1
|
Zhang X, Hartung JE, Gold MS. Persistent (Na v 1.9) sodium currents in human dorsal root ganglion neurons. Pain 2025; 166:448-459. [PMID: 39297710 PMCID: PMC11723807 DOI: 10.1097/j.pain.0000000000003394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/07/2024] [Indexed: 10/12/2024]
Abstract
ABSTRACT Na v 1.9 is of interest to the pain community for a number of reasons, including the human mutations in the gene encoding Na v 1.9, SCN11a , that are associated with both pain and loss of pain phenotypes. However, because much of what we know about the biophysical properties of Na v 1.9 has been learned through the study of rodent sensory neurons, and there is only 76% identity between human and rodent homologs of SCN11a , there is reason to suggest that there may be differences in the biophysical properties of the channels in human and rodent sensory neurons, and consequently, the contribution of these channels to the control of sensory neuron excitability, if not pain. Thus, the purpose of this study was to characterize Na v 1.9 currents in human sensory neurons and compare the properties of these currents with those in rat sensory neurons recorded under identical conditions. Whole-cell patch clamp techniques were used to record Na v 1.9 currents in isolated sensory neurons in vitro. Our results indicate that several of the core biophysical properties of the currents, including persistence and a low threshold for activation, are conserved across species. However, we noted a number of potentially important differences between the currents in human and rat sensory neurons including a lower threshold for activation, higher threshold for inactivation, slower deactivation, and faster recovery from slow inactivation. Human Na v 1.9 was inhibited by inflammatory mediators, whereas rat Na v 1.9 was potentiated. Our results may have implications for the role of Na v 1.9 in sensory, if not nociceptive signaling.
Collapse
Affiliation(s)
- Xiulin Zhang
- Department of Neurobiology, and the Pittsburgh Center for Pain Research University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Urology, the Second Hospital of Shandong University, 250032, P.R. China
| | - Jane E Hartung
- Department of Neurobiology, and the Pittsburgh Center for Pain Research University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michael S Gold
- Department of Neurobiology, and the Pittsburgh Center for Pain Research University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Dib-Hajj SD, Waxman SG. Species-specific differences and the role of Na v 1.9 in pain pathophysiology. Pain 2025; 166:231-233. [PMID: 39297718 DOI: 10.1097/j.pain.0000000000003395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 01/12/2025]
Affiliation(s)
- Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
- Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
- Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
3
|
Ghovanloo MR, Tyagi S, Zhao P, Waxman SG. Nav1.8, an analgesic target for nonpsychotomimetic phytocannabinoids. Proc Natl Acad Sci U S A 2025; 122:e2416886122. [PMID: 39835903 DOI: 10.1073/pnas.2416886122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Pain impacts billions of people worldwide, but treatment options are limited and have a spectrum of adverse effects. The search for safe and nonaddictive pain treatments has led to a focus on key mediators of nociceptor excitability. Voltage-gated sodium (Nav) channels in the peripheral nervous system-Nav1.7, Nav1.8, and Nav1.9-play crucial roles in pain signaling. Among these, Nav1.8 has shown promise due to its rapid recovery from inactivation and role in repetitive firing, with recent clinical studies providing proof-of-principal that block of Nav1.8 can reduce pain in humans. We report here that three nonpsychotomimetic cannabinoids-cannabidiol (CBD), cannabigerol (CBG), and cannabinol (CBN)-effectively inhibit Nav1.8, suggesting their potential as analgesic compounds. In particular, CBG shows significant promise due to its ability to effectively inhibit excitability of peripheral sensory neurons. These findings highlight the therapeutic potential of cannabinoids, particularly CBG, as agents that may attenuate pain via block of Nav1.8, warranting further in vivo studies.
Collapse
Affiliation(s)
- Mohammad-Reza Ghovanloo
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Sidharth Tyagi
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT 06520
| | - Peng Zhao
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| |
Collapse
|
4
|
Peng S, Chen M, Wu M, Liu Z, Tang D, Zhou X. Elucidating the roles of voltage sensors in Na V1.9 activation and inactivation through a spider toxin. Biochim Biophys Acta Gen Subj 2025; 1869:130762. [PMID: 39800272 DOI: 10.1016/j.bbagen.2025.130762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
The gating process of voltage-gated sodium (NaV) channels is extraordinary intrinsic and involves numerous factors, such as voltage-sensing domain (VSD), the N-terminus and C-terminus, and the auxiliary subunits. To date, the gating mechanism of NaV channel has not been clearly elucidated. NaV1.9 has garnered significant attention due to its slow gating kinetics. Due to the challenges of NaV1.9 heterologous expression, research on its gating mechanism is relatively limited. Whether there are any differences in the functions of the four VSDs in NaV1.9 compared to those in other subtypes remains an open question. Here, we employed the established chimera method to transplant the S3b-S4 motif from the VSDIV of the toxin-sensitive donor channel (NaV1.9) into the receptor channel (NaV1.9/1.8 DIV S3b-S4 chimera). This modification imparted animal toxin sensitivity to the other three VSDs. Our results demonstrate that all four VSDs of NaV1.9 are involved in channel opening, VSDIII and VSDIV are primarily involved in regulating fast inactivation, and VSDII also regulates the steady-state inactivation of channels. These findings provide a new insight into the gating mechanism of NaV1.9.
Collapse
Affiliation(s)
- Shuijiao Peng
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Hunan Provincial Center for Disease Control and Prevention, Hunan Provincial Key Laboratory of Microbial Molecular Biology, Changsha 410000, Hunan, China
| | - Minzhi Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, Hunan, China
| | - Meijing Wu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, Hunan, China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, Hunan, China
| | - Dongfang Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou, 425199, Hunan, China.
| | - Xi Zhou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, Hunan, China.
| |
Collapse
|
5
|
Song Y, Xu Z, Zhang L, Gao L. Sigma-1 Receptor Modulates CFA-Induced Inflammatory Pain via Sodium Channels in Small DRG Neurons. Biomolecules 2025; 15:73. [PMID: 39858467 PMCID: PMC11764217 DOI: 10.3390/biom15010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/30/2025] Open
Abstract
The sigma-1 receptor (Sig-1R) has emerged as a significant target in the realm of pain management and has been the subject of extensive research. Nonetheless, its specific function in inflammatory pain within dorsal root ganglion (DRG) neurons remains inadequately elucidated. This study utilized whole-cell patch clamp techniques, single-cell real-time PCR, and immunohistochemistry to examine the influence of Sig-1R on inflammatory pain induced by complete Freund's adjuvant (CFA) in a rat model. Our results revealed several key findings: (1) The expression of Sig-1R was found to be upregulated during the progression of inflammatory pain, with a notable translocation from the cytoplasm to the membrane; (2) Inhibition of peripheral Sig-1R using S1RA resulted in a reduction of CFA-induced allodynia; (3) Activation of Sig-1R through PRE-084 led to a decrease in the fast sodium current in isolated DRG neurons from CFA-treated rats, which was associated with a diminished action potential (AP) peak and maximum depolarizing rate (MDR), as well as an increased rheobase; (4) Furthermore, PRE-084 was observed to enhance the slow component of the sodium current, resulting in hyperpolarization of the threshold potential and an increase in AP firing frequency, alongside an elevation in the mRNA expression of the slow sodium channel Nav1.9 in CFA-treated rats. In conclusion, our findings suggest that the modulation of sodium channels by Sig-1R in DRG neurons plays a significant role in the mechanisms underlying inflammatory pain.
Collapse
Affiliation(s)
- Yuanlong Song
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| | - Zifen Xu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Liangpin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| | - Linlin Gao
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| |
Collapse
|
6
|
Heinle JW, Dalessio S, Janicki P, Ouyang A, Vrana KE, Ruiz-Velasco V, Coates MD. Insights into the voltage-gated sodium channel, Na V1.8, and its role in visceral pain perception. Front Pharmacol 2024; 15:1398409. [PMID: 38855747 PMCID: PMC11158627 DOI: 10.3389/fphar.2024.1398409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 06/11/2024] Open
Abstract
Pain is a major issue in healthcare throughout the world. It remains one of the major clinical issues of our time because it is a common sequela of numerous conditions, has a tremendous impact on individual quality of life, and is one of the top drivers of cost in medicine, due to its influence on healthcare expenditures and lost productivity in those affected by it. Patients and healthcare providers remain desperate to find new, safer and more effective analgesics. Growing evidence indicates that the voltage-gated sodium channel Nav1.8 plays a critical role in transmission of pain-related signals throughout the body. For that reason, this channel appears to have strong potential to help develop novel, more selective, safer, and efficacious analgesics. However, many questions related to the physiology, function, and clinical utility of Nav1.8 remain to be answered. In this article, we discuss the latest studies evaluating the role of Nav1.8 in pain, with a particular focus on visceral pain, as well as the steps taken thus far to evaluate its potential as an analgesic target. We also review the limitations of currently available studies related to this topic, and describe the next scientific steps that have already been undertaken, or that will need to be pursued, to fully unlock the capabilities of this potential therapeutic target.
Collapse
Affiliation(s)
- J. Westley Heinle
- Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA, United States
| | - Shannon Dalessio
- Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA, United States
| | - Piotr Janicki
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA, United States
| | - Ann Ouyang
- Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA, United States
| | - Kent E. Vrana
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA, United States
| | - Matthew D. Coates
- Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA, United States
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
7
|
Zhao C, Zhou X, Shi X. The influence of Nav1.9 channels on intestinal hyperpathia and dysmotility. Channels (Austin) 2023; 17:2212350. [PMID: 37186898 DOI: 10.1080/19336950.2023.2212350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
The Nav1.9 channel is a voltage-gated sodium channel. It plays a vital role in the generation of pain and the formation of neuronal hyperexcitability after inflammation. It is highly expressed in small diameter neurons of dorsal root ganglions and Dogiel II neurons in enteric nervous system. The small diameter neurons in dorsal root ganglions are the primary sensory neurons of pain conduction. Nav1.9 channels also participate in regulating intestinal motility. Functional enhancements of Nav1.9 channels to a certain extent lead to hyperexcitability of small diameter dorsal root ganglion neurons. The hyperexcitability of the neurons can cause visceral hyperalgesia. Intestinofugal afferent neurons and intrinsic primary afferent neurons in enteric nervous system belong to Dogiel type II neurons. Their excitability can also be regulated by Nav1.9 channels. The hyperexcitability of intestinofugal afferent neurons abnormally activate entero-enteric inhibitory reflexes. The hyperexcitability of intrinsic primary afferent neurons disturb peristaltic waves by abnormally activating peristaltic reflexes. This review discusses the role of Nav1.9 channels in intestinal hyperpathia and dysmotility.
Collapse
Affiliation(s)
- Chenyu Zhao
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiaoliu Shi
- Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
8
|
Jessup D, Woods K, Thakker S, Damaj MI, Akbarali HI. Short-chain fatty acid, butyrate prevents morphine-and paclitaxel-induced nociceptive hypersensitivity. Sci Rep 2023; 13:17805. [PMID: 37853033 PMCID: PMC10584825 DOI: 10.1038/s41598-023-44857-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023] Open
Abstract
Nociceptive hypersensitivity is a significant side effect with the chronic administration of opioids as well as chemotherapeutics. Both opioid-induced hypersensitivity (OIH) and chemotherapy-induced hypersensitivity (CIH) are characterized by an increased sensitivity to painful stimuli which can significantly reduce the quality of life for individuals on either drug(s). Here we demonstrate the nociceptive hypersensitivity associated with repeated administration of morphine (opioid) and paclitaxel (chemotherapeutic) treatment can be reversed by oral supplementation with the short-chain fatty acid (SCFA) sodium butyrate (NaBut). In two separate mouse behavioral models for nociceptive hypersensitivity, we found that thermal hyperalgesia (for OIH) and cold allodynia (for CIH) were prevented by treatment with oral butyrate (p.o, b.i.d). Electrophysiological recordings of small diameter dorsal root ganglia (DRG) neurons from morphine and paclitaxel treated mice showed an increase in neuronal hyperexcitability in both drug models which was likewise prevented by oral butyrate treatment. Using colonic conditioned media obtained from excised colon segments we found that gut mediators of morphine treated mice can induce hyperexcitability in naïve DRG neurons, but such enhanced excitability is not present when animals are co-treated with NaBut suggesting gut derived mediators modulate neuronal hyperexcitability. In-vitro NaBut treatment did not prevent morphine-induced excitability, suggesting an indirect role of butyrate in modulating neuronal hypersensitivity. These data taken together suggest that gut derived mediators affect opioid and chemotherapeutic-induced neuronal hypersensitivity that is prevented by the SCFA butyrate.
Collapse
Affiliation(s)
- Dawn Jessup
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, 23298, USA
| | - Kareem Woods
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, 23298, USA
| | - Sach Thakker
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, 23298, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, 23298, USA
| | - Hamid I Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, 23298, USA.
| |
Collapse
|
9
|
Marchese-Rojas M, Islas ÁA, Mancilla-Simbro C, Millan-PerezPeña L, León JS, Salinas-Stefanon EM. Inhibition of the Human Neuronal Sodium Channel Na v1.9 by Arachidonyl-2-Chloroethylamide, An Analogue of Anandamide in a hNa v1.9/rNa v1.4 Chimera, An Experimental and in Silico Study. Neuroscience 2023; 511:39-52. [PMID: 36156289 DOI: 10.1016/j.neuroscience.2022.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/12/2022] [Accepted: 09/16/2022] [Indexed: 01/23/2023]
Abstract
Cannabinoids regulate analgesia, which has aroused much interest in identifying new pharmacological therapies in the management of refractory pain. Voltage-gated Na+ channels (Navs) play an important role in inflammatory and neuropathic pain. In particular, Nav1.9 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 hNav1.9_C4, that comprises the extracellular and transmembrane domains of hNav1.9, 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 (ki = 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 hNav1.9, contacting residues that bind cannabinol (CBD) in the NavMs 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.
Collapse
Affiliation(s)
- Mario Marchese-Rojas
- Laboratory of Biophysics, Institute of Physiology, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Ángel A Islas
- Vicerrectoría de Investigación y Estudios de Posgrado, Benemérita Universidad Autónoma de Puebla, Mexico; Laboratory of Computational Molecular Simulations, Department of Pharmacy, Benemérita Universidad Autónoma de Puebla, Mexico.
| | - Claudia Mancilla-Simbro
- Laboratory of Biophysics, Institute of Physiology, Benemérita Universidad Autónoma de Puebla, Mexico
| | | | - Jorge S León
- Laboratory of Biophysics, Institute of Physiology, Benemérita Universidad Autónoma de Puebla, Mexico
| | | |
Collapse
|
10
|
Plumbly W, Patikas N, Field SF, Foskolou S, Metzakopian E. Derivation of nociceptive sensory neurons from hiPSCs with early patterning and temporally controlled NEUROG2 overexpression. CELL REPORTS METHODS 2022; 2:100341. [PMID: 36452863 PMCID: PMC9701618 DOI: 10.1016/j.crmeth.2022.100341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 06/09/2022] [Accepted: 10/24/2022] [Indexed: 06/17/2023]
Abstract
Despite development of protocols to differentiate human pluripotent stem cells (hPSCs), those used to produce sensory neurons remain difficult to replicate and result in heterogenous populations. There is a growing clinical burden of chronic pain conditions, highlighting the need for relevant human cellular models. This study presents a hybrid differentiation method to produce nociceptive sensory neurons from hPSCs. Lines harboring an inducible NEUROG2 construct were patterned toward precursors with small molecules followed by NEUROG2 overexpression. Neurons expressed key markers, including BRN3A and ISL1, with single-cell RNA sequencing, revealing populations of nociceptors expressing SCN9A and TRP channels. Physiological profiling with multi-electrode arrays revealed that neurons responded to noxious stimuli, including capsaicin. Finally, we modeled pain-like states to identify genes and pathways involved in pain transduction. This study presents an optimized method to efficiently produce nociceptive sensory neurons and provides a tool to aid development of chronic pain research.
Collapse
Affiliation(s)
- William Plumbly
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Nikolaos Patikas
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| | - Sarah F. Field
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Stefanie Foskolou
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Emmanouil Metzakopian
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
11
|
Role of the Guanidinium Groups in Ligand–Receptor Binding of Arginine-Containing Short Peptides to the Slow Sodium Channel: Quantitative Approach to Drug Design of Peptide Analgesics. Int J Mol Sci 2022; 23:ijms231810640. [PMID: 36142549 PMCID: PMC9505852 DOI: 10.3390/ijms231810640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/07/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
Several arginine-containing short peptides have been shown by the patch-clamp method to effectively modulate the NaV1.8 channel activation gating system, which makes them promising candidates for the role of a novel analgesic medicinal substance. As demonstrated by the organotypic tissue culture method, all active and inactive peptides studied do not trigger the downstream signaling cascades controlling neurite outgrowth and should not be expected to evoke adverse side effects on the tissue level upon their medicinal administration. The conformational analysis of Ac-RAR-NH2, Ac-RER-NH2, Ac-RAAR-NH2, Ac-REAR-NH2, Ac-RERR-NH2, Ac-REAAR-NH2, Ac-PRERRA-NH2, and Ac-PRARRA-NH2 has made it possible to find the structural parameter, the value of which is correlated with the target physiological effect of arginine-containing short peptides. The distances between the positively charged guanidinium groups of the arginine side chains involved in intermolecular ligand–receptor ion–ion bonds between the attacking peptide molecules and the NaV1.8 channel molecule should fall within a certain range, the lower threshold of which is estimated to be around 9 Å. The distance values have been calculated to be below 9 Å in the inactive peptide molecules, except for Ac-RER-NH2, and in the range of 9–12 Å in the active peptide molecules.
Collapse
|
12
|
Contribution of tetrodotoxin-resistant persistent Na + currents to the excitability of C-type dural afferent neurons in rats. J Headache Pain 2022; 23:73. [PMID: 35764917 PMCID: PMC9238149 DOI: 10.1186/s10194-022-01443-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/20/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Growing evidence supports the important role of persistent sodium currents (INaP) in the neuronal excitability of various central neurons. However, the role of tetrodotoxin-resistant (TTX-R) Na+ channel-mediated INaP in the neuronal excitability of nociceptive neurons remains poorly understood. METHODS We investigated the functional role of TTX-R INaP in the excitability of C-type nociceptive dural afferent neurons, which was identified using a fluorescent dye, 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchloride (DiI), and a whole-cell patch-clamp technique. RESULTS TTX-R INaP were found in most DiI-positive neurons, but their density was proportional to neuronal size. Although the voltage dependence of TTX-R Na+ channels did not differ among DiI-positive neurons, the extent of the onset of slow inactivation, recovery from inactivation, and use-dependent inhibition of these channels was highly correlated with neuronal size and, to a great extent, the density of TTX-R INaP. In the presence of TTX, treatment with a specific INaP inhibitor, riluzole, substantially decreased the number of action potentials generated by depolarizing current injection, suggesting that TTX-R INaP are related to the excitability of dural afferent neurons. In animals treated chronically with inflammatory mediators, the density of TTX-R INaP was significantly increased, and it was difficult to inactivate TTX-R Na+ channels. CONCLUSIONS TTX-R INaP apparently contributes to the differential properties of TTX-R Na+ channels and neuronal excitability. Consequently, the selective modulation of TTX-R INaP could be, at least in part, a new approach for the treatment of migraine headaches.
Collapse
|
13
|
Wong C, Barkai O, Wang F, Thörn Pérez C, Lev S, Cai W, Tansley S, Yousefpour N, Hooshmandi M, Lister KC, Latif M, Cuello AC, Prager-Khoutorsky M, Mogil JS, Séguéla P, De Koninck Y, Ribeiro-da-Silva A, Binshtok AM, Khoutorsky A. mTORC2 mediates structural plasticity in distal nociceptive endings that contributes to pain hypersensitivity following inflammation. J Clin Invest 2022; 132:152635. [PMID: 35579957 PMCID: PMC9337825 DOI: 10.1172/jci152635] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 05/13/2022] [Indexed: 11/29/2022] Open
Abstract
The encoding of noxious stimuli into action potential firing is largely mediated by nociceptive free nerve endings. Tissue inflammation, by changing the intrinsic properties of the nociceptive endings, leads to nociceptive hyperexcitability and thus to the development of inflammatory pain. Here, we showed that tissue inflammation–induced activation of the mammalian target of rapamycin complex 2 (mTORC2) triggers changes in the architecture of nociceptive terminals and leads to inflammatory pain. Pharmacological activation of mTORC2 induced elongation and branching of nociceptor peripheral endings and caused long-lasting pain hypersensitivity. Conversely, nociceptor-specific deletion of the mTORC2 regulatory protein rapamycin-insensitive companion of mTOR (Rictor) prevented inflammation-induced elongation and branching of cutaneous nociceptive fibers and attenuated inflammatory pain hypersensitivity. Computational modeling demonstrated that mTORC2-mediated structural changes in the nociceptive terminal tree are sufficient to increase the excitability of nociceptors. Targeting mTORC2 using a single injection of antisense oligonucleotide against Rictor provided long-lasting alleviation of inflammatory pain hypersensitivity. Collectively, we showed that tissue inflammation–induced activation of mTORC2 causes structural plasticity of nociceptive free nerve endings in the epidermis and inflammatory hyperalgesia, representing a therapeutic target for inflammatory pain.
Collapse
Affiliation(s)
- Calvin Wong
- Department of Anesthesia, McGill University, Montreal, Canada
| | - Omer Barkai
- Department of Medical Neurobiology, The Hebrew University, Jerusalem, Israel
| | - Feng Wang
- Department of Psychiatry and Neuroscience, Université Laval, Quebec City, Canada
| | | | - Shaya Lev
- Department of Medical Neurobiology, The Hebrew University, Jerusalem, Israel
| | - Weihua Cai
- Department of Anesthesia, McGill University, Montreal, Canada
| | - Shannon Tansley
- Department of Psychology, McGill University, Montreal, Canada
| | - Noosha Yousefpour
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | | | - Kevin C Lister
- Department of Anesthesia, McGill University, Montreal, Canada
| | - Mariam Latif
- Department of Anesthesia, McGill University, Montreal, Canada
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | | | - Jeffrey S Mogil
- Department of Psychology, McGill University, Montreal, Canada
| | - Philippe Séguéla
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Yves De Koninck
- Department of Psychiatry and Neuroscience, Université Laval, Quebec City, Canada
| | | | | | | |
Collapse
|
14
|
Ma T, Li L, Chen R, Yang L, Sun H, Du S, Xu X, Cao Z, Zhang X, Zhang L, Shi X, Liu JY. Protein arginine methyltransferase 7 modulates neuronal excitability by interacting with NaV1.9. Pain 2022; 163:753-764. [PMID: 34326297 PMCID: PMC8929296 DOI: 10.1097/j.pain.0000000000002421] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 11/26/2022]
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.
Collapse
Affiliation(s)
- Tingbin Ma
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Lulu Li
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Rui Chen
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Luyao Yang
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Hao Sun
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Shiyue Du
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Xuan Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhijian Cao
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Xianwei Zhang
- Department of Anesthesiology, Tongji Hospital of HUST, Wuhan, China
| | - Luoying Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Xiaoliu Shi
- Department of Medical Genetics, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Yu Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
15
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
16
|
Zaytseva AK, Boitsov AS, Kostareva AA, Zhorov BS. Possible Interactions of Extracellular Loop IVP2-S6 With Voltage-Sensing Domain III in Cardiac Sodium Channel. Front Pharmacol 2021; 12:742508. [PMID: 34721031 PMCID: PMC8551724 DOI: 10.3389/fphar.2021.742508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
Motion transmission from voltage sensors to inactivation gates is an important problem in the general physiology of ion channels. In a cryo-EM structure of channel hNav1.5, residues N1736 and R1739 in the extracellular loop IVP2-S6 approach glutamates E1225 and E1295, respectively, in the voltage-sensing domain III (VSD-III). ClinVar-reported variants E1230K, E1295K, and R1739W/Q and other variants in loops IVP2-S6, IIIS1-S2, and IIIS3-S4 are associated with cardiac arrhythmias, highlighting the interface between IVP2-S6 and VSD-III as a hot spot of disease mutations. Atomic mechanisms of the channel dysfunction caused by these mutations are unknown. Here, we generated mutants E1295R, R1739E, E1295R/R1739E, and N1736R, expressed them in HEK-293T cells, and explored biophysical properties. Mutation E1295R reduced steady-state fast inactivation and enhanced steady-state slow inactivation. In contrast, mutation R1739E slightly enhanced fast inactivation and attenuated slow inactivation. Characteristics of the double mutant E1295R/R1739E were rather similar to those of the wild-type channel. Mutation N1736R attenuated slow inactivation. Molecular modeling predicted salt bridging of R1739E with the outermost lysine in the activated voltage-sensing helix IIIS4. In contrast, the loss-of-function substitution E1295R repelled R1739, thus destabilizing the activated VSD-III in agreement with our data that E1295R caused a depolarizing shift of the G-V curve. In silico deactivation of VSD-III with constraint-maintained salt bridge E1295-R1739 resulted in the following changes: 1) contacts between IIIS4 and IVS5 were switched; 2) contacts of the linker-helix IIIS4-S5 with IVS5, IVS6, and fast inactivation tripeptide IFM were modified; 3) contacts of the IFM tripeptide with helices IVS5 and IVS6 were altered; 4) mobile loop IVP2-S6 shifted helix IVP2 that contributes to the slow inactivation gate and helix IVS6 that contributes to the fast inactivation gate. The likelihood of salt bridge E1295-R1739 in deactivated VSD-III is supported by Poisson–Boltzmann calculations and state-dependent energetics of loop IVP2-S6. Taken together, our results suggest that loop IVP2-S6 is involved in motion transmission from VSD-III to the inactivation gates.
Collapse
Affiliation(s)
- Anastasia K Zaytseva
- Almazov National Medical Research Centre, St. Petersburg, Russia.,Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | | | - Anna A Kostareva
- Almazov National Medical Research Centre, St. Petersburg, Russia.,Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Boris S Zhorov
- Almazov National Medical Research Centre, St. Petersburg, Russia.,Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
17
|
Bagheri S, Haddadi R, Saki S, Kourosh-Arami M, Komaki A. The effect of sodium channels on neurological/neuronal disorders: A systematic review. Int J Dev Neurosci 2021; 81:669-685. [PMID: 34687079 DOI: 10.1002/jdn.10153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/06/2021] [Accepted: 10/19/2021] [Indexed: 12/19/2022] Open
Abstract
Neurological and neuronal disorders are associated with structural, biochemical, or electrical abnormalities in the nervous system. Many neurological diseases have not yet been discovered. Interventions used for the treatment of these disorders include avoidance measures, lifestyle changes, physiotherapy, neurorehabilitation, pain management, medication, and surgery. In the sodium channelopathies, alterations in the structure, expression, and function of voltage-gated sodium channels (VGSCs) are considered as the causes of neurological and neuronal diseases. Online databases, including Scopus, Science Direct, Google Scholar, and PubMed were assessed for studies published between 1977 and 2020 using the keywords of review, sodium channels blocker, neurological diseases, and neuronal diseases. VGSCs consist of one α subunit and two β subunits. These subunits are known to regulate the gating kinetics, functional characteristics, and localization of the ion channel. These channels are involved in cell migration, cellular connections, neuronal pathfinding, and neurite outgrowth. Through the VGSC, the action potential is triggered and propagated in the neurons. Action potentials are physiological functions and passage of impermeable ions. The electrophysiological properties of these channels and their relationship with neurological and neuronal disorders have been identified. Subunit mutations are involved in the development of diseases, such as epilepsy, multiple sclerosis, autism, and Alzheimer's disease. Accordingly, we conducted a review of the link between VGSCs and neurological and neuronal diseases. Also, novel therapeutic targets were introduced for future drug discoveries.
Collapse
Affiliation(s)
- Shokufeh Bagheri
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology, School of Pharmacy, Hamadan University of Medical Science, Hamadan, Iran
| | - Sahar Saki
- Vice-Chancellor for Research and Technology, Hamadan University of Medical Science, Hamadan, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
18
|
Pierre O, Fouchard M, Le Goux N, Buscaglia P, Leschiera R, Lewis RJ, Mignen O, Fluhr JW, Misery L, Le Garrec R. Pacific-Ciguatoxin-2 and Brevetoxin-1 Induce the Sensitization of Sensory Receptors Mediating Pain and Pruritus in Sensory Neurons. Mar Drugs 2021; 19:387. [PMID: 34356812 PMCID: PMC8306505 DOI: 10.3390/md19070387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 11/24/2022] Open
Abstract
Ciguatera fish poisoning (CFP) and neurotoxic shellfish poisoning syndromes are induced by the consumption of seafood contaminated by ciguatoxins and brevetoxins. Both toxins cause sensory symptoms such as paresthesia, cold dysesthesia and painful disorders. An intense pruritus, which may become chronic, occurs also in CFP. No curative treatment is available and the pathophysiology is not fully elucidated. Here we conducted single-cell calcium video-imaging experiments in sensory neurons from newborn rats to study in vitro the ability of Pacific-ciguatoxin-2 (P-CTX-2) and brevetoxin-1 (PbTx-1) to sensitize receptors and ion channels, (i.e., to increase the percentage of responding cells and/or the response amplitude to their pharmacological agonists). In addition, we studied the neurotrophin release in sensory neurons co-cultured with keratinocytes after exposure to P-CTX-2. Our results show that P-CTX-2 induced the sensitization of TRPA1, TRPV4, PAR2, MrgprC, MrgprA and TTX-r NaV channels in sensory neurons. P-CTX-2 increased the release of nerve growth factor and brain-derived neurotrophic factor in the co-culture supernatant, suggesting that those neurotrophins could contribute to the sensitization of the aforementioned receptors and channels. Our results suggest the potential role of sensitization of sensory receptors/ion channels in the induction or persistence of sensory disturbances in CFP syndrome.
Collapse
Affiliation(s)
- Ophélie Pierre
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Maxime Fouchard
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Nelig Le Goux
- Lymphocytes B et Autoimmunité, Faculty of Medicine and Health Sciences, University of Brest, Inserm, UMR1227, F-29200 Brest, France; (N.L.G.); (P.B.); (O.M.)
| | - Paul Buscaglia
- Lymphocytes B et Autoimmunité, Faculty of Medicine and Health Sciences, University of Brest, Inserm, UMR1227, F-29200 Brest, France; (N.L.G.); (P.B.); (O.M.)
- Department of Molecular Physiology and Biophysics, Fraternal Order of Eagle Diabetes Research Center, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Raphaël Leschiera
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Richard J. Lewis
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Olivier Mignen
- Lymphocytes B et Autoimmunité, Faculty of Medicine and Health Sciences, University of Brest, Inserm, UMR1227, F-29200 Brest, France; (N.L.G.); (P.B.); (O.M.)
| | - Joachim W. Fluhr
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
- Department of Dermatology and Allergology, Universitaetsmedizin Charité Berlin, D-10117 Berlin, Germany
| | - Laurent Misery
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Raphaële Le Garrec
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| |
Collapse
|
19
|
Goodwin G, McMahon SB. The physiological function of different voltage-gated sodium channels in pain. Nat Rev Neurosci 2021; 22:263-274. [PMID: 33782571 DOI: 10.1038/s41583-021-00444-w] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 02/01/2023]
Abstract
Evidence from human genetic pain disorders shows that voltage-gated sodium channel α-subtypes Nav1.7, Nav1.8 and Nav1.9 are important in the peripheral signalling of pain. Nav1.7 is of particular interest because individuals with Nav1.7 loss-of-function mutations are congenitally insensitive to acute and chronic pain, and there is considerable hope that phenocopying these effects with a pharmacological antagonist will produce a new class of analgesic drug. However, studies in these rare individuals do not reveal how and where voltage-gated sodium channels contribute to pain signalling, which is of critical importance for drug development. More than a decade of research utilizing rodent genetic models and pharmacological tools to study voltage-gated sodium channels in pain has begun to unravel the role of different subtypes. Here, we review the contribution of individual channel subtypes in three key physiological processes necessary for transmission of sensory information to the CNS: transduction of stimuli at peripheral nerve terminals, axonal transmission of action potentials and neurotransmitter release from central terminals. These data suggest that drugs seeking to recapitulate the analgesic effects of loss of function of Nav1.7 will need to be brain-penetrant - which most of those developed to date are not.
Collapse
Affiliation(s)
- George Goodwin
- Pain and Neurorestoration Group, King's College London, London, UK.
| | | |
Collapse
|
20
|
Sapio MR, Vazquez FA, Loydpierson AJ, Maric D, Kim JJ, LaPaglia DM, Puhl HL, Lu VB, Ikeda SR, Mannes AJ, Iadarola MJ. Comparative Analysis of Dorsal Root, Nodose and Sympathetic Ganglia for the Development of New Analgesics. Front Neurosci 2021; 14:615362. [PMID: 33424545 PMCID: PMC7793666 DOI: 10.3389/fnins.2020.615362] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Interoceptive and exteroceptive signals, and the corresponding coordinated control of internal organs and sensory functions, including pain, are received and orchestrated by multiple neurons within the peripheral, central and autonomic nervous systems. A central aim of the present report is to obtain a molecularly informed basis for analgesic drug development aimed at peripheral rather than central targets. We compare three key peripheral ganglia: nodose, sympathetic (superior cervical), and dorsal root ganglia in the rat, and focus on their molecular composition using next-gen RNA-Seq, as well as their neuroanatomy using immunocytochemistry and in situ hybridization. We obtained quantitative and anatomical assessments of transmitters, receptors, enzymes and signaling pathways mediating ganglion-specific functions. Distinct ganglionic patterns of expression were observed spanning ion channels, neurotransmitters, neuropeptides, G-protein coupled receptors (GPCRs), transporters, and biosynthetic enzymes. The relationship between ganglionic transcript levels and the corresponding protein was examined using immunohistochemistry for select, highly expressed, ganglion-specific genes. Transcriptomic analyses of spinal dorsal horn and intermediolateral cell column (IML), which form the termination of primary afferent neurons and the origin of preganglionic innervation to the SCG, respectively, disclosed pre- and post-ganglionic molecular-level circuits. These multimodal investigations provide insight into autonomic regulation, nodose transcripts related to pain and satiety, and DRG-spinal cord and IML-SCG communication. Multiple neurobiological and pharmacological contexts can be addressed, such as discriminating drug targets and predicting potential side effects, in analgesic drug development efforts directed at the peripheral nervous system.
Collapse
Affiliation(s)
- Matthew R Sapio
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Fernando A Vazquez
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Amelia J Loydpierson
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jenny J Kim
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Danielle M LaPaglia
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Henry L Puhl
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Van B Lu
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Stephen R Ikeda
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Andrew J Mannes
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Michael J Iadarola
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| |
Collapse
|
21
|
Pierre O, Fouchard M, Buscaglia P, Le Goux N, Leschiera R, Mignen O, Fluhr JW, Misery L, Le Garrec R. Calcium Increase and Substance P Release Induced by the Neurotoxin Brevetoxin-1 in Sensory Neurons: Involvement of PAR2 Activation through Both Cathepsin S and Canonical Signaling. Cells 2020; 9:E2704. [PMID: 33348659 PMCID: PMC7767211 DOI: 10.3390/cells9122704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
Red tides involving Karenia brevis expose humans to brevetoxins (PbTxs). Oral exposition triggers neurotoxic shellfish poisoning, whereas inhalation induces a respiratory syndrome and sensory disturbances. No curative treatment is available and the pathophysiology is not fully elucidated. Protease-activated receptor 2 (PAR2), cathepsin S (Cat-S) and substance P (SP) release are crucial mediators of the sensory effects of ciguatoxins (CTXs) which are PbTx analogs. This work explored the role of PAR2 and Cat-S in PbTx-1-induced sensory effects and deciphered the signaling pathway involved. We performed calcium imaging, PAR2 immunolocalization and SP release experiments in monocultured sensory neurons or co-cultured with keratinocytes treated with PbTx-1 or P-CTX-2. We demonstrated that PbTx-1-induced calcium increase and SP release involved Cat-S, PAR2 and transient receptor potential vanilloid 4 (TRPV4). The PbTx-1-induced signaling pathway included protein kinase A (PKA) and TRPV4, which are compatible with the PAR2 biased signaling induced by Cat-S. Internalization of PAR2 and protein kinase C (PKC), inositol triphosphate receptor and TRPV4 activation evoked by PbTx-1 are compatible with the PAR2 canonical signaling. Our results suggest that PbTx-1-induced sensory disturbances involve the PAR2-TRPV4 pathway. We identified PAR2, Cat-S, PKA, and PKC that are involved in TRPV4 sensitization induced by PbTx-1 in sensory neurons.
Collapse
Affiliation(s)
- Ophélie Pierre
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Maxime Fouchard
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Paul Buscaglia
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Nelig Le Goux
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Raphaël Leschiera
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Olivier Mignen
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Joachim W. Fluhr
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
- Department of Dermatology and Allergology, Universitaetsmedizin Charit Berlin, D-10117 Berlin, Germany
| | - Laurent Misery
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Raphaële Le Garrec
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| |
Collapse
|
22
|
Mini-review - Sodium channels and beyond in peripheral nerve disease: Modulation by cytokines and their effector protein kinases. Neurosci Lett 2020; 741:135446. [PMID: 33166641 DOI: 10.1016/j.neulet.2020.135446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022]
Abstract
Peripheral neuropathy is associated with enhanced activity of primary afferents which is often manifested as pain. Voltage-gated sodium channels (VGSCs) are critical for the initiation and propagation of action potentials and are thus essential for the transmission of the noxious stimuli from the periphery. Human peripheral sensory neurons express multiple VGSCs, including Nav1.7, Nav1.8, and Nav1.9 that are almost exclusively expressed in the peripheral nervous system. Distinct biophysical properties of Nav1.7, Nav1.8, and Nav1.9 underlie their differential contributions to finely tuned neuronal firing of nociceptors, and mutations in these channels have been associated with several inherited human pain disorders. Functional characterization of these mutations has provided additional insights into the role of these channels in electrogenesis in nociceptive neurons and pain sensation. Peripheral tissue damage activates an inflammatory response and triggers generation and release of inflammatory mediators, which can act through diverse signaling cascades to modulate expression and activity of ion channels including VGSCs, contributing to the development and maintenance of pathological pain conditions. In this review, we discuss signaling pathways that are activated by pro-nociceptive inflammatory mediators that regulate peripheral sodium channels, with a specific focus on direct phosphorylation of these channels by multiple protein kinases.
Collapse
|
23
|
Alsaloum M, Higerd GP, Effraim PR, Waxman SG. Status of peripheral sodium channel blockers for non-addictive pain treatment. Nat Rev Neurol 2020; 16:689-705. [PMID: 33110213 DOI: 10.1038/s41582-020-00415-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
The effective and safe treatment of pain is an unmet health-care need. Current medications used for pain management are often only partially effective, carry dose-limiting adverse effects and are potentially addictive, highlighting the need for improved therapeutic agents. Most common pain conditions originate in the periphery, where dorsal root ganglion and trigeminal ganglion neurons feed pain information into the CNS. Voltage-gated sodium (NaV) channels drive neuronal excitability and three subtypes - NaV1.7, NaV1.8 and NaV1.9 - are preferentially expressed in the peripheral nervous system, suggesting that their inhibition might treat pain while avoiding central and cardiac adverse effects. Genetic and functional studies of human pain disorders have identified NaV1.7, NaV1.8 and NaV1.9 as mediators of pain and validated them as targets for pain treatment. Consequently, multiple NaV1.7-specific and NaV1.8-specific blockers have undergone clinical trials, with others in preclinical development, and the targeting of NaV1.9, although hampered by technical constraints, might also be moving ahead. In this Review, we summarize the clinical and preclinical literature describing compounds that target peripheral NaV channels and discuss the challenges and future prospects for the field. Although the potential of peripheral NaV channel inhibition for the treatment of pain has yet to be realized, this remains a promising strategy to achieve non-addictive analgesia for multiple pain conditions.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Grant P Higerd
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - Philip R Effraim
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA. .,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA. .,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
24
|
PAR2, Keratinocytes, and Cathepsin S Mediate the Sensory Effects of Ciguatoxins Responsible for Ciguatera Poisoning. J Invest Dermatol 2020; 141:648-658.e3. [PMID: 32800876 DOI: 10.1016/j.jid.2020.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 01/14/2023]
Abstract
Ciguatera fish poisoning is caused by the consumption of fish contaminated with ciguatoxins (CTXs). The most distressing symptoms are cutaneous sensory disturbances, including cold dysesthesia and itch. CTXs are neurotoxins known to activate voltage-gated sodium channels, but no specific treatment exists. Peptidergic neurons have been critically involved in ciguatera fish poisoning sensory disturbances. Protease-activated receptor-2 (PAR2) is an itch- and pain-related G protein‒coupled receptor whose activation leads to a calcium-dependent neuropeptide release. In this study, we studied the role of voltage-gated sodium channels, PAR2, and the PAR2 agonist cathepsin S in the cytosolic calcium increase and subsequent release of the neuropeptide substance P elicited by Pacific CTX-2 (P-CTX-2) in rat sensory neurons and human epidermal keratinocytes. In sensory neurons, the P-CTX-2‒evoked calcium response was driven by voltage-gated sodium channels and PAR2-dependent mechanisms. In keratinocytes, P-CTX-2 also induced voltage-gated sodium channels and PAR2-dependent marked calcium response. In the cocultured cells, P-CTX-2 significantly increased cathepsin S activity, and cathepsin S and PAR2 antagonists almost abolished P-CTX-2‒elicited substance P release. Keratinocytes synergistically favored the induced substance P release. Our results demonstrate that the sensory effects of CTXs involve the cathepsin S-PAR2 pathway and are potentiated by their direct action on nonexcitable keratinocytes through the same pathway.
Collapse
|
25
|
Painful and painless mutations of SCN9A and SCN11A voltage-gated sodium channels. Pflugers Arch 2020; 472:865-880. [PMID: 32601768 PMCID: PMC7351857 DOI: 10.1007/s00424-020-02419-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
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.
Collapse
|
26
|
McMahon KL, Tay B, Deuis JR, Tanaka BS, Peigneur S, Jin AH, Tytgat J, Waxman SG, Dib-Hajj SD, Vetter I, Schroeder CI. Pharmacological activity and NMR solution structure of the leech peptide HSTX-I. Biochem Pharmacol 2020; 181:114082. [PMID: 32524995 DOI: 10.1016/j.bcp.2020.114082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 10/24/2022]
Abstract
The role of voltage-gated sodium (NaV) channels in pain perception is indisputable. Of particular interest as targets for the development of pain therapeutics are the tetrodotoxin-resistant isoforms NaV1.8 and NaV1.9, based on animal as well as human genetic studies linking these ion channel subtypes to the pathogenesis of pain. However, only a limited number of inhibitors selectively targeting these channels have been reported. HSTX-I is a peptide toxin identified from saliva of the leech Haemadipsa sylvestris. The native 23-residue peptide, stabilised by two disulfide bonds, has been reported to inhibit rat NaV1.8 and mouse NaV1.9 with low micromolar activity, and may therefore represent a scaffold for development of novel modulators with activity at human tetrodotoxin-resistant NaV isoforms. We synthetically produced this hydrophobic peptide in high yield using a one-pot oxidation and single step purification and determined the three-dimensional solution structure of HSTX-I using NMR solution spectroscopy. However, in our hands, the synthetic HSTX-I displayed only very modest activity at human NaV1.8 and NaV1.9, and lacked analgesic efficacy in a murine model of inflammatory pain.
Collapse
Affiliation(s)
- Kirsten L McMahon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Bryan Tay
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Brian S Tanaka
- Center for Neuroscience and Regeneration Research, New Haven, CT, United States; Department of Neurology, Yale University School of Medicine, New Haven, CT, United States; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Steve Peigneur
- Toxicology and Pharmacology, Katholieke Universiteit (KU) Leuven, Campus Gasthuisberg, Leuven, Belgium
| | - Ai-Hua Jin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jan Tytgat
- Toxicology and Pharmacology, Katholieke Universiteit (KU) Leuven, Campus Gasthuisberg, Leuven, Belgium
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, New Haven, CT, United States; Department of Neurology, Yale University School of Medicine, New Haven, CT, United States; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, New Haven, CT, United States; Department of Neurology, Yale University School of Medicine, New Haven, CT, United States; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4103, Australia.
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia; National Cancer Institute, National Institutes of Health, Frederick, MD 21702, United States.
| |
Collapse
|
27
|
Zhou Y, Cai S, Gomez K, Wijeratne EMK, Ji Y, Bellampalli SS, Luo S, Moutal A, Gunatilaka AAL, Khanna R. 1-O-Acetylgeopyxin A, a derivative of a fungal metabolite, blocks tetrodotoxin-sensitive voltage-gated sodium, calcium channels and neuronal excitability which correlates with inhibition of neuropathic pain. Mol Brain 2020; 13:73. [PMID: 32393368 PMCID: PMC7216607 DOI: 10.1186/s13041-020-00616-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/04/2020] [Indexed: 01/03/2023] Open
Abstract
Chronic pain can be the result of an underlying disease or condition, medical treatment, inflammation, or injury. The number of persons experiencing this type of pain is substantial, affecting upwards of 50 million adults in the United States. Pharmacotherapy of most of the severe chronic pain patients includes drugs such as gabapentinoids, re-uptake blockers and opioids. Unfortunately, gabapentinoids are not effective in up to two-thirds of this population and although opioids can be initially effective, their long-term use is associated with multiple side effects. Therefore, there is a great need to develop novel non-opioid alternative therapies to relieve chronic pain. For this purpose, we screened a small library of natural products and their derivatives in the search for pharmacological inhibitors of voltage-gated calcium and sodium channels, which are outstanding molecular targets due to their important roles in nociceptive pathways. We discovered that the acetylated derivative of the ent-kaurane diterpenoid, geopyxin A, 1-O-acetylgeopyxin A, blocks voltage-gated calcium and tetrodotoxin-sensitive voltage-gated sodium channels but not tetrodotoxin-resistant sodium channels in dorsal root ganglion (DRG) neurons. Consistent with inhibition of voltage-gated sodium and calcium channels, 1-O-acetylgeopyxin A reduced reduce action potential firing frequency and increased firing threshold (rheobase) in DRG neurons. Finally, we identified the potential of 1-O-acetylgeopyxin A to reverse mechanical allodynia in a preclinical rat model of HIV-induced sensory neuropathy. Dual targeting of both sodium and calcium channels may permit block of nociceptor excitability and of release of pro-nociceptive transmitters. Future studies will harness the core structure of geopyxins for the generation of antinociceptive drugs.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Clinical Laboratory, the First Hospital of Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Song Cai
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - E M Kithsiri Wijeratne
- Southwest Center for Natural Products Research, School of Natural Resources & the Environment, College of Agriculture & Life Sciences, The University of Arizona, Tucson, AZ, 85724, USA
| | - Yingshi Ji
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Shreya S Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Shizhen Luo
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - A A Leslie Gunatilaka
- Southwest Center for Natural Products Research, School of Natural Resources & the Environment, College of Agriculture & Life Sciences, The University of Arizona, Tucson, AZ, 85724, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA.
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, Tucson, AZ, 85724, USA.
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, 85724, USA.
| |
Collapse
|
28
|
Gade AR, Marx SO, Pitt GS. An interaction between the III-IV linker and CTD in NaV1.5 confers regulation of inactivation by CaM and FHF. J Gen Physiol 2020; 152:e201912434. [PMID: 31865383 PMCID: PMC7062510 DOI: 10.1085/jgp.201912434] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/25/2019] [Indexed: 12/19/2022] Open
Abstract
Voltage gated sodium channel (VGSC) activation drives the action potential upstroke in cardiac myocytes, skeletal muscles, and neurons. After opening, VGSCs rapidly enter a non-conducting, inactivated state. Impaired inactivation causes persistent inward current and underlies cardiac arrhythmias. VGSC auxiliary proteins calmodulin (CaM) and fibroblast growth factor homologous factors (FHFs) bind to the channel's C-terminal domain (CTD) and limit pathogenic persistent currents. The structural details and mechanisms mediating these effects are not clear. Building on recently published cryo-EM structures, we show that CaM and FHF limit persistent currents in the cardiac NaV1.5 VGSC by stabilizing an interaction between the channel's CTD and III-IV linker region. Perturbation of this intramolecular interaction increases persistent current and shifts the voltage dependence of steady-state inactivation. Interestingly, the NaV1.5 residues involved in the interaction are sites mutated in the arrhythmogenic long QT3 syndrome (LQT3). Along with electrophysiological investigations of this interaction, we present structural models that suggest how CaM and FHF stabilize the interaction and thereby limit the persistent current. The critical residues at the interaction site are conserved among VGSC isoforms, and subtle substitutions provide an explanation for differences in inactivation among the isoforms.
Collapse
Affiliation(s)
- Aravind R. Gade
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY
| | - Steven O. Marx
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Pharmacology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY
| |
Collapse
|
29
|
Adachi K, Yamada T, Ishizuka H, Oki M, Tsunogae S, Shimada N, Chiba O, Orihara T, Hidaka M, Hirokawa T, Odagi M, Konoki K, Yotsu‐Yamashita M, Nagasawa K. Synthesis of C12‐Keto Saxitoxin Derivatives with Unusual Inhibitory Activity Against Voltage‐Gated Sodium Channels. Chemistry 2020; 26:2025-2033. [DOI: 10.1002/chem.201904184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/04/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Kanna Adachi
- Department of Biotechnology and Life Science Faculty of Technology Tokyo University of Agriculture and Technology 2-24-16 Naka-cho Koganei Tokyo 184-8588 Japan
| | - Tomoshi Yamada
- Graduate School of Agriculture Science Tohoku University 468-1 Aramaki-Aza-Aoba, Aoba-ku Sendai 980-8572 Japan
| | - Hayate Ishizuka
- Department of Biotechnology and Life Science Faculty of Technology Tokyo University of Agriculture and Technology 2-24-16 Naka-cho Koganei Tokyo 184-8588 Japan
| | - Mana Oki
- Department of Biotechnology and Life Science Faculty of Technology Tokyo University of Agriculture and Technology 2-24-16 Naka-cho Koganei Tokyo 184-8588 Japan
| | - Shunsuke Tsunogae
- Graduate School of Agriculture Science Tohoku University 468-1 Aramaki-Aza-Aoba, Aoba-ku Sendai 980-8572 Japan
| | - Noriko Shimada
- Graduate School of Agriculture Science Tohoku University 468-1 Aramaki-Aza-Aoba, Aoba-ku Sendai 980-8572 Japan
| | - Osamu Chiba
- Graduate School of Agriculture Science Tohoku University 468-1 Aramaki-Aza-Aoba, Aoba-ku Sendai 980-8572 Japan
| | - Tatsuya Orihara
- Department of Biotechnology and Life Science Faculty of Technology Tokyo University of Agriculture and Technology 2-24-16 Naka-cho Koganei Tokyo 184-8588 Japan
| | - Masafumi Hidaka
- Graduate School of Agriculture Science Tohoku University 468-1 Aramaki-Aza-Aoba, Aoba-ku Sendai 980-8572 Japan
| | - Takatsugu Hirokawa
- Transborder Medical Research Center University of Tsukuba 1-1-1 Tennodai Tsukuba 305-8575 Japan
- Division of Biomedical Science University of Tsukuba 1-1-1 Tennodai Tsukuba 305-8575 Japan
- Molecular Profiling Research Center for Drug Discovery National Institute of Advanced Industrial Science and Technology 2-4-7 Aomi, Koto-ward Tokyo 135-0064 Japan
| | - Minami Odagi
- Department of Biotechnology and Life Science Faculty of Technology Tokyo University of Agriculture and Technology 2-24-16 Naka-cho Koganei Tokyo 184-8588 Japan
| | - Keiichi Konoki
- Graduate School of Agriculture Science Tohoku University 468-1 Aramaki-Aza-Aoba, Aoba-ku Sendai 980-8572 Japan
| | - Mari Yotsu‐Yamashita
- Graduate School of Agriculture Science Tohoku University 468-1 Aramaki-Aza-Aoba, Aoba-ku Sendai 980-8572 Japan
| | - Kazuo Nagasawa
- Department of Biotechnology and Life Science Faculty of Technology Tokyo University of Agriculture and Technology 2-24-16 Naka-cho Koganei Tokyo 184-8588 Japan
| |
Collapse
|
30
|
Synthetic Approaches to Zetekitoxin AB, a Potent Voltage-Gated Sodium Channel Inhibitor. Mar Drugs 2019; 18:md18010024. [PMID: 31888062 PMCID: PMC7024329 DOI: 10.3390/md18010024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are membrane proteins that are involved in the generation and propagation of action potentials in neurons. Recently, the structure of a complex made of a tetrodotoxin-sensitive (TTX-s) NaV subtype with saxitoxin (STX), a shellfish toxin, was determined. STX potently inhibits TTX-s NaV, and is used as a biological tool to investigate the function of NaVs. More than 50 analogs of STX have been isolated from nature. Among them, zetekitoxin AB (ZTX) has a distinctive chemical structure, and is the most potent inhibitor of NaVs, including tetrodotoxin-resistant (TTX-r) NaV. Despite intensive synthetic studies, total synthesis of ZTX has not yet been achieved. Here, we review recent efforts directed toward the total synthesis of ZTX, including syntheses of 11-saxitoxinethanoic acid (SEA), which is considered a useful synthetic model for ZTX, since it contains a key carbon-carbon bond at the C11 position.
Collapse
|
31
|
Hamed SA, Oseily AM. Peripheral and central auditory function in adults with epilepsy and treated with carbamazepine. HEARING, BALANCE AND COMMUNICATION 2019. [DOI: 10.1080/21695717.2019.1630975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Sherifa A. Hamed
- Department of Neurology and Psychiatry, Assiut University Hospital, Assiut, Egypt
| | - Amira M. Oseily
- Department of ENT, Audiology Unit, Assiut University Hospital, Assiut, Egypt
| |
Collapse
|
32
|
Maladaptive activation of Nav1.9 channels by nitric oxide causes triptan-induced medication overuse headache. Nat Commun 2019; 10:4253. [PMID: 31534133 PMCID: PMC6751217 DOI: 10.1038/s41467-019-12197-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 08/26/2019] [Indexed: 01/03/2023] Open
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.
Collapse
|
33
|
Voinsky I, McCarthy MJ, Shekhtman T, Kelsoe JR, Gurwitz D. SCN11A mRNA levels in female bipolar disorder PBMCs as tentative biomarker for distinct patient sub-phenotypes. Drug Dev Res 2019; 80:1128-1135. [PMID: 31498915 DOI: 10.1002/ddr.21598] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 01/12/2023]
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 NaV 1.9 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 NaV 1.9 voltage-gated sodium channel thus deserves consideration as a tentative phenotype modifier in BD.
Collapse
Affiliation(s)
- Irena Voinsky
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Michael J McCarthy
- Department of Psychiatry, University of California San Diego, La Jolla, California.,VA San Diego Healthcare System, San Diego, California
| | - Tatyana Shekhtman
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - John R Kelsoe
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
34
|
Lee J, Kim S, Kim HM, Kim HJ, Yu FH. NaV1.6 and NaV1.7 channels are major endogenous voltage-gated sodium channels in ND7/23 cells. PLoS One 2019; 14:e0221156. [PMID: 31419255 PMCID: PMC6697327 DOI: 10.1371/journal.pone.0221156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/31/2019] [Indexed: 01/02/2023] Open
Abstract
ND7/23 cells are gaining traction as a host model to express peripheral sodium channels such as NaV1.8 and NaV1.9 that have been difficult to express in widely utilized heterologous cells, like CHO and HEK293. Use of ND7/23 as a model cell to characterize the properties of sodium channels requires clear understanding of the endogenous ion channels. To define the nature of the background sodium currents in ND7/23 cells, we aimed to comprehensively profile the voltage-gated sodium channel subunits by endpoint and quantitative reverse transcription-PCR and by whole-cell patch clamp electrophysiology. We found that untransfected ND7/23 cells express endogenous peak sodium currents that average -2.12nA (n = 15) and with kinetics typical of fast sodium currents having activation and inactivation completed within few milliseconds. Furthermore, sodium currents were reduced to virtually nil upon exposure to 100nM tetrodotoxin, indicating that ND7/23 cells have essentially null background for tetrodotoxin-resistant (TTX-R) currents. qRT-PCR profiling indicated a major expression of TTX-sensitive (TTX-S) NaV1.6 and NaV1.7 at similar levels and very low expression of TTX-R NaV1.9 transcripts. There was no expression of TTX-R NaV1.8 in ND7/23 cells. There was low expression of NaV1.1, NaV1.2, NaV1.3 and no expression of cardiac or skeletal muscle sodium channels. As for the sodium channel auxiliary subunits, β1 and β3 subunits were expressed, but not the β2 and β4 subunits that covalently associate with the α-subunits. In addition, our results also showed that only the mouse forms of NaV1.6, NaV1.7 and NaV1.9 sodium channels were expressed in ND7/23 cells that was originally generated as a hybridoma of rat embryonic DRG and mouse neuroblastoma cell-line. By molecular profiling of auxiliary β- and principal α-subunits of the voltage gated sodium channel complex, our results define the background sodium channels expressed in ND7/23 cells, and confirm their utility for detailed functional studies of emerging pain channelopathies ascribed to mutations of the TTX-R sodium channels of sensory neurons.
Collapse
Affiliation(s)
- Jisoo Lee
- Department of Pharmacology and Dental Therapeutics, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Shinae Kim
- Department of Pharmacology and Dental Therapeutics, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Hye-mi Kim
- Department of Pharmacology and Dental Therapeutics, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Hyun Jeong Kim
- Department of Dental Anesthesiology, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Frank H. Yu
- Department of Pharmacology and Dental Therapeutics, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
35
|
Lee K, Lee BM, Park CK, Kim YH, Chung G. Ion Channels Involved in Tooth Pain. Int J Mol Sci 2019; 20:ijms20092266. [PMID: 31071917 PMCID: PMC6539952 DOI: 10.3390/ijms20092266] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/03/2019] [Accepted: 05/03/2019] [Indexed: 01/05/2023] Open
Abstract
The tooth has an unusual sensory system that converts external stimuli predominantly into pain, yet its sensory afferents in teeth demonstrate cytochemical properties of non-nociceptive neurons. This review summarizes the recent knowledge underlying this paradoxical nociception, with a focus on the ion channels involved in tooth pain. The expression of temperature-sensitive ion channels has been extensively investigated because thermal stimulation often evokes tooth pain. However, temperature-sensitive ion channels cannot explain the sudden intense tooth pain evoked by innocuous temperatures or light air puffs, leading to the hydrodynamic theory emphasizing the microfluidic movement within the dentinal tubules for detection by mechanosensitive ion channels. Several mechanosensitive ion channels expressed in dental sensory systems have been suggested as key players in the hydrodynamic theory, and TRPM7, which is abundant in the odontoblasts, and recently discovered PIEZO receptors are promising candidates. Several ligand-gated ion channels and voltage-gated ion channels expressed in dental primary afferent neurons have been discussed in relation to their potential contribution to tooth pain. In addition, in recent years, there has been growing interest in the potential sensory role of odontoblasts; thus, the expression of ion channels in odontoblasts and their potential relation to tooth pain is also reviewed.
Collapse
Affiliation(s)
- Kihwan Lee
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 406-799, Korea.
| | - Byeong-Min Lee
- Department of Oral Physiology and Program in Neurobiology, School of Dentistry, Seoul National University, Seoul 08826, Korea.
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 406-799, Korea.
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 406-799, Korea.
| | - Gehoon Chung
- Department of Oral Physiology and Program in Neurobiology, School of Dentistry, Seoul National University, Seoul 08826, Korea.
- Dental Research Institute, Seoul National University, Seoul 03080, Korea.
| |
Collapse
|
36
|
Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD. The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev 2019; 99:1079-1151. [DOI: 10.1152/physrev.00052.2017] [Citation(s) in RCA: 256] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute pain signaling has a key protective role and is highly evolutionarily conserved. Chronic pain, however, is maladaptive, occurring as a consequence of injury and disease, and is associated with sensitization of the somatosensory nervous system. Primary sensory neurons are involved in both of these processes, and the recent advances in understanding sensory transduction and human genetics are the focus of this review. Voltage-gated sodium channels (VGSCs) are important determinants of sensory neuron excitability: they are essential for the initial transduction of sensory stimuli, the electrogenesis of the action potential, and neurotransmitter release from sensory neuron terminals. Nav1.1, Nav1.6, Nav1.7, Nav1.8, and Nav1.9 are all expressed by adult sensory neurons. The biophysical characteristics of these channels, as well as their unique expression patterns within subtypes of sensory neurons, define their functional role in pain signaling. Changes in the expression of VGSCs, as well as posttranslational modifications, contribute to the sensitization of sensory neurons in chronic pain states. Furthermore, gene variants in Nav1.7, Nav1.8, and Nav1.9 have now been linked to human Mendelian pain disorders and more recently to common pain disorders such as small-fiber neuropathy. Chronic pain affects one in five of the general population. Given the poor efficacy of current analgesics, the selective expression of particular VGSCs in sensory neurons makes these attractive targets for drug discovery. The increasing availability of gene sequencing, combined with structural modeling and electrophysiological analysis of gene variants, also provides the opportunity to better target existing therapies in a personalized manner.
Collapse
Affiliation(s)
- David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Alex J. Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jianying Huang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G. Waxman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Sulayman D. Dib-Hajj
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
37
|
Cai S, Bellampalli SS, Yu J, Li W, Ji Y, Wijeratne EMK, Dorame A, Luo S, Shan Z, Khanna M, Moutal A, Streicher JM, Gunatilaka AAL, Khanna R. (-)-Hardwickiic Acid and Hautriwaic Acid Induce Antinociception via Blockade of Tetrodotoxin-Sensitive Voltage-Dependent Sodium Channels. ACS Chem Neurosci 2019; 10:1716-1728. [PMID: 30525440 DOI: 10.1021/acschemneuro.8b00617] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
For an affliction that debilitates an estimated 50 million adults in the United States, the current chronic pain management approaches are inadequate. The Centers for Disease Control and Prevention have called for a minimization in opioid prescription and use for chronic pain conditions, and thus, it is imperative to discover alternative non-opioid based strategies. For the realization of this call, a library of natural products was screened in search of pharmacological inhibitors of both voltage-gated calcium channels and voltage-gated sodium channels, which are excellent targets due to their well-established roles in nociceptive pathways. We discovered (-)-hardwickiic acid ((-)-HDA) and hautriwaic acid (HTA) isolated from plants, Croton californicus and Eremocarpus setigerus, respectively, inhibited tetrodotoxin-sensitive sodium, but not calcium or potassium, channels in small diameter, presumptively nociceptive, dorsal root ganglion (DRG) neurons. Failure to inhibit spontaneous postsynaptic excitatory currents indicated a preferential targeting of voltage-gated sodium channels over voltage-gated calcium channels by these extracts. Neither compound was a ligand at opioid receptors. Finally, we identified the potential of both (-)-HDA and HTA to reverse chronic pain behavior in preclinical rat models of HIV-sensory neuropathy, and for (-)-HDA specifically, in chemotherapy-induced peripheral neuropathy. Our results illustrate the therapeutic potential for (-)-HDA and HTA for chronic pain management and could represent a scaffold, that, if optimized by structure-activity relationship studies, may yield novel specific sodium channel antagonists for pain relief.
Collapse
Affiliation(s)
| | | | - Jie Yu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310058, P.R. China
| | | | - Yingshi Ji
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | | | | | | | - Zhiming Shan
- Department of Anesthesiology, Shenzhen People’s Hospital & Second Clinical Medical College of Jinan University, Shenzhen 518020, P.R. China
| | - May Khanna
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, United States
| | | | | | | | - Rajesh Khanna
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, United States
| |
Collapse
|
38
|
Chew LA, Bellampalli SS, Dustrude ET, Khanna R. Mining the Na v1.7 interactome: Opportunities for chronic pain therapeutics. Biochem Pharmacol 2019; 163:9-20. [PMID: 30699328 DOI: 10.1016/j.bcp.2019.01.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/24/2019] [Indexed: 12/14/2022]
Abstract
The peripherally expressed voltage-gated sodium NaV1.7 (gene SCN9A) channel boosts small stimuli to initiate firing of pain-signaling dorsal root ganglia (DRG) neurons and facilitates neurotransmitter release at the first synapse within the spinal cord. Mutations in SCN9A produce distinct human pain syndromes. Widely acknowledged as a "gatekeeper" of pain, NaV1.7 has been the focus of intense investigation but, to date, no NaV1.7-selective drugs have reached the clinic. Elegant crystallographic studies have demonstrated the potential of designing highly potent and selective NaV1.7 compounds but their therapeutic value remains untested. Transcriptional silencing of NaV1.7 by a naturally expressed antisense transcript has been reported in rodents and humans but whether this represents a viable opportunity for designing NaV1.7 therapeutics is currently unknown. The demonstration that loss of NaV1.7 function is associated with upregulation of endogenous opioids and potentiation of mu- and delta-opioid receptor activities, suggests that targeting only NaV1.7 may be insufficient for analgesia. However, the link between opioid-dependent analgesic mechanisms and function of sodium channels and intracellular sodium-dependent signaling remains controversial. Thus, additional new targets - regulators, modulators - are needed. In this context, we mine the literature for the known interactome of NaV1.7 with a focus on protein interactors that affect the channel's trafficking or link it to opioid signaling. As a case study, we present antinociceptive evidence of allosteric regulation of NaV1.7 by the cytosolic collapsin response mediator protein 2 (CRMP2). Throughout discussions of these possible new targets, we offer thoughts on the therapeutic implications of modulating NaV1.7 function in chronic pain.
Collapse
Affiliation(s)
- Lindsey A Chew
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Shreya S Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Erik T Dustrude
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Neuroscience, College of Medicine, University of Arizona, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ 85724, USA.
| |
Collapse
|
39
|
Hoffstaetter LJ, Mastrotto M, Merriman DK, Dib-Hajj SD, Waxman SG, Bagriantsev SN, Gracheva EO. Somatosensory Neurons Enter a State of Altered Excitability during Hibernation. Curr Biol 2018; 28:2998-3004.e3. [PMID: 30174191 DOI: 10.1016/j.cub.2018.07.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 06/28/2018] [Accepted: 07/06/2018] [Indexed: 12/19/2022]
Abstract
Hibernation in mammals involves prolonged periods of inactivity, hypothermia, hypometabolism, and decreased somatosensation. Peripheral somatosensory neurons play an essential role in the detection and transmission of sensory information to CNS and in the generation of adaptive responses. During hibernation, when body temperature drops to as low as 2°C, animals dramatically reduce their sensitivity to physical cues [1, 2]. It is well established that, in non-hibernators, cold exposure suppresses energy production, leading to dissipation of the ionic and electrical gradients across the plasma membrane and, in the case of neurons, inhibiting the generation of action potentials [3]. Conceivably, such cold-induced elimination of electrogenesis could be part of a general mechanism that inhibits sensory abilities in hibernators. However, when hibernators become active, the bodily functions-including the ability to sense environmental cues-return to normal within hours, suggesting the existence of mechanisms supporting basal functionality of cells during torpor and rapid restoration of activity upon arousal. We tested this by comparing properties of somatosensory neurons from active and torpid thirteen-lined ground squirrels (Ictidomys tridecemlineatus). We found that torpid neurons can compensate for cold-induced functional deficits, resulting in unaltered resting potential, input resistance, and rheobase. Torpid neurons can generate action potentials but manifest markedly altered firing patterns, partially due to decreased activity of voltage-gated sodium channels. Our results provide insights into the mechanism that preserves somatosensory neurons in a semi-active state, enabling fast restoration of sensory function upon arousal. These findings contribute to the development of strategies enabling therapeutic hypothermia and hypometabolism.
Collapse
Affiliation(s)
- Lydia J Hoffstaetter
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Marco Mastrotto
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Dana K Merriman
- Department of Biology, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI 54901, USA
| | - Sulayman D Dib-Hajj
- The Center for Neuroscience and Regeneration Research and Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Center for Restoration of Nervous System Function, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- The Center for Neuroscience and Regeneration Research and Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA; Center for Restoration of Nervous System Function, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA.
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA.
| |
Collapse
|
40
|
Touska F, Turnquist B, Vlachova V, Reeh PW, Leffler A, Zimmermann K. Heat-resistant action potentials require TTX-resistant sodium channels Na V1.8 and Na V1.9. J Gen Physiol 2018; 150:1125-1144. [PMID: 29970412 PMCID: PMC6080895 DOI: 10.1085/jgp.201711786] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 12/31/2017] [Accepted: 05/29/2018] [Indexed: 12/13/2022] Open
Abstract
Nociceptors prevent damage by being able to detect and transmit noxious stimuli, such as hot temperatures. Touska et al. show that the TTX-resistant NaV channels, NaV1.8 and NaV1.9, are required for heat-resistant nociceptors to encode noxious heat and that the current through NaV1.9 increases at higher temperatures. 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.
Collapse
Affiliation(s)
- Filip Touska
- Klinik für Anästhesiologie am Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Cellular Neurophysiology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Brian Turnquist
- Department of Mathematics and Computer Science, Bethel University, St. Paul, MN
| | - Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Peter W Reeh
- Department of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Leffler
- Klinik für Anästhesiologie und Intensivmedizin, Medizinische Hochschule Hannover, Hannover, Germany
| | - Katharina Zimmermann
- Klinik für Anästhesiologie am Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
41
|
Kondo C, Clark RB, Al‐Jezani N, Kim TY, Belke D, Banderali U, Szerencsei RT, Jalloul AH, Schnetkamp PPM, Spitzer KW, Giles WR. ATP triggers a robust intracellular [Ca 2+ ]-mediated signalling pathway in human synovial fibroblasts. Exp Physiol 2018; 103:1101-1122. [PMID: 29791754 DOI: 10.1113/ep086851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/26/2018] [Indexed: 01/05/2023]
Abstract
NEW FINDINGS What is the central question of this study? What are the main [Ca2+ ]i signalling pathways activated by ATP in human synovial fibroblasts? What is the main finding and its importance? In human synovial fibroblasts ATP acts through a linked G-protein (Gq ) and phospholipase C signalling mechanism to produce IP3 , which then markedly enhances release of Ca2+ from the endoplasmic reticulum. These results provide new information for the detection of early pathophysiology of arthritis. ABSTRACT In human articular joints, synovial fibroblasts (HSFs) have essential physiological functions that include synthesis and secretion of components of the extracellular matrix and essential articular joint lubricants, as well as release of paracrine substances such as ATP. Although the molecular and cellular processes that lead to a rheumatoid arthritis (RA) phenotype are not fully understood, HSF cells exhibit significant changes during this disease progression. The effects of ATP on HSFs were studied by monitoring changes in intracellular Ca2+ ([Ca2+ ]i ), and measuring electrophysiological properties. ATP application to HSF cell populations that had been enzymatically released from 2-D cell culture revealed that ATP (10-100 μm), or its analogues UTP or ADP, consistently produced a large transient increase in [Ca2+ ]i . These changes (i) were initiated by activation of the P2 Y purinergic receptor family, (ii) required Gq -mediated signal transduction, (iii) did not involve a transmembrane Ca2+ influx, but instead (iv) arose almost entirely from activation of endoplasmic reticulum (ER)-localized inositol 1,4,5-trisphosphate (IP3 ) receptors that triggered Ca2+ release from the ER. Corresponding single cell electrophysiological studies revealed that these ATP effects (i) were insensitive to [Ca2+ ]o removal, (ii) involved an IP3 -mediated intracellular Ca2+ release process, and (iii) strongly turned on Ca2+ -activated K+ current(s) that significantly hyperpolarized these cells. Application of histamine produced very similar effects in these HSF cells. Since ATP is a known paracrine agonist and histamine is released early in the inflammatory response, these findings may contribute to identification of early steps/defects in the initiation and progression of RA.
Collapse
Affiliation(s)
- C Kondo
- Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | - R B Clark
- Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | | | - T Y Kim
- Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | - D Belke
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | | | - R T Szerencsei
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - A H Jalloul
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - P P M Schnetkamp
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - K W Spitzer
- Nora Eccles Harrison Cardiovascular Centre, Salt Lake City, UT, USA
| | - W R Giles
- Faculty of Kinesiology, University of Calgary, Calgary, Canada.,Faculty of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
42
|
Marler TL, Wright AB, Elmslie KL, Heier AK, Remily E, Kim-Han JS, Ramachandra R, Elmslie KS. Na V1.9 channels in muscle afferent neurons and axons. J Neurophysiol 2018; 120:1032-1044. [PMID: 29847236 DOI: 10.1152/jn.00573.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
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 NaV current 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.
Collapse
Affiliation(s)
- Tyler L Marler
- The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A. T. Still University of Health Sciences, Kirksville, Missouri
| | - Andrew B Wright
- The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A. T. Still University of Health Sciences, Kirksville, Missouri
| | - Kristina L Elmslie
- The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A. T. Still University of Health Sciences, Kirksville, Missouri
| | - Ankeeta K Heier
- The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A. T. Still University of Health Sciences, Kirksville, Missouri
| | - Ethan Remily
- The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A. T. Still University of Health Sciences, Kirksville, Missouri
| | - Jeong Sook Kim-Han
- The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A. T. Still University of Health Sciences, Kirksville, Missouri
| | - Renuka Ramachandra
- The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A. T. Still University of Health Sciences, Kirksville, Missouri
| | - Keith S Elmslie
- The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A. T. Still University of Health Sciences, Kirksville, Missouri
| |
Collapse
|
43
|
Behavioral, cellular and molecular maladaptations covary with exposure to pyridostigmine bromide in a rat model of gulf war illness pain. Toxicol Appl Pharmacol 2018; 352:119-131. [PMID: 29803855 DOI: 10.1016/j.taap.2018.05.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/16/2018] [Accepted: 05/20/2018] [Indexed: 12/12/2022]
Abstract
Many veterans of Operation Desert Storm (ODS) struggle with the chronic pain of Gulf War Illness (GWI). Exposure to insecticides and pyridostigmine bromide (PB) have been implicated in the etiology of this multisymptom disease. We examined the influence of 3 (DEET (N,N-diethyl-meta-toluamide), permethrin, chlorpyrifos) or 4 GW agents (DEET, permethrin, chlorpyrifos, pyridostigmine bromide (PB)) on the post-exposure ambulatory and resting behaviors of rats. In three independent studies, rats that were exposed to all 4 agents consistently developed both immediate and delayed ambulatory deficits that persisted at least 16 weeks after exposures had ceased. Rats exposed to a 3 agent protocol (PB excluded) did not develop any ambulatory deficits. Cellular and molecular studies on nociceptors harvested from 16WP (weeks post-exposure) rats indicated that vascular nociceptor Nav1.9 mediated currents were chronically potentiated following the 4 agent protocol but not following the 3 agent protocol. Muscarinic linkages to muscle nociceptor TRPA1 were also potentiated in the 4 agent but not the 3 agent, PB excluded, protocol. Although Kv7 activity changes diverged from the behavioral data, a Kv7 opener, retigabine, transiently reversed ambulation deficits. We concluded that PB played a critical role in the development of pain-like signs in a GWI rat model and that shifts in Nav1.9 and TRPA1 activity were critical to the expression of these pain behaviors.
Collapse
|
44
|
Hoffstaetter LJ, Bagriantsev SN, Gracheva EO. TRPs et al.: a molecular toolkit for thermosensory adaptations. Pflugers Arch 2018; 470:745-759. [PMID: 29484488 PMCID: PMC5945325 DOI: 10.1007/s00424-018-2120-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/03/2018] [Accepted: 02/05/2018] [Indexed: 12/19/2022]
Abstract
The ability to sense temperature is crucial for the survival of an organism. Temperature influences all biological operations, from rates of metabolic reactions to protein folding, and broad behavioral functions, from feeding to breeding, and other seasonal activities. The evolution of specialized thermosensory adaptations has enabled animals to inhabit extreme temperature niches and to perform specific temperature-dependent behaviors. The function of sensory neurons depends on the participation of various types of ion channels. Each of the channels involved in neuronal excitability, whether through the generation of receptor potential, action potential, or the maintenance of the resting potential have temperature-dependent properties that can tune the neuron's response to temperature stimuli. Since the function of all proteins is affected by temperature, animals need adaptations not only for detecting different temperatures, but also for maintaining sensory ability at different temperatures. A full understanding of the molecular mechanism of thermosensation requires an investigation of all channel types at each step of thermosensory transduction. A fruitful avenue of investigation into how different molecules can contribute to the fine-tuning of temperature sensitivity is to study the specialized adaptations of various species. Given the diversity of molecular participants at each stage of sensory transduction, animals have a toolkit of channels at their disposal to adapt their thermosensitivity to their particular habitats or behavioral circumstances.
Collapse
Affiliation(s)
- Lydia J Hoffstaetter
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
| |
Collapse
|
45
|
Foadi N. Modulation of sodium channels as pharmacological tool for pain therapy-highlights and gaps. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:481-488. [PMID: 29572558 DOI: 10.1007/s00210-018-1487-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023]
Abstract
Voltage-gated sodium channels are crucially involved in the transduction and transmission of nociceptive signals and pathological pain states. In the past decades, a lot of effort has been spent examining and characterizing biophysical properties of the different sodium channels and their role in signaling pathways. Several gains of function mutations of the sodium channels Nav1.7, Nav1.8, and Nav1.9 are associated with pain disorders. Due to their critical role in nociceptive pathways voltage-gated sodium channels are regarded interesting targets for pharmacological pain treatment. However we still need to fill the gap that exists in the translation of efficacy in preclinical in vitro experiments and in models of pain into the clinic. This review summarizes biological and electrophysiological properties of voltage-gated sodium channels and aims to discuss limitations and promising pharmacological strategies in sodium channel research in the context of pain therapy.
Collapse
Affiliation(s)
- Nilufar Foadi
- Clinic for Anaesthesia and Critical Care Medicine, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
46
|
Wang G, Long C, Liu W, Xu C, Zhang M, Li Q, Lu Q, Meng P, Li D, Rong M, Sun Z, Luo X, Lai R. Novel Sodium Channel Inhibitor From Leeches. Front Pharmacol 2018; 9:186. [PMID: 29559913 PMCID: PMC5845541 DOI: 10.3389/fphar.2018.00186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/19/2018] [Indexed: 12/16/2022] Open
Abstract
Considering blood-sucking habits of leeches from surviving strategy of view, it can be hypothesized that leech saliva has analgesia or anesthesia functions for leeches to stay undetected by the host. However, no specific substance with analgesic function has been reported from leech saliva although clinical applications strongly indicated that leech therapy produces a strong and long lasting pain-reducing effect. Herein, a novel family of small peptides (HSTXs) including 11 members which show low similarity with known peptides was identified from salivary glands of the leech Haemadipsa sylvestris. A typical HSTX is composed of 22-25 amino acid residues including four half-cysteines, forming two intra-molecular disulfide bridges, and an amidated C-terminus. HSTX-I exerts significant analgesic function by specifically inhibiting voltage-gated sodium (NaV) channels (NaV1.8 and NaV1.9) which contribute to action potential electrogenesis in neurons and potential targets to develop analgesics. This study reveals that sodium channel inhibitors are analgesic substances in the leech. HSTXs are excellent candidates or templates for development of analgesics.
Collapse
Affiliation(s)
- Gan Wang
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China
| | - Chengbo Long
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China
| | - Weihui Liu
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China
| | - Cheng Xu
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China
| | - Min Zhang
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China.,Graduate School of University of Chinese Academy of Sciences, Beijing, China
| | - Qiong Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Qiumin Lu
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China.,Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, China
| | - Ping Meng
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China
| | - Dongsheng Li
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China
| | - Mingqiang Rong
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China.,Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, China
| | - Zhaohui Sun
- Department of Clinical Laboratory, Guangzhou General Hospital of Guangzhou Military Command of PLA, Guangzhou, China
| | - Xiaodong Luo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Ren Lai
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, China.,Life Sciences College of Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
47
|
Erickson A, Deiteren A, Harrington AM, Garcia‐Caraballo S, Castro J, Caldwell A, Grundy L, Brierley SM. Voltage-gated sodium channels: (Na V )igating the field to determine their contribution to visceral nociception. J Physiol 2018; 596:785-807. [PMID: 29318638 PMCID: PMC5830430 DOI: 10.1113/jp273461] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic visceral pain, altered motility and bladder dysfunction are common, yet poorly managed symptoms of functional and inflammatory disorders of the gastrointestinal and urinary tracts. Recently, numerous human channelopathies of the voltage-gated sodium (NaV ) channel family have been identified, which induce either painful neuropathies, an insensitivity to pain, or alterations in smooth muscle function. The identification of these disorders, in addition to the recent utilisation of genetically modified NaV mice and specific NaV channel modulators, has shed new light on how NaV channels contribute to the function of neuronal and non-neuronal tissues within the gastrointestinal tract and bladder. Here we review the current pre-clinical and clinical evidence to reveal how the nine NaV channel family members (NaV 1.1-NaV 1.9) contribute to abdominal visceral function in normal and disease states.
Collapse
Affiliation(s)
- Andelain Erickson
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Annemie Deiteren
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Andrea M. Harrington
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Sonia Garcia‐Caraballo
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Joel Castro
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Ashlee Caldwell
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Luke Grundy
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Stuart M. Brierley
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| |
Collapse
|
48
|
Wu Y, Ma H, Zhang F, Zhang C, Zou X, Cao Z. Selective Voltage-Gated Sodium Channel Peptide Toxins from Animal Venom: Pharmacological Probes and Analgesic Drug Development. ACS Chem Neurosci 2018; 9:187-197. [PMID: 29161016 DOI: 10.1021/acschemneuro.7b00406] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Navs) play critical roles in action potential generation and propagation. Nav channelopathy as well as pathological sensitization contribute to allodynia and hyperalgesia. Recent evidence has demonstrated the significant roles of Nav subtypes (Nav1.3, 1.7, 1.8, and 1.9) in nociceptive transduction, and therefore these Navs may represent attractive targets for analgesic drug discovery. Animal toxins are structurally diverse peptides that are highly potent yet selective on ion channel subtypes and therefore represent valuable probes to elucidate the structures, gating properties, and cellular functions of ion channels. In this review, we summarize recent advances on peptide toxins from animal venom that selectively target Nav1.3, 1.7, 1.8, and 1.9, along with their potential in analgesic drug discovery.
Collapse
Affiliation(s)
- Ying Wu
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Hui Ma
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Fan Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chunlei Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaohan Zou
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Zhengyu Cao
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
49
|
Zhou X, Xiao Z, Xu Y, Zhang Y, Tang D, Wu X, Tang C, Chen M, Shi X, Chen P, Liang S, Liu Z. Electrophysiological and Pharmacological Analyses of Na v1.9 Voltage-Gated Sodium Channel by Establishing a Heterologous Expression System. Front Pharmacol 2017; 8:852. [PMID: 29213238 PMCID: PMC5702848 DOI: 10.3389/fphar.2017.00852] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/07/2017] [Indexed: 11/13/2022] Open
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.
Collapse
Affiliation(s)
- Xi Zhou
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zhen Xiao
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yan Xu
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yunxiao Zhang
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Dongfang Tang
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xinzhou Wu
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Cheng Tang
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.,Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Minzhi Chen
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.,Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaoliu Shi
- Laboratory of Clinical Diagnosis and Research, Department of Medical Genetics, Second Xiangya Hospital of Central South University, Changsha, China
| | - Ping Chen
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.,Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Songping Liang
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.,Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zhonghua Liu
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.,Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
50
|
Hamed SA. The auditory and vestibular toxicities induced by antiepileptic drugs. Expert Opin Drug Saf 2017; 16:1281-1294. [PMID: 28838247 DOI: 10.1080/14740338.2017.1372420] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Sherifa A Hamed
- Department of Neurology and Psychiatry, Assiut University Hospital, Assiut, Egypt
| |
Collapse
|