arthritis in Mice
- Author
-
Xiu-Qi Xu, Guangqin Zhang, Shijia Zhang, Jie Qiu, and Guang Li
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Action potential ,Chemistry ,Immunology ,Depolarization ,medicine.disease ,Rheumatology ,Gout ,Nav1.9 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Endocrinology ,Dorsal root ganglion ,030220 oncology & carcinogenesis ,Internal medicine ,NAV1 ,medicine ,Immunology and Allergy ,Ankle - Abstract
The aim of the present study was to observe the changes of TTX-R, Nav1.8, and Nav1.9 Na+ currents in MSU-induced gouty arthritis mice, and to explore the possibility of Nav1.8 and Nav1.9 channels as potential targets for gout pain treatment. Acute gouty arthritis was induced by monosodium urate (MSU) in mice. Swelling degree was evaluated by measuring the circumference of the ankle joint. Mechanical allodynia was assessed by applying the electronic von Frey. Na+ currents were recorded by patch-clamp techniques in acute isolated dorsal root ganglion (DRG) neurons. MSU treatment significantly increased the swelling degree of ankle joint and decreased the mechanical pain threshold. The amplitude of TTX-R Na+ current was significantly increased and reached its peak on the 4th day after injection of MSU. For TTX-R Na+ channel subunits, Nav1.8 current density was significantly increased, but Nav1.9 current density was markedly decreased after MSU treatment. MSU treatment shifted the steady-state activation curves of TTX-R Na+ channel, Nav1.8 and Nav1.9 channels, and the inactivation curves of TTX-R Na+ channel and Nav1.8 channels to the depolarizing direction, but did not affect the inactivation curve of Nav1.9 channel. Compared with the normal group, the recovery of Nav1.8 channel was faster, while that of Nav1.9 channel was slower. The recovery of TTX-R Na+ channel remained unchanged after MSU treatment. Additionally, MSU treatment increased DRG neurons excitability by reducing action potential threshold. Nav1.8 channel, not Nav1.9 channel, may be involved in MSU-induced gout pain by increasing nerve excitability.
- Published
- 2021
- Full Text
- View/download PDF
26. Antinociceptive effects of AGAP, a recombinant neurotoxic polypeptide: Possible involvement of the tetrodotoxin-resistant sodium channels in small dorsal root ganglia neurons
- Author
-
Li Chunli, Liu Xifang, Li Guixia, Ban Mengqi, Chen Jianzhao, Cui Yong, Zhang Jinghai, and Wu Chunfu
- Subjects
Pain ,Nav1.8 ,voltage-gated sodium channels ,Scorpion toxins ,Nav1.9 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Antitumor-analgesic peptide(AGAP)is a novel recombinant polypeptide. The primary study showed that AGAP 1.0 mg/kg exhibited strong analgesic and antitumor effects. The tail vein administration of AGAP potently reduced pain behaviors in mice induced by intraplantar injection of formalin or intraperitoneal injection of acetic acid, without affecting basal pain perception. To further assess the mechanisms of AGAP, the effects of AGAP on sodium channels were assessed using the whole-cell patch clamp recordings in dorsal root ganglia (DRG) neurons. The results showed that AGAP (3-1000 nM) inhibited the sodium currents in small-diameter DRG neurons in a dose-dependent manner. 1000 nM AGAP could inhibit the current density-voltage relationship curve of sodium channels in a voltage-dependent manner and negatively shift the activation curves. 1000 nM AGAP could reduce the tetrodotoxin-resistant (TTX-R) sodium currents by 42.8% in small-diameter DRG neurons. Further analysis revealed that AGAP potently inhibited NaV1.8 currents by 59.4%, and negatively shifted the activation and inactivation kinetics. 1000 nM AGAP also reduced the NaV1.9 currents by 33.7%, but had no significant effect on activation and inactivation kinetics. Thus, our results demonstrated that submicromolar concentrations of AGAP inhibited TTX-R sodium channel in rat small-diameter DRG neurons. It is concluded that these new results may better explain, at least in part, the analgesic properties of this polypeptide.
- Published
- 2016
- Full Text
- View/download PDF
27. Painful and painless mutations of SCN9A and SCN11A voltage-gated sodium channels
- Author
-
Mohammed A. Nassar and Mark D. Baker
- Subjects
0301 basic medicine ,Central Nervous System ,Physiology ,Clinical Biochemistry ,Central nervous system ,Analgesic ,Population ,Pain ,Sensory system ,Nav1.9 ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Ganglia, Spinal ,Noxious stimulus ,medicine ,Animals ,Humans ,education ,Dorsal root ganglia ,NAV1.9 Voltage-Gated Sodium Channel ,Nav1.7 ,education.field_of_study ,Invited Review ,business.industry ,Sodium channel ,NAV1.7 Voltage-Gated Sodium Channel ,Chronic pain ,Human mutations ,medicine.disease ,Painful conditions ,030104 developmental biology ,medicine.anatomical_structure ,Mutation ,Chronic Pain ,business ,Voltage-gated sodium channels ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Chronic pain is a global problem affecting up to 20% of the world’s population and has a significant economic, social and personal cost to society. Sensory neurons of the dorsal root ganglia (DRG) detect noxious stimuli and transmit this sensory information to regions of the central nervous system (CNS) where activity is perceived as pain. DRG neurons express multiple voltage-gated sodium channels that underlie their excitability. Research over the last 20 years has provided valuable insights into the critical roles that two channels, NaV1.7 and NaV1.9, play in pain signalling in man. Gain of function mutations in NaV1.7 cause painful conditions while loss of function mutations cause complete insensitivity to pain. Only gain of function mutations have been reported for NaV1.9. However, while most NaV1.9 mutations lead to painful conditions, a few are reported to cause insensitivity to pain. The critical roles these channels play in pain along with their low expression in the CNS and heart muscle suggest they are valid targets for novel analgesic drugs.
- Published
- 2020
28. Conotoxins Targeting Neuronal Voltage-Gated Sodium Channel Subtypes: Potential Analgesics?
- Author
-
Jeffrey R. McArthur, Oliver Knapp, and David J. Adams
- Subjects
voltage-gated sodium channel ,Nav1.3 ,Nav1.7 ,Nav1.8 ,Nav1.9 ,m-conotoxin ,mO-conotoxin ,nociception ,analgesic ,pain ,Medicine - Abstract
Voltage-gated sodium channels (VGSC) are the primary mediators of electrical signal amplification and propagation in excitable cells. VGSC subtypes are diverse, with different biophysical and pharmacological properties, and varied tissue distribution. Altered VGSC expression and/or increased VGSC activity in sensory neurons is characteristic of inflammatory and neuropathic pain states. Therefore, VGSC modulators could be used in prospective analgesic compounds. VGSCs have specific binding sites for four conotoxin families: μ-, μO-, δ- and ί-conotoxins. Various studies have identified that the binding site of these peptide toxins is restricted to well-defined areas or domains. To date, only the μ- and μO-family exhibit analgesic properties in animal pain models. This review will focus on conotoxins from the μ- and μO-families that act on neuronal VGSCs. Examples of how these conotoxins target various pharmacologically important neuronal ion channels, as well as potential problems with the development of drugs from conotoxins, will be discussed.
- Published
- 2012
- Full Text
- View/download PDF
29. Animal Toxins Can Alter the Function of Nav1.8 and Nav1.9
- Author
-
John Gilchrist and Frank Bosmans
- Subjects
Nav1.8 ,Nav1.9 ,pain ,animal toxins ,voltage sensor ,voltage-activated sodium channel ,Medicine - Abstract
Human voltage-activated sodium (Nav) channels are adept at rapidly transmitting electrical signals across long distances in various excitable tissues. As such, they are amongst the most widely targeted ion channels by drugs and animal toxins. Of the nine isoforms, Nav1.8 and Nav1.9 are preferentially expressed in DRG neurons where they are thought to play an important role in pain signaling. Although the functional properties of Nav1.8 have been relatively well characterized, difficulties with expressing Nav1.9 in established heterologous systems limit our understanding of the gating properties and toxin pharmacology of this particular isoform. This review summarizes our current knowledge of the role of Nav1.8 and Nav1.9 in pain perception and elaborates on the approaches used to identify molecules capable of influencing their function.
- Published
- 2012
- Full Text
- View/download PDF
30. Author Correction: Maladaptive activation of Nav1.9 channels by nitric oxide causes triptan-induced medication overuse headache
- Author
-
Caroline Bonnet, Virginie Penalba, Anne Donnet, Jérôme Ruel, Nancy Osorio, Patrick Delmas, and Jizhe Hao
- Subjects
Male ,Science ,Calcitonin Gene-Related Peptide ,Migraine Disorders ,General Physics and Astronomy ,Pain ,Diseases ,Nitric Oxide ,General Biochemistry, Genetics and Molecular Biology ,Cell Degranulation ,Nitric oxide ,Nav1.9 ,chemistry.chemical_compound ,Mice ,Headache Disorders, Secondary ,Medicine ,Animals ,Mast Cells ,Neurons, Afferent ,Author Correction ,NAV1.9 Voltage-Gated Sodium Channel ,Migraine ,Cells, Cultured ,Prescription Drug Overuse ,Mice, Knockout ,Multidisciplinary ,business.industry ,Headache ,Nociceptors ,General Chemistry ,Tryptamines ,Mice, Inbred C57BL ,chemistry ,Hyperalgesia ,Anesthesia ,Female ,Medication overuse ,business ,Neurological disorders - Abstract
Medication-overuse headaches (MOH) occur with both over-the-counter and pain-relief medicines, including paracetamol, opioids and combination analgesics. The mechanisms that lead to MOH are still uncertain. Here, we show that abnormal activation of Nav1.9 channels by Nitric Oxide (NO) is responsible for MOH induced by triptan migraine medicine. Deletion of the Scn11a gene in MOH mice abrogates NO-mediated symptoms, including cephalic and extracephalic allodynia, photophobia and phonophobia. NO strongly activates Nav1.9 in dural afferent neurons from MOH but not normal mice. Abnormal activation of Nav1.9 triggers CGRP secretion, causing artery dilatation and degranulation of mast cells. In turn, released mast cell mediators potentiates Nav1.9 in meningeal nociceptors, exacerbating inflammation and pain signal. Analysis of signaling networks indicates that PKA is downregulated in trigeminal neurons from MOH mice, relieving its inhibitory action on NO-Nav1.9 coupling. Thus, anomalous activation of Nav1.9 channels by NO, as a result of chronic medication, promotes MOH.
- Published
- 2021
31. The role of voltage-gated sodium channels in modality-specific pain pathways.
- Author
-
Louloudis, Georgios
- Subjects
- *
SODIUM channels , *DORSAL root ganglia - Abstract
Pain is a distressing physical and emotional experience associated with actual or potential tissue injury, or an experience described in terms of such injury. The primary function of nociceptors, such as some dorsal root ganglion (DRG) neurons, is to transduce noxious sensory modalities, e.g. mechanical pressure, cold and heat, into electrical impulses and to transmit these to processing centres in the central nervous system (CNS). Modality-specific pain pathways have been identified through in vivo deletion of voltage-gated Na+ channels in mouse DRG neurons. Deletion of Nav1.8 channels has been shown to result in loss of mechanosensory and cold-induced pain, but not-heat induced pain, whereas deletion of Nav1.7 channels has been seen to abolish responses to noxious heat and mechanical stimuli. The present review constitutes an attempt to elucidate the mechanisms through which voltage-gated Na+ channels are involved in modality-specific pain pathways. It has been found that Nav1.8 and Nav1.9 channels are resistant to slow inactivation upon cooling, maintaining activity even though channels on other sensory afferents may be inactivated. Nav1.7 channel activity is reported to be coupled to substance P release into the dorsal horn of dorsal root ganglion (DRG) neurons in heat-specific pain pathways. Recent research has also offered insight into the role of Nav1.7 and Nav1.9 mutations in pain-related conditions, e.g. inherited erythromelalgia and cold-aggravated pain, respectively, as these influence kinetic parameters, such as open state probability. Therefore, voltage-gated Na+ channels appear to be playing an important role in segregating modality-specific pain pathways. The identification of markers for mechanisms implicated in the activation of these pathways could potentially pave the way towards the development of more effective analgesics. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
32. Antinociceptive Effects of AGAP, a Recombinant Neurotoxic Polypeptide: Possible Involvement of the Tetrodotoxin-Resistant Sodium Channels in Small Dorsal Root Ganglia Neurons.
- Author
-
Chun-Li Li, Xi-Fang Liu, Gui-Xia Li, Meng-qi Ban, Jian-Zhao Chen, Yong Cui, Jing-Hai Zhang, and Chun-Fu Wu
- Subjects
DRUG resistance in cancer cells ,POLYPEPTIDES ,DORSAL root ganglia - Abstract
Antitumor-analgesic peptide (AGAP) is a novel recombinant polypeptide. The primary study showed that AGAP 1.0 mg/kg exhibited strong analgesic and antitumor effects. The tail vein administration of AGAP potently reduced pain behaviors in mice induced by intraplantar injection of formalin or intraperitoneal injection of acetic acid, without affecting basal pain perception. To further assess the mechanisms of AGAP, the effects of AGAP on sodium channels were assessed using the whole-cell patch clamp recordings in dorsal root ganglia (DRG) neurons. The results showed that AGAP (3-1000 nM) inhibited the sodium currents in small-diameter DRG neurons in a dose-dependent manner . 1000 nM AGAP could inhibit the current density-voltage relationship curve of sodium channels in a voltage-dependent manner and negatively shift the activation curves. 1000 nM AGAP could reduce the tetrodotoxin-resistant (TTX-R) sodium currents by 42.8% in small-diameter DRG neurons. Further analysis revealed that AGAP potently inhibited Na V1.8 currents by 59.4%, and negatively shifted the activation and inactivation kinetics. 1000 nM AGAP also reduced the Na 1.9 currents by 33.7%, but had no significant effect on activation and inactivation kinetics. Thus, our results demonstrated that submicromolar concentrations of AGAP inhibited TTX-R sodium channel in rat small-diameter DRG neurons. It is concluded that these new results may better explain, at least in part, the analgesic properties of this polypeptide. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
33. Decreased Nav1.9 channel expression in Hirschsprung's disease.
- Author
-
O'Donnell, Anne-Marie, Coyle, David, and Puri, Prem
- Abstract
Aim Voltage-gated sodium channel subtype 9 (Na v 1.9) are expressed in dorsal root ganglion neurons and are known to be involved in pain during inflammation. Animal studies have reported Na v 1.9 channel expression in myenteric intrinsic primary afferent neurons (IPANs). More recently, a study involving Na v 1.9 knockout mice showed clear evidence of colonic dysmotility. However, there are no data regarding the expression of these channels in the human intestine, thus, the aim of our study was to determine Na v 1.9 channel expression within the human colon and to elucidate if Na v 1.9 channel expression is altered in Hirschsprung's disease (HD). Methods HD tissue specimens ( n = 10) were collected at the time of pull-through surgery, while normal controls were obtained at the time of colostomy closure in patients with imperforate anus ( n = 10). Na v 1.9 immunofluorescence was visualized using confocal microscopy to assess the distribution of the protein. Western blot analysis was undertaken to determine Na v 1.9 protein quantification. Results Confocal microscopy revealed Na v 1.9-immunoreactive neurons within the submucosal and myenteric plexus in normal controls, with a reduction in the HD specimens. Calbindin double-labeling showed that Na v 1.9-immunoreactive neurons were IPANs. Na v 1.9 channels were also seen to be co-localized on smooth muscle cells in all tissues. Western blotting revealed high levels of Na v 1.9 protein expression in normal controls, while there was a marked decrease in Na v 1.9 protein expression in the HD tissue. Conclusion Our results show the expression of Na v 1.9 channels within the human colon for the first time. Furthermore, Na v 1.9 channel expression is decreased in HD versus normal controls. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
34. The voltage-gated sodium channel NaV1.9 in visceral pain.
- Author
-
Hockley, J. R. F., Winchester, W. J., and Bulmer, D. C.
- Subjects
- *
ENTERIC nervous system , *INFLAMMATORY bowel diseases , *IRRITABLE colon , *NOCICEPTORS , *VISCERAL pain , *SODIUM channels - Abstract
Background Visceral pain is a common symptom for patients with gastrointestinal ( GI) disease. It is unpleasant, debilitating, and represents a large unmet medical need for effective clinical treatments. Recent studies have identified NaV1.9 as an important regulator of afferent sensitivity in visceral pain pathways to mechanical and inflammatory stimuli, suggesting that NaV1.9 could represent an important therapeutic target for the treatment of visceral pain. This potential has been highlighted by the identification of patients who have an insensitivity to pain or painful neuropathies associated with mutations in SCN11A, the gene encoding voltage-gated sodium channel subtype 1.9 (NaV1.9). Purpose Here, we address the role of NaV1.9 in visceral pain and what known human NaV1.9 mutants can tell us about NaV1.9 function in gut physiology and pathophysiology. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
35. P2Y Receptors Sensitize Mouse and Human Colonic Nociceptors.
- Author
-
Hockley, James R. F., Tranter, Michael M., McGuire, Cian, Boundouki, George, Cibert-Goton, Vincent, Thaha, Mohamed A., Blackshaw, L. Ashley, Michael, Gregory J., Baker, Mark D., Knowles, Charles H., Winchester, Wendy J., and Bulmer, David C.
- Subjects
- *
SENSORY receptors , *NOCICEPTORS , *INFLAMMATORY mediators , *PYRIMIDINE nucleotides , *EPITHELIAL cells , *ADENOSINE triphosphate - Abstract
Activation of visceral nociceptors by inflammatory mediators contributes to visceral hypersensitivity and abdominal pain associated with many gastrointestinal disorders. Purine and pyrimidine nucleotides (e.g., ATP and UTP) are strongly implicated in this process following their release from epithelial cells during mechanical stimulation of the gut, and from immune cells during inflammation. Actions of ATP are mediated through both ionotropic P2X receptors and metabotropic P2Y receptors. P2X receptor activation causes excitation of visceral afferents; however, the impact of P2Y receptor activation on visceral afferents innervating the gut is unclear. Here we investigate the effects of stimulating P2Y receptors in isolated mouse colonic sensory neurons, and visceral nociceptor fibers in mouse and human nerve-gut preparations. Additionally, we investigate the role of Navl .9 in mediating murine responses. The application of UTP (P2Y2 and P2Y4 agonist) sensitized colonic sensory neurons by increasing action potential firing to current injection and depolarizing the membrane potential. The application of ADP (P2Y2,, P2Y12, and P2Y13 agonist) also increased action potential firing, an effect blocked by the selective P2Y, receptor antagonist MRS2500. UTP or ADP stimulated afferents, including mouse and human visceral nociceptors, in nerve-gut preparations. P2Y! and P2Y2 transcripts were detected in 80% and 56% of retrogradely labeled colonic neurons, respectively. Nav1.9 transcripts colocalized in 86% of P2Y1,-positive and 100% of P2Y2-positive colonic neurons, consistent with reduced afferent fiber responses to UTP and ADP in Nav1.9-/- mice. These data demonstrate that P2Y receptor activation stimulates mouse and human visceral nociceptors, highlighting P2Y-dependent mechanisms in the generation of visceral pain during gastrointestinal disease. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
36. A 49-residue sequence motif in the C terminus of Nav1.9 regulates trafficking of the channel to the plasma membrane
- Author
-
Sulayman D. Dib-Hajj, Carolina Gomis-Perez, Elizabeth J. Akin, Mark Estacion, Stephen G. Waxman, Jianying Huang, and Daria Sizova
- Subjects
0301 basic medicine ,Amino Acid Motifs ,Green Fluorescent Proteins ,Biochemistry ,Nav1.9 ,Structure-Activity Relationship ,03 medical and health sciences ,Chimera (genetics) ,Cytosol ,0302 clinical medicine ,Protein Domains ,Neurobiology ,Live cell imaging ,Humans ,Amino Acid Sequence ,NAV1.9 Voltage-Gated Sodium Channel ,Molecular Biology ,Chemistry ,Sodium channel ,C-terminus ,Cell Membrane ,NAV1.7 Voltage-Gated Sodium Channel ,HEK 293 cells ,Cell Biology ,Cell biology ,Kinetics ,Protein Transport ,HEK293 Cells ,030104 developmental biology ,Heterologous expression ,Sequence motif ,Ion Channel Gating ,030217 neurology & neurosurgery - Abstract
Genetic and functional studies have confirmed an important role for the voltage-gated sodium channel Nav1.9 in human pain disorders. However, low functional expression of Nav1.9 in heterologous systems (e.g. in human embryonic kidney 293 (HEK293) cells) has hampered studies of its biophysical and pharmacological properties and the development of high-throughput assays for drug development targeting this channel. The mechanistic basis for the low level of Nav1.9 currents in heterologous expression systems is not understood. Here, we implemented a multidisciplinary approach to investigate the mechanisms that govern functional Nav1.9 expression. Recombinant expression of a series of Nav1.9-Nav1.7 C-terminal chimeras in HEK293 cells identified a 49-amino-acid-long motif in the C terminus of the two channels that regulates expression levels of these chimeras. We confirmed the critical role of this motif in the context of a full-length channel chimera, Nav1.9-Ct49aaNav1.7, which displayed significantly increased current density in HEK293 cells while largely retaining the characteristic Nav1.9-gating properties. High-resolution live microscopy indicated that the newly identified C-terminal motif dramatically increases the number of channels on the plasma membrane of HEK293 cells. Molecular modeling results suggested that this motif is exposed on the cytoplasmic face of the folded C terminus, where it might interact with other channel partners. These findings reveal that a 49-residue-long motif in Nav1.9 regulates channel trafficking to the plasma membrane.
- Published
- 2019
- Full Text
- View/download PDF
37. Protein kinase C‐α upregulates sodium channel Nav1.9 in nociceptive dorsal root ganglion neurons in an inflammatory arthritis pain model of rat
- Author
-
Jing Cao, Weihua Cai, Qian Bai, Tieli Dong, Jinping Shao, Weidong Zang, Zhiyong Tan, Sanjeeth George, Xiuhua Ren, and Songxue Su
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Protein Kinase C-alpha ,Inflammatory arthritis ,Freund's Adjuvant ,Pain ,Inflammation ,Biochemistry ,Nav1.9 ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Dorsal root ganglion ,Ganglia, Spinal ,Internal medicine ,medicine ,Animals ,NAV1.9 Voltage-Gated Sodium Channel ,Molecular Biology ,Protein kinase C ,Behavior, Animal ,Chemistry ,Nociceptors ,Cell Biology ,medicine.disease ,Arthritis, Experimental ,Rats ,Disease Models, Animal ,030104 developmental biology ,Nociception ,Endocrinology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Hyperalgesia ,Cold sensitivity ,medicine.symptom - Abstract
Previous studies have found that increased expression of Nav1.9 and protein kinase C (PKC) contributes to pain hypersensitivity in a couple of inflammatory pain models. Here we want to observe if PKC can regulate the expression of Nav1.9 in dorsal root ganglion (DRG) in rheumatoid arthritis (RA) pain model. A chronic knee joint inflammation model was produced by intra-articular injection of the complete Freund's adjuvant (CFA) in rats. Nociceptive behaviors including mechanical, cold, and heat hyperalgesia were examined. The expression of Nav1.9 and PKCα in DRG was detected by a quantitative polymerase chain reaction, Western blot, and immunofluorescence. The in vitro and in vivo effects of a PKC activator (phorbol 12-myristate 13-acetate [PMA]) and a PKC inhibitor (GF-109203X) on the expression of Nav1.9 were examined. Moreover, the effects of PKC modulators on nociceptive behaviors were studied. Increased mechanical, heat, and cold sensitivity was observed 3 to 14 days after CFA injection. Parallel increases in messenger RNA and protein expression of Nav1.9 and PKCα were found. Immunofluorescence experiments found that Nav1.9 was preferentially colocalized with IB4+DRG neurons in RA rats. In cultured DRG neurons, PMA increased Nav1.9 expression while GF-109203X prevented the effect of PMA. PMA increased Nav1.9 expression in naïve rats while GF-109203X decreased Nav1.9 expression in RA rats. In naïve rats, PMA caused mechanical and cold hyperalgesia. On the other hand, GF-109203X attenuated mechanical and cold hyperalgesia in RA-pain model. Nav1.9 might be upregulated by PKCα in DRG, which contributes to pain hypersensitivity in CFA-induced chronic knee joint inflammation model of RA pain.
- Published
- 2019
- Full Text
- View/download PDF
38. Positive shift of Nav1.8 current inactivation curve in injured neurons causes neuropathic pain following chronic constriction injury.
- Author
-
GUIXIA LI, XIFANG LIU, JINGNAN DU, JIANZHAO CHEN, FENGLIN SHE, CHUNFU WU, and CHUNLI LI
- Subjects
- *
NEUROPATHY , *SODIUM channel inactivation , *HYPERPOLARIZATION (Cytology) , *NOCICEPTORS , *NEURALGIA , *NEURONS , *MESSENGER RNA , *SENSORY ganglia , *WOUNDS & injuries - Abstract
Neuropathic pain is a global medical concern, characterized by spontaneous pain, heat hyperalgesia and mechanical allodynia. The condition has been associated with alterations in the voltage-gated sodium channels, Nav1.8 and Nav1.9, in nociceptive neurons termed nociceptors. However, an explanation for the contribution of these channels to the phenotype observed in neuropathic pain remains to be elucidated. The changes induced by chronic constriction injury (CCI) to Nav1.8 and Nav1.9 mRNA and protein levels, as well as electrical currents in injured and contralateral non-injured dorsal root ganglion (DRG) neurons are described in the present study. A marked downregulation was observed for each Nav isoform transcript and protein expressed in injured neurons with the exception of the Nav1.9 protein, which exhibited no change, while in contralateral non-injured neurons, the levels of protein and mRNA remained unchanged. Nav isoform functional analysis was then performed in L4-6 DRG neurons 14 days after CCI. The Nav1.8 current density was markedly decreased in injured DRG neurons following CCI. The voltage-dependent activation of the Nav1.8 channel in these neurons was shifted to depolarized potentials by 5.3 mV, while it was shifted to hyperpolarized potentials by 10 mV for inactivation. The electrophysiological function of Nav1.9 was not affected by CCI. The present study demonstrated that ectopic discharge following CCI, which was likely induced by a positive shift in the Nav1.8 current inactivation curve in injured neurons, enhanced the excitability of the neurons by facilitating tetrodotoxin-resistant sodium channels into the fast inactivation state and did not occur as a result of a compensatory redistribution in the contralateral uninjured neurons. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
39. The Domain II S4-S5 Linker in Nav1.9: A Missense Mutation Enhances Activation, Impairs Fast Inactivation, and Produces Human Painful Neuropathy.
- Author
-
Han, Chongyang, Yang, Yang, Greef, Bianca, Hoeijmakers, Janneke, Gerrits, Monique, Verhamme, Camiel, Qu, Jian, Lauria, Giuseppe, Merkies, Ingemar, Faber, Catharina, Dib-Hajj, Sulayman, and Waxman, Stephen
- Abstract
Painful small fiber neuropathy is a challenging medical condition with no effective treatment. Non-genetic causes can be identified in one half of the subjects. Gain-of-function variants of sodium channels Nav1.7 and Nav1.8 have recently been associated with painful small fiber neuropathy. More recently, mutations of sodium channel Nav1.9 have been linked to human pain disorders, with two gain-of-function mutations found in patients with painful small fiber neuropathy. Here we report a novel Nav1.9 mutation, a glycine 699 substitution by arginine (G699R) in the domain II S4-S5 linker, identified in a patient with painful small fiber neuropathy. In this study, we assayed the mutant channels by voltage-clamp in superior cervical ganglion neurons, which do not produce endogenous Nav1.8 or Nav1.9 currents, and provide a novel platform where Nav1.9 is expressed at relatively high levels. Voltage-clamp analysis showed that the mutation hyperpolarizes (−10.1 mV) channel activation, depolarizes (+6.3 mV) steady-state fast inactivation, slows deactivation, and enhances ramp responses compared with wild-type Nav1.9 channels. Current-clamp analysis showed that the G699R mutant channels render dorsal root ganglion neurons hyperexcitable, via depolarized resting membrane potential, reduced current threshold and increased evoked firing. These observations show that the domain II S4-S5 linker plays an important role in the gating of Nav1.9 and demonstrates that a mutation in this linker is linked to a common pain disorder. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
40. Expression and functional role of Nav1.9 sodium channel in cartwheel cells of the dorsal cochlear nucleus.
- Author
-
ZHIYU YAN, YANJUN XU, MIN LIANG, and XIAOWEI REN
- Subjects
- *
INTERNEURONS , *SODIUM channels , *AUDITORY perception , *COCHLEAR nucleus , *DEPOLARIZATION (Cytology) , *WESTERN immunoblotting , *IMMUNOCHEMISTRY - Abstract
In the central auditory system, cartwheel cells (CWCs) are a group of interneurons in the dorsal cochlear nucleus (DCN). While other DCN neurons respond to stimuli with a simple discharge pattern of single action potentials (SAPs), CWCs respond with complex action potentials (CAPs), consisting of SAPs superimposed on a slow depolarization. The CAPs in CWCs may participate in various auditory or non-auditory signaling processing but its intrinsic mechanisms are largely unknown. In the present study, in vitro whole-cell current clamp recordings on neonatal mice brain slices were used to demonstrate that CWCs respond to brief voltage stimulation with CAPs. Western blotting and immunohistochemistry were also utilized to demonstrate that Nav1.9 was expressed in the CWCs. Finally, when Nav1.9 was genetically silenced, CWCs responded to voltage stimulation with SAPs, not CAPs. The results strongly suggested that Nav1.9 was expressed and functionally contributed to the signaling processing in the central auditory pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
41. The insecticide deltamethrin enhances sodium channel slow inactivation of human Nav1.9, Nav1.8 and Nav1.7
- Author
-
Angelika Lampert and Stefanie Bothe
- Subjects
Pharmacology ,Membrane potential ,Insecticides ,Pyrethroid ,Dose-Response Relationship, Drug ,Sodium channel ,NAV1.7 Voltage-Gated Sodium Channel ,Toxicology ,Membrane Potentials ,Nav1.9 ,NAV1.8 Voltage-Gated Sodium Channel ,chemistry.chemical_compound ,Electrophysiology ,HEK293 Cells ,Deltamethrin ,chemistry ,Nitriles ,Pyrethrins ,parasitic diseases ,NAV1 ,Biophysics ,Humans ,Patch clamp ,NAV1.9 Voltage-Gated Sodium Channel - Abstract
Toxicology and applied pharmacology : TAP 428, 115676 (2021). doi:10.1016/j.taap.2021.115676, Published by Academic Press, Orlando, Fla.
- Published
- 2021
- Full Text
- View/download PDF
42. Persistent modification of Nav1.9 following chronic exposure to insecticides and pyridostigmine bromide.
- Author
-
Nutter, Thomas J. and Cooper, Brian Y.
- Subjects
- *
PAIN management , *PHYSIOLOGICAL effects of sodium , *INSECTICIDES , *PYRIDOSTIGMINE bromide , *CHRONIC pain , *CHLORPYRIFOS - Abstract
Abstract: Many veterans of the 1991 Gulf War (GW) returned from that conflict with a widespread chronic pain affecting deep tissues. Recently, we have shown that a 60day exposure to the insecticides permethrin, chlorpyrifos, and pyridostigmine bromide (NTPB) had little influence on nociceptor action potential forming Nav1.8, but increased Kv7 mediated inhibitory currents 8weeks after treatment. Using the same exposure regimen, we used whole cell patch methods to examine whether the influences of NTPB could be observed on Nav1.9 expressed in muscle and vascular nociceptors. During a 60day exposure to NTPB, rats exhibited lowered muscle pain thresholds and increased rest periods, but these measures subsequently returned to normal levels. Eight and 12weeks after treatments ceased, DRG neurons were excised from the sensory ganglia. Whole cell patch studies revealed little change in voltage dependent activation and deactivation of Nav1.9, but significant increases in the amplitude of Nav1.9 were observed 8weeks after exposure. Cellular studies, at the 8week delay, revealed that NTPB also significantly prolonged action potential duration and afterhyperpolarization (22°C). Acute application of permethrin (10μM) also increased the amplitude of Nav1.9 in skin, muscle and vascular nociceptors. In conclusion, chronic exposure to Gulf War agents produced long term changes in the amplitude of Nav1.9 expressed in muscle and vascular nociceptors. The reported increases in Kv7 amplitude may have been an adaptive response to increased Nav1.9, and effectively suppressed behavioral pain measures in the post treatment period. Factors that alter the balance between Nav1.9 and Kv7 could release spontaneous discharge and produce chronic deep tissue pain. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
43. Spider venom-derived peptide induces hyperalgesia in Nav1.7 knockout mice by activating Nav1.9 channels
- Author
-
Lulu Li, Minzhi Chen, Shuijiao Peng, Xi Zhou, Yazhou Huang, Jing Yu Liu, Zhen Xiao, Zhouquan Wang, Luyao Yang, Songping Liang, Zhonghua Liu, Qingfeng Zhang, Li Wang, Xianwei Zhang, and Tingbin Ma
- Subjects
0301 basic medicine ,Male ,Science ,General Physics and Astronomy ,Pain ,Spider Venoms ,Pharmacology ,Ion channels in the nervous system ,General Biochemistry, Genetics and Molecular Biology ,Article ,Nav1.9 ,NAV1.8 Voltage-Gated Sodium Channel ,03 medical and health sciences ,0302 clinical medicine ,Dorsal root ganglion ,Ganglia, Spinal ,medicine ,Animals ,Humans ,Amino Acid Sequence ,lcsh:Science ,NAV1.9 Voltage-Gated Sodium Channel ,Mice, Knockout ,Neurons ,Multidisciplinary ,Chemistry ,Sodium channel ,NAV1.7 Voltage-Gated Sodium Channel ,General Chemistry ,medicine.disease ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Nociception ,Hyperalgesia ,Knockout mouse ,Nociceptor ,lcsh:Q ,Female ,medicine.symptom ,Peripheral nervous system ,Peptides ,Ion Channel Gating ,030217 neurology & neurosurgery ,Congenital insensitivity to pain - Abstract
The sodium channels Nav1.7, Nav1.8 and Nav1.9 are critical for pain perception in peripheral nociceptors. Loss of function of Nav1.7 leads to congenital insensitivity to pain in humans. Here we show that the spider peptide toxin called HpTx1, first identified as an inhibitor of Kv4.2, restores nociception in Nav1.7 knockout (Nav1.7-KO) mice by enhancing the excitability of dorsal root ganglion neurons. HpTx1 inhibits Nav1.7 and activates Nav1.9 but does not affect Nav1.8. This toxin produces pain in wild-type (WT) and Nav1.7-KO mice, and attenuates nociception in Nav1.9-KO mice, but has no effect in Nav1.8-KO mice. These data indicate that HpTx1-induced hypersensitivity is mediated by Nav1.9 activation and offers pharmacological insight into the relationship of the three Nav channels in pain signalling., Loss of function of Nav1.7 leads to congenital insensitivity to pain in humans. Here the authors found that activation of Nav1.9 can restore nociception in Nav1.7 knockout mice, revealed by a venom-derived peptide as a probe.
- Published
- 2020
44. NaV1.9 channels in muscle afferent neurons and axons
- Author
-
Ethan A. Remily, Ankeeta K Heier, Tyler L Marler, Renuka Ramachandra, Jeong Sook Kim-Han, Kristina L Elmslie, Keith S. Elmslie, and Andrew B. Wright
- Subjects
Male ,Reflex, Stretch ,0301 basic medicine ,Patch-Clamp Techniques ,Small diameter ,Physiology ,Normal Distribution ,Action Potentials ,Muscle blood flow ,Signal ,Afferent Neurons ,Nav1.9 ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Ganglia, Spinal ,Animals ,Neurons, Afferent ,Patch clamp ,Muscle activity ,Furans ,Muscle, Skeletal ,NAV1.9 Voltage-Gated Sodium Channel ,Aniline Compounds ,Chemistry ,General Neuroscience ,Immunohistochemistry ,Axons ,Rats ,030104 developmental biology ,Microscopy, Fluorescence ,Neuroscience ,030217 neurology & neurosurgery ,Research Article ,Muscle Contraction ,Sodium Channel Blockers - Abstract
The exercise pressor reflex (EPR) is activated by muscle contractions to increase heart rate and blood pressure during exercise. While this reflex is beneficial in healthy individuals, the reflex activity is exaggerated in patients with cardiovascular disease, which is associated with increased mortality. Group III and IV afferents mediate the EPR and have been shown to express both tetrodotoxin-sensitive (TTX-S, NaV1.6, and NaV1.7) and -resistant (TTX-R, NaV1.8, and NaV1.9) voltage-gated sodium (NaV) channels, but NaV1.9 current has not yet been demonstrated. Using a F−-containing internal solution, we found a NaVcurrent in muscle afferent neurons that activates at around −70 mV with slow activation and inactivation kinetics, as expected from NaV1.9 current. However, this current ran down with time, which resulted, at least in part, from increased steady-state inactivation since it was slowed by both holding potential hyperpolarization and a depolarized shift of the gating properties. We further show that, following NaV1.9 current rundown (internal F−), application of the NaV1.8 channel blocker A803467 inhibited significantly more TTX-R current than we had previously observed (internal Cl−), which suggests that NaV1.9 current did not rundown with that internal solution. Using immunohistochemistry, we found that the majority of group IV somata and axons were NaV1.9 positive. The majority of small diameter myelinated afferent somata (putative group III) were also NaV1.9 positive, but myelinated muscle afferent axons were rarely labeled. The presence of NaV1.9 channels in muscle afferents supports a role for these channels in activation and maintenance of the EPR.NEW & NOTEWORTHY Small diameter muscle afferents signal pain and muscle activity levels. The muscle activity signals drive the cardiovascular system to increase muscle blood flow, but these signals can become exaggerated in cardiovascular disease to exacerbate cardiac damage. The voltage-dependent sodium channel NaV1.9 plays a unique role in controlling afferent excitability. We show that NaV1.9 channels are expressed in muscle afferents, which supports these channels as a target for drug development to control hyperactivity of these neurons.
- Published
- 2018
- Full Text
- View/download PDF
45. Heat-resistant action potentials require TTX-resistant sodium channels NaV1.8 and NaV1.9
- Author
-
Andreas Leffler, Filip Touska, Katharina Zimmermann, Peter W. Reeh, Viktorie Vlachova, and Brian Turnquist
- Subjects
0301 basic medicine ,Heat resistant ,Physiology ,Subthreshold conduction ,Sodium channel ,Nav1.9 ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,nervous system ,chemistry ,Threshold of pain ,NAV1 ,Tetrodotoxin ,Biophysics ,Nociceptor ,030217 neurology & neurosurgery - Abstract
Damage-sensing nociceptors in the skin provide an indispensable protective function thanks to their specialized ability to detect and transmit hot temperatures that would block or inflict irreversible damage in other mammalian neurons. Here we show that the exceptional capacity of skin C-fiber nociceptors to encode noxiously hot temperatures depends on two tetrodotoxin (TTX)-resistant sodium channel α-subunits: NaV1.8 and NaV1.9. We demonstrate that NaV1.9, which is commonly considered an amplifier of subthreshold depolarizations at 20°C, undergoes a large gain of function when temperatures rise to the pain threshold. We also show that this gain of function renders NaV1.9 capable of generating action potentials with a clear inflection point and positive overshoot. In the skin, heat-resistant nociceptors appear as two distinct types with unique and possibly specialized features: one is blocked by TTX and relies on NaV1.9, and the second type is insensitive to TTX and composed of both NaV1.8 and NaV1.9. Independent of rapidly gated TTX-sensitive NaV channels that form the action potential at pain threshold, NaV1.8 is required in all heat-resistant nociceptors to encode temperatures higher than ∼46°C, whereas NaV1.9 is crucial for shaping the action potential upstroke and keeping the NaV1.8 voltage threshold within reach.
- Published
- 2018
- Full Text
- View/download PDF
46. Correlation of Nav1.8 and Nav1.9 sodium channel expression with neuropathic pain in human subjects with lingual nerve neuromas.
- Author
-
Bird, Emma V., Christmas, Claire R., Loescher, Alison R., Smith, Keith G., Robinson, Peter P., Black, Joel A., Waxman, Stephen G., and Boissonade, Fiona M.
- Subjects
- *
NERVOUS system injuries , *TRIGEMINAL nerve , *NEUROPATHY , *SENSORY neurons , *HUMAN research subjects - Abstract
Background Voltage-gated sodium channels Nav1.8 and Nav1.9 are expressed preferentially in small diameter sensory neurons, and are thought to play a role in the generation of ectopic activity in neuronal cell bodies and/or their axons following peripheral nerve injury. The expression of Nav1.8 and Nav1.9 has been quantified in human lingual nerves that have been previously injured inadvertently during lower third molar removal, and any correlation between the expression of these ion channels and the presence or absence of dysaesthesia investigated. Results Immunohistochemical processing and quantitative image analysis revealed that Nav1.8 and Nav1.9 were expressed in human lingual nerve neuromas from patients with or without symptoms of dysaesthesia. The level of Nav1.8 expression was significantly higher in patients reporting pain compared with no pain, and a significant positive correlation was observed between levels of Nav1.8 expression and VAS scores for the symptom of tingling. No significant differences were recorded in the level of expression of Nav1.9 between patients with or without pain. Conclusions These results demonstrate that Nav1.8 and Nav1.9 are present in human lingual nerve neuromas, with significant correlations between the level of expression of Nav1.8 and symptoms of pain. These data provide further evidence that changes in expression of Nav1.8 are important in the development and/or maintenance of nerve injury-induced pain, and suggest that Nav1.8 may be a potential therapeutic target. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
47. The scorpion toxin Amm VIII induces pain hypersensitivity through gain-of-function of TTX-sensitive Na+ channels.
- Author
-
Abbas, Najwa, Gaudioso-Tyzra, Christelle, Bonnet, Caroline, Gabriac, Mélanie, Amsalem, Muriel, Lonigro, Aurélie, Padilla, Françoise, Crest, Marcel, Martin-Eauclaire, Marie-France, and Delmas, Patrick
- Subjects
- *
ALLERGIES , *ANDROCTONUS australis , *HYPERALGESIA , *SCORPION venom , *SODIUM channels , *LABORATORY mice ,PHYSIOLOGICAL effects of venom - Abstract
Abstract: Voltage-gated Na+ channels (Nav) are the targets of a variety of scorpion toxins. Here, we investigated the effects of Amm VIII, a toxin isolated from the venom of the scorpion Androctonus mauretanicus mauretanicus, on pain-related behaviours in mice. The effects of Amm VIII were compared with the classic scorpion α-toxin AaH II from Androctonus australis. Contrary to AaH II, intraplantar injection of Amm VIII at relatively high concentrations caused little nocifensive behaviours. However, Amm VIII induced rapid mechanical and thermal pain hypersensitivities. We evaluated the toxins’ effects on Nav currents in nociceptive dorsal root ganglion (DRG) neurons and immortalized DRG neuron-derived F11 cells. Amm VIII and AaH II enhanced tetrodotoxin-sensitive (TTX-S) Nav currents in DRG and F11 cells. Both toxins impaired fast inactivation and negatively shifted activation. AaH II was more potent than Amm VIII at modulating TTX-S Nav currents with EC50 of 5nM and 1μM, respectively. AaH II and Amm VIII also impaired fast inactivation of Nav1.7, with EC50 of 6.8nM and 1.76μM, respectively. Neither Nav1.8 nor Nav1.9 was affected by the toxins. AaH II and Amm VIII reduced first spike latency and lowered action potential threshold. Amm VIII was less efficient than AaH II in increasing the gain of the firing frequency-stimulation relationship. In conclusion, our data show that Amm VIII, although less potent than AaH II, acts as a gating-modifier peptide reminiscent of classic α-toxins, and suggest that its hyperalgesic effects can be ascribed to gain-of-function of TTX-S Na+ channels in nociceptors. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
48. Effect of amitriptyline on tetrodotoxin-resistant Nav1.9 currents in nociceptive trigeminal neurons.
- Author
-
Jingyao Liang, Xiaoyan Liu, Jianquan Zheng, and Shengyuan Yu
- Subjects
- *
AMITRIPTYLINE , *TETRODOTOXIN , *ANTIDEPRESSANTS , *PAIN management , *MIGRAINE , *LABORATORY rats - Abstract
Background: Amitriptyline (AMI) is tricyclic antidepressant that has been widely used to manage various chronic pains such as migraines. Its efficacy is attributed to its blockade of voltage-gated sodium channels (VGSCs). However, the effects of AMI on the tetrodotoxin-resistant (TTX-r) sodium channel Nav1.9 currents have been unclear to present. Results: Using a whole-cell patch clamp technique, this study showed that AMI efficiently inhibited Nav1.9 currents in a concentration-dependent manner and had an IC50 of 15.16 μM in acute isolated trigeminal ganglion (TG) neurons of the rats. 10 μM AMI significantly shifted the steady-state inactivation of Nav1.9 channels in the hyperpolarizing direction without affecting voltage-dependent activation. Surprisingly, neither 10 nor 50 μM AMI caused a use-dependent blockade of Nav1.9 currents elicited by 60 pulses at 1 Hz. Conclusion: These data suggest that AMI is a state-selective blocker of Nav1.9 channels in rat nociceptive trigeminal neurons, which likely contributes to the efficacy of AMI in treating various pains, including migraines. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
49. Conotoxins Targeting Neuronal Voltage-Gated Sodium Channel Subtypes: Potential Analgesics?
- Author
-
Knapp, Oliver, McArthur, Jeffrey R., and Adams, David J.
- Subjects
CONOTOXINS ,ANALGESICS ,CELLS ,NEURONS ,IMMUNOSPECIFICITY ,IMMUNOGLOBULIN idiotypes ,NERVOUS system ,ANTIBODY diversity - Abstract
Voltage-gated sodium channels (VGSC) are the primary mediators of electrical signal amplification and propagation in excitable cells. VGSC subtypes are diverse, with different biophysical and pharmacological properties, and varied tissue distribution. Altered VGSC expression and/or increased VGSC activity in sensory neurons is characteristic of inflammatory and neuropathic pain states. Therefore, VGSC modulators could be used in prospective analgesic compounds. VGSCs have specific binding sites for four conotoxin families: μ-, μO-, δ- and í-conotoxins. Various studies have identified that the binding site of these peptide toxins is restricted to well-defined areas or domains. To date, only the μ- and μO-family exhibit analgesic properties in animal pain models. This review will focus on conotoxins from the μ- and μO-families that act on neuronal VGSCs. Examples of how these conotoxins target various pharmacologically important neuronal ion channels, as well as potential problems with the development of drugs from conotoxins, will be discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
50. The distribution of low-threshold TTX-resistant Na+ currents in rat trigeminal ganglion cells
- Author
-
Scroggs, R.S.
- Subjects
- *
SODIUM ions , *LABORATORY rats , *GANGLIA , *TETRODOTOXIN , *DRUG resistance , *TEMPOROMANDIBULAR joint , *EVOKED potentials (Electrophysiology) - Abstract
Abstract: The distribution of low-threshold tetrodotoxin-resistant (TTX-r) Na+ current and its co-expression with high-threshold TTX-r Na+ current were studied in randomly selected acutely dissociated rat trigeminal ganglion (non-identified TG cells) and TG cells serving the temporomandibular joint (TMJ-TG cells). Conditions previously shown to enhance NaV1.9 channel-mediated currents (holding potential (HP) −80mV, 130-mM fluoride internally) were employed to amplify the low-threshold Na+ current. Under these conditions, detectable low-threshold Na+ current was exhibited by 16 out of 21 non-identified TG cells (average, 1810±358pA), and by nine of 14 TMJ-TG cells (average, 959±525pA). The low-threshold Na+ current began to activate around −55mV and was inactivated by holding TG cells at −60mV and delivering 40-ms test potentials (TPs) to 0mV. The inactivation was long lasting, recovering only 8±3% over a 5-min period after the HP was returned to −80mV. Following low-threshold Na+ current inactivation, high-threshold TTX-r Na+ current, evoked from HP −60mV, was observed. High-threshold Na+ current amplitude averaged 16,592±3913pA for TPs to 0mV, was first detectable at an average TP of −34±1.3mV, and was ½ activated at −7.1±2.3mV. In TG cells expressing prominent low-threshold Na+ currents, changing the external solution to one containing 0mM Na+ reduced the amount of current required to hold the cells at −80mV through −50mV, the peak effect being observed at HP −60mV. TG cells recorded from with a more physiological pipette solution containing chloride instead of fluoride exhibited small low-threshold Na+ currents, which were greatly increased upon superfusion of the TG cells with the adenylyl cyclase (AC) activator forskolin. These data suggest two hypotheses: (1) low- and high-threshold NaV1.9 and NaV1.8 channels, respectively, are frequently co-expressed in TG neurons serving the TMJ and other structures, and (2), NaV1.9 channel-mediated currents are small under physiological conditions, but may be enhanced by inflammatory mediators that increase AC activity, and may mediate an inward leak that depolarizes TG neurons, increasing their excitability. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.