1
|
Köster PA, Leipold E, Tigerholm J, Maxion A, Namer B, Stiehl T, Lampert A. Nociceptor sodium channels shape subthreshold phase, upstroke, and shoulder of action potentials. J Gen Physiol 2025; 157:e202313526. [PMID: 39836077 PMCID: PMC11748974 DOI: 10.1085/jgp.202313526] [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: 12/13/2023] [Revised: 09/30/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Voltage-gated sodium channels (VGSCs) in the peripheral nervous system shape action potentials (APs) and thereby support the detection of sensory stimuli. Most of the nine mammalian VGSC subtypes are expressed in nociceptors, but predominantly, three are linked to several human pain syndromes: while Nav1.7 is suggested to be a (sub-)threshold channel, Nav1.8 is thought to support the fast AP upstroke. Nav1.9, as it produces large persistent currents, is attributed a role in determining the resting membrane potential. We characterized the gating of Nav1.1-Nav1.3 and Nav1.5-Nav1.9 in manual patch clamp with a focus on the AP subthreshold depolarization phase. Nav1.9 exhibited the most hyperpolarized activation, while its fast inactivation resembled the depolarized inactivation of Nav1.8. For some VGSCs (e.g., Nav1.1 and Nav1.2), a positive correlation between ramp current and window current was detected. Using a modified Hodgkin-Huxley model that accounts for the time needed for inactivation to occur, we used the acquired data to simulate two nociceptive nerve fiber types (an Aδ- and a mechano-insensitive C-nociceptor) containing VGSC conductances according to published human RNAseq data. Our simulations suggest that Nav1.9 is supporting both the AP upstroke and its shoulder. A reduced threshold for AP generation was induced by enhancing Nav1.7 conductivity or shifting its activation to more hyperpolarized potentials, as observed in Nav1.7-related pain disorders. Here, we provide a comprehensive, comparative functional characterization of VGSCs relevant in nociception and describe their gating with Hodgkin-Huxley-like models, which can serve as a tool to study their specific contributions to AP shape and sodium channel-related diseases.
Collapse
Affiliation(s)
- Phil Alexander Köster
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Enrico Leipold
- Department of Anesthesiology and Intensive Care and CBBM-Center of Brain, Behavior and Metabolism, University of Luebeck, Lübeck, Germany
| | - Jenny Tigerholm
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Joint Research Center for Computational Biomedicine (JRCC), Uniklinik RWTH Aachen University, Aachen, Germany
| | - Anna Maxion
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, Research Group Neurosciences, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Barbara Namer
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, Research Group Neurosciences, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Thomas Stiehl
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Joint Research Center for Computational Biomedicine (JRCC), Uniklinik RWTH Aachen University, Aachen, Germany
- Institute for Computational Biomedicine and Disease Modelling With Focus on Phase Transitions Between Phenotypes, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Angelika Lampert
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| |
Collapse
|
2
|
Alsaloum M, Dib-Hajj SD, Page DA, Ruben PC, Krainer AR, Waxman SG. Voltage-gated sodium channels in excitable cells as drug targets. Nat Rev Drug Discov 2025:10.1038/s41573-024-01108-x. [PMID: 39901031 DOI: 10.1038/s41573-024-01108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 02/05/2025]
Abstract
Excitable cells - including neurons, muscle cells and cardiac myocytes - are unique in expressing high densities of voltage-gated sodium (NaV) channels. This molecular adaptation enables these cells to produce action potentials, and is essential to their function. With the advent of the molecular revolution, the concept of 'the' sodium channel has been supplanted by understanding that excitable cells in mammals can express any of nine different forms of sodium channels (NaV1.1-NaV1.9). Selective expression in particular types of cells, together with a key role in controlling action potential firing, makes some of these NaV subtypes especially attractive molecular targets for drug development. Although these different channel subtypes display a common overall structure, differences in their amino acid sequences have provided a basis for the development of subtype-specific drugs. This approach has resulted in exciting progress in the development of drugs for epilepsy, cardiac disorders and pain. In this Review, we discuss recent progress in the development of drugs that selectively target each of the sodium channel subtypes.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Dana A Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Su M, Ouyang X, Zhou P, Dong L, Shao L, Wang K, Liu Y. Inhibition of TTX-S Na + currents by a novel blocker QLS-278 for antinociception. J Pharmacol Exp Ther 2025; 392:100030. [PMID: 40023600 DOI: 10.1124/jpet.124.002273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/15/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
Genetic loss-of-function mutations of the NaV1.7 channel, abundantly expressed in peripheral nociceptive neurons, cause congenital insensitivity to pain in humans, indicating that selective inhibition of the channel may lead to potential therapy for pain disorders. In this study, we investigated a novel compound, 5-chloro-N-(cyclopropylsulfonyl)-2-fluoro-4-(2-(8-(furan-2-ylmethyl)-8-azaspiro [4.5] decan-2-yl) ethoxy) benzamide (QLS-278) that inhibits NaV1.7 channels and exhibits antinociceptive activity. Compound QLS-278 exhibits inactivation- and concentration-dependent inhibition of macroscopic currents of NaV1.7 channels stably expressed in HEK293 cells with an IC50 of 1.2 ± 0.2 μM. QLS-278 causes a hyperpolarization shift of the channel inactivation and delays recovery from inactivation, without any noticeable effect on voltage-dependent activation. In mouse dorsal root ganglion neurons, QLS-278 suppresses native tetrodotoxin-sensitive NaV currents and also reduces neuronal firings. Moreover, QLS-278 dose-dependently relieves neuropathic pain induced by spared nerve injury and inflammatory pain induced by formalin without significantly altering spontaneous locomotor activity in mice. Therefore, our identification of the novel compound QLS-278 may hold developmental potential in chronic pain treatment. SIGNIFICANCE STATEMENT: QLS-278, a novel voltage-gated sodium NaV1.7 channel blocker, inhibits native tetrodotoxin-sensitive Na+ current and reduces action potential firings in dorsal root ganglion sensory neurons. QLS-278 also exhibits antinociceptive activity in mouse models of pain, demonstrating the potential for the development of a chronic pain treatment.
Collapse
Affiliation(s)
- Min Su
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Xiangshuo Ouyang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Ping Zhou
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Liying Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China.
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China; Institute of Innovative Drugs, Qingdao University, Qingdao, China
| | - Yani Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China; Institute of Innovative Drugs, Qingdao University, Qingdao, China.
| |
Collapse
|
4
|
Motzkin JC, Basbaum AI, Crowther AJ. Neuroanatomy of the nociceptive system: From nociceptors to brain networks. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:1-39. [PMID: 39580210 DOI: 10.1016/bs.irn.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
This chapter reviews the neuroanatomy of the nociceptive system and its functional organization. We describe three main compartments of the nervous system that underlie normal nociception and the resulting pain percept: Peripheral, Spinal Cord, and Brain. We focus on how ascending nociceptive processing streams traverse these anatomical compartments, culminating in the multidimensional experience of pain. We also describe neuropathic pain conditions, in which nociceptive processing is abnormal, not only because of the primary effects of a lesion or disease affecting peripheral nerves or the central nervous system (CNS), but also due to secondary effects on ascending pathways and brain networks. We discuss how the anatomical components (circuits/networks) reorganize under various etiologies of neuropathic pain and how these changes can give rise to pathological pain states.
Collapse
Affiliation(s)
- Julian C Motzkin
- Department of Neurology and Department Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States.
| | - Allan I Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Andrew J Crowther
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
5
|
Wang L, Hao H, Meng X, Zhang W, Zhang Y, Chai T, Wang X, Gao Z, Zheng Y, Yang J. A novel isoquinoline alkaloid HJ-69 isolated from Zanthoxylum bungeanum attenuates inflammatory pain by inhibiting voltage-gated sodium and potassium channels. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118218. [PMID: 38677570 DOI: 10.1016/j.jep.2024.118218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Zanthoxylum bungeanum Maxim. (Z. bungeanum), a member of the Rutaceae family, has a rich history of traditional use in Asia for treating arthritis and toothache conditions. As characteristic chemical components, numerous kinds of alkaloids have been extracted from plants and their diverse biological activities have been reported. However, research on the isoquinoline alkaloid, a specific type of alkaloids, in Z. bungeanum was scarce. AIM OF THE STUDY The study aimed to isolate a novel isoquinoline alkaloid from Z. bungeanum and explore its pharmacological activity in vitro and analgesic activity in vivo. MATERIALS AND METHODS Isoquinoline alkaloid isolation and identification from Z. bungeanum were conducted using chromatographic and spectroscopic methods. The whole-cell patch-clamp technique was applied to assess its impact on neuronal excitability, and endogenous voltage-gated potassium (Kv) and sodium (Nav) currents in acutely isolated mouse small-diameter dorsal root ganglion (DRG) neurons. Its inhibitory impacts on channels were further validated with HEK293 cells stably expressing Nav1.7 and Nav1.8, and Chinese hamster ovary (CHO) cells transiently expressing Kv2.1. The formalin inflammatory pain model was utilized to evaluate the potential analgesic activity in vivo. RESULTS A novel isoquinoline alkaloid named HJ-69 (N-13-(3-methoxyprop-1-yl)rutaecarpine) was isolated and identified from Z. bungeanum for the first time. HJ-69 significantly suppressed the firing frequency and amplitudes of action potentials in DRG neurons. Consistently, it state-dependently inhibited endogenous Nav currents of DRG neurons, with half maximal inhibitory concentration (IC50) values of 13.06 ± 2.06 μM and 30.19 ± 2.07 μM for the inactivated and resting states, respectively. HJ-69 significantly suppressed potassium currents in DRG neurons, which notably inhibited the delayed rectifier potassium (IK) currents (IC50 = 6.95 ± 1.29 μM) and slightly affected the transient outward potassium (IA) currents (IC50 = 523.50 ± 39.16 μM). Furtherly, HJ-69 exhibited similar potencies on heterologously expressed Nav1.7, Nav1.8, and Kv2.1 channels, which correspondingly represent the main components in neurons. Notably, intraperitoneal administration of 30 mg/kg and 100 mg/kg HJ-69 significantly alleviated pain behaviors in the mouse inflammatory pain model induced by formalin. CONCLUSION The study concluded that HJ-69 is a novel and active isoquinoline alkaloid, and the inhibition of Nav and Kv channels contributes to its analgesic activity. HJ-69 may be a promising prototype for future analgesic drug discovery based on the isoquinoline alkaloid.
Collapse
Affiliation(s)
- Long Wang
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Haishuang Hao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xianhua Meng
- Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Wenbo Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yin Zhang
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tian Chai
- Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Xingrong Wang
- Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Zhaobing Gao
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yueming Zheng
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Junli Yang
- Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
| |
Collapse
|
6
|
Le Franc A, Da Silva A, Lepetre-Mouelhi S. Nanomedicine and voltage-gated sodium channel blockers in pain management: a game changer or a lost cause? Drug Deliv Transl Res 2024; 14:2112-2145. [PMID: 38861139 DOI: 10.1007/s13346-024-01615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 06/12/2024]
Abstract
Pain, a complex and debilitating condition affecting millions globally, is a significant concern, especially in the context of post-operative recovery. This comprehensive review explores the complexity of pain and its global impact, emphasizing the modulation of voltage-gated sodium channels (VGSC or NaV channels) as a promising avenue for pain management with the aim of reducing reliance on opioids. The article delves into the role of specific NaV isoforms, particularly NaV 1.7, NaV 1.8, and NaV 1.9, in pain process and discusses the development of sodium channel blockers to target these isoforms precisely. Traditional local anesthetics and selective NaV isoform inhibitors, despite showing varying efficacy in pain management, face challenges in systemic distribution and potential side effects. The review highlights the potential of nanomedicine in improving the delivery of local anesthetics, toxins and selective NaV isoform inhibitors for a targeted and sustained release at the site of pain. This innovative strategy seeks to improve drug bioavailability, minimize systemic exposure, and optimize therapeutic outcomes, holding significant promise for secure pain management and enhancing the quality of life for individuals recovering from surgical procedures or suffering from chronic pain.
Collapse
Affiliation(s)
- Adélaïde Le Franc
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Alexandre Da Silva
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | | |
Collapse
|
7
|
Zhao M, Wu J, Jin Y, Li M, Yu K, Yu H. Schisandrin B from Schisandra chinensis alleviated pain via glycine receptors, Nav1.7 channels and Cav2.2 channels. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117996. [PMID: 38431110 DOI: 10.1016/j.jep.2024.117996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/18/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Schisandra chinensis, the dried and ripe fruit of the magnolia family plant Schisandra chinensis (Turcz.) Baill, was commonly used in traditional analgesic prescription. Studies have shown that the extract of Schisandra chinensis (SC) displayed analgesic activity. However, the analgesic active component and the exact mechanisms have yet to be revealed. AIM OF THE STUDY The present study was to investigate the anti-nociceptive constituent of Schisandra chinensis, assess its analgesic effect, and explore the potential molecular mechanisms. MATERIALS AND METHODS The effects of a series of well-recognized compounds from SC on glycine receptors were investigated. The analgesic effect of the identified compound was evaluated in three pain models. Mechanistic studies were performed using patch clamp technique on various targets expressed in recombinant cells. These targets included glycine receptors, Nav1.7 sodium channels, Cav2.2 calcium channels et al. Meanwhile, primary cultured spinal dorsal horn (SDH) neurons and dorsal root ganglion (DRG) neurons were also utilized. RESULTS Schisandrin B (SchB) was a positive allosteric modulator of glycine receptors in spinal dorsal horn neurons. The EC50 of SchB on glycine receptors in spinal dorsal horn neurons was 2.94 ± 0.28 μM. In three pain models, the analgesic effect of SchB was comparable to that of indomethacin at the same dose. Besides, SchB rescued PGE2-induced suppression of α3 GlyR activity and alleviated persistent pain. Notably, SchB could also potently decrease the frequency of action potentials and inhibit sodium and calcium channels in DRG neurons. Consistent with the data from DRG neurons, SchB was also found to significantly block Nav1.7 sodium channels and Cav2.2 channels in recombinant cells. CONCLUSION Our results demonstrated that, Schisandrin B, the primary lignan component of Schisandra chinensis, may exert its analgesic effect by acting on multiple ion channels, including glycine receptors, Nav1.7 channels, and Cav2.2 channels.
Collapse
Affiliation(s)
- Miao Zhao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Jun Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Yuchen Jin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Min Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - KeXin Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Haibo Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
8
|
Salvage SC, Rahman T, Eagles DA, Rees JS, King GF, Huang CL, Jackson AP. The β3-subunit modulates the effect of venom peptides ProTx-II and OD1 on Na V 1.7 gating. J Cell Physiol 2023; 238:1354-1367. [PMID: 37042220 PMCID: PMC10953403 DOI: 10.1002/jcp.31018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 04/13/2023]
Abstract
The voltage-gated sodium channel NaV 1.7 is involved in various pain phenotypes and is physiologically regulated by the NaV -β3-subunit. Venom toxins ProTx-II and OD1 modulate NaV 1.7 channel function and may be useful as therapeutic agents and/or research tools. Here, we use patch-clamp recordings to investigate how the β3-subunit can influence and modulate the toxin-mediated effects on NaV 1.7 function, and we propose a putative binding mode of OD1 on NaV 1.7 to rationalise its activating effects. The inhibitor ProTx-II slowed the rate of NaV 1.7 activation, whilst the activator OD1 reduced the rate of fast inactivation and accelerated recovery from inactivation. The β3-subunit partially abrogated these effects. OD1 induced a hyperpolarising shift in the V1/2 of steady-state activation, which was not observed in the presence of β3. Consequently, OD1-treated NaV 1.7 exhibited an enhanced window current compared with OD1-treated NaV 1.7-β3 complex. We identify candidate OD1 residues that are likely to prevent the upward movement of the DIV S4 helix and thus impede fast inactivation. The binding sites for each of the toxins and the predicted location of the β3-subunit on the NaV 1.7 channel are distinct. Therefore, we infer that the β3-subunit influences the interaction of toxins with NaV 1.7 via indirect allosteric mechanisms. The enhanced window current shown by OD1-treated NaV 1.7 compared with OD1-treated NaV 1.7-β3 is discussed in the context of differing cellular expressions of NaV 1.7 and the β3-subunit in dorsal root ganglion (DRG) neurons. We propose that β3, as the native binding partner for NaV 1.7 in DRG neurons, should be included during screening of molecules against NaV 1.7 in relevant analgesic discovery campaigns.
Collapse
Affiliation(s)
| | - Taufiq Rahman
- Department of PharmacologyUniversity of CambridgeCambridgeUK
| | - David A. Eagles
- Institute of Molecular BioscienceUniversity of QueenslandBrisbaneQLDAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQLDAustralia
| | - Johanna S. Rees
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Present address:
Babraham Research CampusPetMedix Ltd.CambridgeUK
| | - Glenn F. King
- Institute of Molecular BioscienceUniversity of QueenslandBrisbaneQLDAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQLDAustralia
| | - Christopher L‐H. Huang
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
9
|
Zuo L, Su A, Shi Y, Li N, Chen S, Yang X. Case report: Spinal cord stimulation in the treatment of pediatric erythromelalgia. Front Neurol 2023; 14:1143241. [PMID: 37273700 PMCID: PMC10233004 DOI: 10.3389/fneur.2023.1143241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/27/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction In children, erythromelalgia is a rare chronic pain syndrome characterized by erythema, severe burning pain, and itching of affected feet. Unfortunately, there is no definitive therapy available currently. Case report Here, we report a case of primary erythromelalgia and the treatment response in a 10-year-old boy, whose genetic findings for mutations in the SCN9A gene were positive and skin biopsy results were diagnosed as small fiber neuropathy, while he has suffered from excruciating burning pain, itching, erythema, and recurrent infections over the past 3 years. He did not respond well to conventional treatment, and the only way to receive minimal relief was to immerse his feet in ice water. After a successful trial of spinal cord stimulation (SCS), the implantable pulse generator (IPG) was successfully implanted without complications, and it proved partial response to therapy. Conclusion There is no specific, efficient treatment for pediatric erythromelalgia currently, but this case demonstrates neuromodulation serves as part of the multimodal regimen to treat pediatric erythromelalgia.
Collapse
|
10
|
Almomani R, Sopacua M, Marchi M, Ślęczkowska M, Lindsey P, de Greef BTA, Hoeijmakers JGJ, Salvi E, Merkies ISJ, Ferdousi M, Malik RA, Ziegler D, Derks KWJ, Boenhof G, Martinelli-Boneschi F, Cazzato D, Lombardi R, Dib-Hajj S, Waxman SG, Smeets HJM, Gerrits MM, Faber CG, Lauria G. Genetic Profiling of Sodium Channels in Diabetic Painful and Painless and Idiopathic Painful and Painless Neuropathies. Int J Mol Sci 2023; 24:ijms24098278. [PMID: 37175987 PMCID: PMC10179245 DOI: 10.3390/ijms24098278] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/15/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Neuropathic pain is a frequent feature of diabetic peripheral neuropathy (DPN) and small fiber neuropathy (SFN). Resolving the genetic architecture of these painful neuropathies will lead to better disease management strategies, counselling and intervention. Our aims were to profile ten sodium channel genes (SCG) expressed in a nociceptive pathway in painful and painless DPN and painful and painless SFN patients, and to provide a perspective for clinicians who assess patients with painful peripheral neuropathy. Between June 2014 and September 2016, 1125 patients with painful-DPN (n = 237), painless-DPN (n = 309), painful-SFN (n = 547) and painless-SFN (n = 32), recruited in four different centers, were analyzed for SCN3A, SCN7A-SCN11A and SCN1B-SCN4B variants by single molecule Molecular inversion probes-Next Generation Sequence. Patients were grouped based on phenotype and the presence of SCG variants. Screening of SCN3A, SCN7A-SCN11A, and SCN1B-SCN4B revealed 125 different (potential) pathogenic variants in 194 patients (17.2%, n = 194/1125). A potential pathogenic variant was present in 18.1% (n = 142/784) of painful neuropathy patients vs. 15.2% (n = 52/341) of painless neuropathy patients (17.3% (n = 41/237) for painful-DPN patients, 14.9% (n = 46/309) for painless-DPN patients, 18.5% (n = 101/547) for painful-SFN patients, and 18.8% (n = 6/32) for painless-SFN patients). Of the variants detected, 70% were in SCN7A, SCN9A, SCN10A and SCN11A. The frequency of SCN9A and SCN11A variants was the highest in painful-SFN patients, SCN7A variants in painful-DPN patients, and SCN10A variants in painless-DPN patients. Our findings suggest that rare SCG genetic variants may contribute to the development of painful neuropathy. Genetic profiling and SCG variant identification should aid in a better understanding of the genetic variability in patients with painful and painless neuropathy, and may lead to better risk stratification and the development of more targeted and personalized pain treatments.
Collapse
Affiliation(s)
- Rowida Almomani
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Maurice Sopacua
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Margherita Marchi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Milena Ślęczkowska
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick Lindsey
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Erika Salvi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Ingemar S J Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
- Department of Neurology, Curaçao Medical Center, 4365+37Q, J. H. J. Hamelbergweg, Willemstad, Curacao
| | - Maryam Ferdousi
- Institute of Cardiovascular Sciences, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9P, UK
| | - Rayaz A Malik
- Institute of Cardiovascular Sciences, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9P, UK
- Weill Cornell Medicine-Qatar, Doha P.O. Box 24144, Qatar
| | - Dan Ziegler
- German Diabetes Centre, 40225 Düsseldorf, Germany
| | - Kasper W J Derks
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Gidon Boenhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, 40225 Düsseldorf, Germany
| | - Filippo Martinelli-Boneschi
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniele Cazzato
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Raffaella Lombardi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Hubert J M Smeets
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Monique M Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, 20157 Milan, Italy
| |
Collapse
|
11
|
Li S, Jin Y, Li M, Yu H. NAN-190, a 5-HT 1A antagonist, alleviates inflammatory pain by targeting Nav1.7 sodium channels. Life Sci 2023; 319:121520. [PMID: 36828129 DOI: 10.1016/j.lfs.2023.121520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/10/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
AIMS In the present study, NAN-190 [1-(2-methoxyphenyl)-4-[4-(2-phthalimido) butyl] piperazine] was identified as a Nav1.7 blocker. In the meantime, the compound could alleviate the Complete Freund's Adjuvant (CFA)-induced inflammatory pain. To understand the molecular mechanisms of NAN-190 on pain, the effect of NAN-190 on Nav1.7 sodium channels was studied. MAIN METHODS Inflammatory pain was induced by injection of CFA solution into the plantar side of the left hindpaw. Thermal hyperalgesia and mechanical allodynia were measured. Whole-cell patch clamp methods were used to record sodium channels and other pain-related targets in the cultured recombinant cells and dorsal root ganglion neurons. KEY FINDINGS Nan-190 was identified as an inhibitor of Nav1.7 sodium channels and animal experiments showed that NAN-190 significantly alleviated CFA-induced inflammatory pain. Mechanism studies demonstrated that NAN-190 was a state-dependent Nav1.7 blocker with IC50 value on the inactivated state ten-fold more potent than that on the rest state. NAN-190 leftward-shifted the fast and slow inactivation curves about 9.07 mV and 38.56 mV, respectively, but had no effects on channel activation. The compound also slowed the recovery from fast and slow inactivation and showed use-dependent properties. Further, the site-directed mutagenesis experiments demonstrated that NAN-190 mainly worked on the open state of Nav1.7 channels by interacting with sites similar as local anesthetics. In DRG neurons, NAN-190 mainly blocks TTX-sensitive currents but is less sensitive to TTX-R sodium currents. SIGNIFICANCE Taken together, our results indicated that NAN-190 alleviated pain behaviors by blocking sodium channels by interacting with the open state.
Collapse
Affiliation(s)
- Shaohua Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yuchen Jin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Min Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Haibo Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
12
|
Small Fiber Neuropathy: A Clinical and Practical Approach. J Nurse Pract 2023. [DOI: 10.1016/j.nurpra.2023.104547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
13
|
Kim SE, Yin MZ, Roh JW, Kim HJ, Choi SW, Wainger BJ, Kim WK, Kim SJ, Nam JH. Multi-target modulation of ion channels underlying the analgesic effects of α-mangostin in dorsal root ganglion neurons. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154791. [PMID: 37094425 DOI: 10.1016/j.phymed.2023.154791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/15/2023] [Accepted: 03/28/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND α-Mangostin is a xanthone isolated from the pericarps of mangosteen fruit with, and has analgesic properties. Although the effects suggest an interaction of α-mangostin with ion channels in the nociceptive neurons, electrophysiological investigation of the underlying mechanism has not been performed. HYPOTHESIS We hypothesized that α-Mangostin exerts its analgesic effects by modulating the activity of various ion channels in dorsal root ganglion (DRG) neurons. METHODS We performed a whole-cell patch clamp study using mouse DRG neurons, HEK293T cells overexpressing targeted ion channels, and ND7/23 cells. Molecular docking (MD) and in silico absorption, distribution, metabolism, and excretion (ADME) analyses were conducted to obtain further insights into the binding sites and pharmacokinetics, respectively. RESULTS Application of α-mangostin (1-3 µM) hyperpolarized the resting membrane potential (RMP) of small-sized DRG neurons by increasing background K+ conductance and thereby inhibited action potential generation. At micromolar levels, α-mangostin activates TREK-1, TREK-2, or TRAAK, members of the two-pore domain K+ channel (K2P) family known to be involved in RMP formation in DRG neurons. Furthermore, capsaicin-induced TRPV1 currents were potently inhibited by α-mangostin (0.43 ± 0.27 µM), and partly suppressed tetrodotoxin-sensitive voltage-gated Na+ channel (NaV) currents. MD simulation revealed that multiple oxygen atoms in α-mangostin may form stable hydrogen bonds with TREKs, TRAAK, TRPV1, and NaV channels. In silico ADME tests suggested that α-mangostin may satisfy the drug-likeness properties without penetrating the blood-brain barrier. CONCLUSION The analgesic properties of α-mangostin might be mediated by the multi-target modulation of ion channels, including TREK/TRAAK activation, TRPV1 inhibition, and reduction of the tetrodotoxin-sensitive NaV current. The findings suggest that the phytochemical can be a multi-ion channel-targeting drug and an alternative drug for effective pain management.
Collapse
Affiliation(s)
- Sung Eun Kim
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Ming Zhe Yin
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jae Won Roh
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyun Jong Kim
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Gyeonggi-do 10326, Republic of Korea
| | - Seong Woo Choi
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Brian J Wainger
- Departments Anesthesia, Critical Care & Pain Medicine and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston 02114, ts, USA
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Gyeonggi-do 10326, Republic of Korea; Department of Internal Medicine Graduate School of Medicine, Dongguk University, Gyeonggi-do 10326, Republic of Korea.
| | - Sung Joon Kim
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Departments Anesthesia, Critical Care & Pain Medicine and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston 02114, ts, USA; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Gyeonggi-do 10326, Republic of Korea.
| |
Collapse
|
14
|
Wang F, Cheung CW, Wong SSC. Use of pain-related gene features to predict depression by support vector machine model in patients with fibromyalgia. Front Genet 2023; 14:1026672. [PMID: 37065490 PMCID: PMC10090498 DOI: 10.3389/fgene.2023.1026672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
The prevalence rate of depression is higher in patients with fibromyalgia syndrome, but this is often unrecognized in patients with chronic pain. Given that depression is a common major barrier in the management of patients with fibromyalgia syndrome, an objective tool that reliably predicts depression in patients with fibromyalgia syndrome could significantly enhance the diagnostic accuracy. Since pain and depression can cause each other and worsen each other, we wonder if pain-related genes can be used to differentiate between those with major depression from those without. This study developed a support vector machine model combined with principal component analysis to differentiate major depression in fibromyalgia syndrome patients using a microarray dataset, including 25 fibromyalgia syndrome patients with major depression, and 36 patients without major depression. Gene co-expression analysis was used to select gene features to construct support vector machine model. The principal component analysis can help reduce the number of data dimensions without much loss of information, and identify patterns in data easily. The 61 samples available in the database were not enough for learning based methods and cannot represent every possible variation of each patient. To address this issue, we adopted Gaussian noise to generate a large amount of simulated data for training and testing of the model. The ability of support vector machine model to differentiate major depression using microarray data was measured as accuracy. Different structural co-expression patterns were identified for 114 genes involved in pain signaling pathway by two-sample KS test (p < 0.001 for the maximum deviation D = 0.11 > Dcritical = 0.05), indicating the aberrant co-expression patterns in fibromyalgia syndrome patients. Twenty hub gene features were further selected based on co-expression analysis to construct the model. The principal component analysis reduced the dimension of the training samples from 20 to 16, since 16 components were needed to retain more than 90% of the original variance. The support vector machine model was able to differentiate between those with major depression from those without in fibromyalgia syndrome patients with an average accuracy of 93.22% based on the expression levels of the selected hub gene features. These findings would contribute key information that can be used to develop a clinical decision-making tool for the data-driven, personalized optimization of diagnosing depression in patients with fibromyalgia syndrome.
Collapse
|
15
|
Gale JR, Gedeon JY, Donnelly CJ, Gold MS. Local translation in primary afferents and its contribution to pain. Pain 2022; 163:2302-2314. [PMID: 35438669 PMCID: PMC9579217 DOI: 10.1097/j.pain.0000000000002658] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 04/08/2022] [Indexed: 02/06/2023]
Abstract
ABSTRACT Chronic pain remains a significant problem due to its prevalence, impact, and limited therapeutic options. Progress in addressing chronic pain is dependent on a better understanding of underlying mechanisms. Although the available evidence suggests that changes within the central nervous system contribute to the initiation and maintenance of chronic pain, it also suggests that the primary afferent plays a critical role in all phases of the manifestation of chronic pain in most of those who suffer. Most notable among the changes in primary afferents is an increase in excitability or sensitization. A number of mechanisms have been identified that contribute to primary afferent sensitization with evidence for both increases in pronociceptive signaling molecules, such as voltage-gated sodium channels, and decreases in antinociceptive signaling molecules, such as voltage-dependent or calcium-dependent potassium channels. Furthermore, these changes in signaling molecules seem to reflect changes in gene expression as well as posttranslational processing. A mechanism of sensitization that has received far less attention, however, is the local or axonal translation of these signaling molecules. A growing body of evidence indicates that this process not only is dynamically regulated but also contributes to the initiation and maintenance of chronic pain. Here, we review the biology of local translation in primary afferents and its relevance to pain pathobiology.
Collapse
Affiliation(s)
- Jenna R Gale
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Jeremy Y Gedeon
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | | | - Michael S Gold
- Corresponding author: Michael S Gold, PhD, Department of Neurobiology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, P: 412-383-5367,
| |
Collapse
|
16
|
Effraim PR, Estacion M, Zhao P, Sosniak D, Waxman SG, Dib-Hajj SD. Fibroblast growth factor homologous factor 2 attenuates excitability of DRG neurons. J Neurophysiol 2022; 128:1258-1266. [PMID: 36222860 PMCID: PMC9909838 DOI: 10.1152/jn.00361.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factor homologous factors (FHFs) are cytosolic members of the superfamily of the FGF proteins. Four members of this subfamily (FHF1-4) are differentially expressed in multiple tissues in an isoform-dependent manner. Mutations in FHF proteins have been associated with multiple neurological disorders. FHF proteins bind to the COOH terminus of voltage-gated sodium (Nav) channels and regulate current amplitude and gating properties of these channels. FHF2, which is expressed in dorsal root ganglia (DRG) neurons, has two main splicing isoforms: FHF2A and FHF2B, which differ in the length and sequence of their NH2 termini, have been shown to differentially regulate gating properties of Nav1.7, a channel that is a major driver of DRG neuron firing. FHF2 expression levels are downregulated after peripheral nerve axotomy, which suggests that they may regulate neuronal excitability via an action on Nav channels after injury. We have previously shown that knockdown of FHF2 leads to gain-of-function changes in Nav1.7 gating properties: enhanced repriming, increased current density, and hyperpolarized activation. From this we posited that knockdown of FHF2 might also lead to DRG hyperexcitability. Here we show that knockdown of either FHF2A alone or all isoforms of FHF2 results in increased DRG neuron excitability. In addition, we demonstrate that supplementation of FHF2A and FHF2B reduces DRG neuron excitability. Overexpression of FHF2A or FHF2B also reduced excitability of DRG neurons treated with a cocktail of inflammatory mediators, a model of inflammatory pain. Our data suggest that increased neuronal excitability after nerve injury might be triggered, in part, via a loss of FHF2-Nav1.7 interaction.NEW & NOTEWORTHY FHF2 is known to bind to and modulate the function of Nav1.7. FHF2 expression is also reduced after nerve injury. We demonstrate that knockdown of FHF2 expression increases DRG neuronal excitability. More importantly, overexpression of FHF2 reduces DRG excitability in basal conditions and in the presence of inflammatory mediators (a model of inflammatory pain). These results suggest that FHF2 could potentially be used as a tool to reduce DRG neuronal excitability and to treat pain.
Collapse
Affiliation(s)
- Philip R. Effraim
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Daniel Sosniak
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
17
|
Gallagher CI, Ha DA, Harvey RJ, Vandenberg RJ. Positive Allosteric Modulators of Glycine Receptors and Their Potential Use in Pain Therapies. Pharmacol Rev 2022; 74:933-961. [PMID: 36779343 PMCID: PMC9553105 DOI: 10.1124/pharmrev.122.000583] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/26/2022] [Accepted: 05/13/2022] [Indexed: 11/22/2022] Open
Abstract
Glycine receptors are ligand-gated ion channels that mediate synaptic inhibition throughout the mammalian spinal cord, brainstem, and higher brain regions. They have recently emerged as promising targets for novel pain therapies due to their ability to produce antinociception by inhibiting nociceptive signals within the dorsal horn of the spinal cord. This has greatly enhanced the interest in developing positive allosteric modulators of glycine receptors. Several pharmaceutical companies and research facilities have attempted to identify new therapeutic leads by conducting large-scale screens of compound libraries, screening new derivatives from natural sources, or synthesizing novel compounds that mimic endogenous compounds with antinociceptive activity. Advances in structural techniques have also led to the publication of multiple high-resolution structures of the receptor, highlighting novel allosteric binding sites and providing additional information for previously identified binding sites. This has greatly enhanced our understanding of the functional properties of glycine receptors and expanded the structure activity relationships of novel pharmacophores. Despite this, glycine receptors are yet to be used as drug targets due to the difficulties in obtaining potent, selective modulators with favorable pharmacokinetic profiles that are devoid of side effects. This review presents a summary of the structural basis for how current compounds cause positive allosteric modulation of glycine receptors and discusses their therapeutic potential as analgesics. SIGNIFICANCE STATEMENT: Chronic pain is a major cause of disability, and in Western societies, this will only increase as the population ages. Despite the high level of prevalence and enormous socioeconomic burden incurred, treatment of chronic pain remains limited as it is often refractory to current analgesics, such as opioids. The National Institute for Drug Abuse has set finding effective, safe, nonaddictive strategies to manage chronic pain as their top priority. Positive allosteric modulators of glycine receptors may provide a therapeutic option.
Collapse
Affiliation(s)
- Casey I Gallagher
- Molecular Biomedicine, School of Medical Sciences, University of Sydney, Sydney, Australia (C.I.G., D.A.H., R.J.V.) and Biomedical Science, School of Health and Behavioural Sciences and Sunshine Coast Health Institute, University of the Sunshine Coast, Maroochydore, Australia (R.J.H.)
| | - Damien A Ha
- Molecular Biomedicine, School of Medical Sciences, University of Sydney, Sydney, Australia (C.I.G., D.A.H., R.J.V.) and Biomedical Science, School of Health and Behavioural Sciences and Sunshine Coast Health Institute, University of the Sunshine Coast, Maroochydore, Australia (R.J.H.)
| | - Robert J Harvey
- Molecular Biomedicine, School of Medical Sciences, University of Sydney, Sydney, Australia (C.I.G., D.A.H., R.J.V.) and Biomedical Science, School of Health and Behavioural Sciences and Sunshine Coast Health Institute, University of the Sunshine Coast, Maroochydore, Australia (R.J.H.)
| | - Robert J Vandenberg
- Molecular Biomedicine, School of Medical Sciences, University of Sydney, Sydney, Australia (C.I.G., D.A.H., R.J.V.) and Biomedical Science, School of Health and Behavioural Sciences and Sunshine Coast Health Institute, University of the Sunshine Coast, Maroochydore, Australia (R.J.H.)
| |
Collapse
|
18
|
Klein-Weigel P, Ruttloff A, König D, Nielitz J, Steindl J, Sander O, Richter JG. [Functional vascular acrosyndromes]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2022; 63:591-600. [PMID: 35925129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 06/15/2023]
Abstract
Vascular acrosyndromes are characterized by sparse, uniform clinical manifestations and a variety of possible pathomechanisms. The present article focuses on the functional entities. Raynaud phenomenon is based on cold- or stress-induced vasospasms of acral arteries. It is defined by the color changes of the skin, in the typical case white-blue-red (tricolore). The long fingers are most commonly affected. The etiology is unknown, and the pathophysiology is poorly understood. A distinction is made between primary and a secondary Raynaud phenomenon. The most important underlying diseases include collagenosis, primarily systemic sclerosis, and malignancies; furthermore, medications and drugs may promote vasospasm. Treatment is aimed at preventing or breaking the vasospasm, but has been only partially effective in doing so. Acrocyanosis is a vasospastic dystonic acral disorder that results in permanent reddish-livid discoloration, especially of the hands and feet. Secondary forms occur in collagenosis, malignancies, and myelodysplastic syndromes. The etiology and pathophysiology are virtually unknown. Targeted pharmacological intervention is not possible. Unlike all other vascular acrosyndromes, erythromelalgia is characterized by hyperemia. The primary form is a genetic sodium channelopathy, while secondary forms include malignancies, connective tissue diseases, and myelodysplastic syndromes. The symptoms are often distressing and disabling. Therapy requires a multimodal approach that includes both nonpharmacological and pharmacological strategies. Close interdisciplinary collaboration is essential for the management of this disease.
Collapse
Affiliation(s)
- Peter Klein-Weigel
- Klinik für Angiologie, Klinikum Ernst von Bergmann, Charlottenstr. 72, 14467, Potsdam, Deutschland.
| | - Andreas Ruttloff
- Klinik für Angiologie, Klinikum Ernst von Bergmann, Charlottenstr. 72, 14467, Potsdam, Deutschland
| | - Dana König
- Klinik für Angiologie, Klinikum Ernst von Bergmann, Charlottenstr. 72, 14467, Potsdam, Deutschland
| | - Jessica Nielitz
- Klinik für Angiologie, Klinikum Ernst von Bergmann, Charlottenstr. 72, 14467, Potsdam, Deutschland
| | - Julia Steindl
- Klinik für Angiologie, Klinikum Ernst von Bergmann, Charlottenstr. 72, 14467, Potsdam, Deutschland
| | - Oliver Sander
- Poliklinik und Funktionsbereich für Rheumatologie & Hiller-Forschungszentrum für Rheumatologie, Universitätsklinikum Düsseldorf, Medizinische Fakultät, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Deutschland
| | - Jutta G Richter
- Poliklinik und Funktionsbereich für Rheumatologie & Hiller-Forschungszentrum für Rheumatologie, Universitätsklinikum Düsseldorf, Medizinische Fakultät, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Deutschland
| |
Collapse
|
19
|
Multitarget nociceptor sensitization by a promiscuous peptide from the venom of the King Baboon spider. Proc Natl Acad Sci U S A 2022; 119:2110932119. [PMID: 35074873 PMCID: PMC8812547 DOI: 10.1073/pnas.2110932119] [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] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
The King Baboon spider, Pelinobius muticus, is a burrowing African tarantula. Its impressive size and appealing coloration are tempered by reports describing severe localized pain, swelling, itchiness, and muscle cramping after accidental envenomation. Hyperalgesia is the most prominent symptom after bites from P. muticus, but the molecular basis by which the venom induces pain is unknown. Proteotranscriptomic analysis of P. muticus venom uncovered a cysteine-rich peptide, δ/κ-theraphotoxin-Pm1a (δ/κ-TRTX-Pm1a), that elicited nocifensive behavior when injected into mice. In small dorsal root ganglion neurons, synthetic δ/κ-TRTX-Pm1a (sPm1a) induced hyperexcitability by enhancing tetrodotoxin-resistant sodium currents, impairing repolarization and lowering the threshold of action potential firing, consistent with the severe pain associated with envenomation. The molecular mechanism of nociceptor sensitization by sPm1a involves multimodal actions over several ion channel targets, including NaV1.8, KV2.1, and tetrodotoxin-sensitive NaV channels. The promiscuous targeting of peptides like δ/κ-TRTX-Pm1a may be an evolutionary adaptation in pain-inducing defensive venoms.
Collapse
|
20
|
Cheng KI, Wang HC, Tseng KY, Wang YH, Chang CY, Chen YJ, Lai CS, Chen DR, Chang LL. Cilostazol Ameliorates Peripheral Neuropathic Pain in Streptozotocin-Induced Type I Diabetic Rats. Front Pharmacol 2022; 12:771271. [PMID: 35115925 PMCID: PMC8804339 DOI: 10.3389/fphar.2021.771271] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/09/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Cilostazol is an antiplatelet agent with vasodilating, endothelial function restoration, and anti-inflammatory effects. This study aims to investigate the efficacy of oral cilostazol for preventing the development of diabetic peripheral neuropathy (DPN). Materials and Methods: Ninety adult male Sprague-Dawley rats were divided into five groups: 1) naïve (control); 2) diabetic (DM); 3) DM receiving 10 mg/kg cilostazol (cilo-10); 4) DM receiving 30 mg/kg cilostazol (cilo-30); and 5) DM receiving 100 mg/kg cilostazol (cilo-100). Hindpaw responses to thermal and mechanical stimuli were measured. Activation of microglia and astrocytes in the spinal dorsal horn (SDH) and expression of NaVs in the dorsal root ganglia (DRG) were examined with Western blots and immunofluorescence. Results: DM rats displayed decreased withdrawal thresholds to mechanical stimuli (mechanical allodynia) and blunted responses to thermal stimuli. In addition, the expression of microglia increased, but astrocytes were reduced in the SDH. Upregulation of Nav −1.1, 1.2, −1.3, −1.6, and −1.7 and downregulation of Nav-1.8 were observed in the DRG. The DM rats receiving cilostazol all returned DM-induced decrease in withdrawal threshold to mechanical stimuli and attenuated neuropathic pain. Additionally, all cilostazol treatments suppressed the level of activated microglial cells and ameliorated the DM-induced decline in astrocyte expression levels in the SDH. However, only the rats treated with cilo-100 demonstrated significant improvements to the aberrant NaV expression in the DRG. Conclusion: Oral cilostazol can blunt the responses of mechanical allodynia and has the potential to treat diabetic neuropathy by attenuating NaV and glial cell dysregulation.
Collapse
Affiliation(s)
- Kuang-I. Cheng
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hung-Chen Wang
- Department of Neurosurgery, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kuang-Yi Tseng
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hsuan Wang
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chung-Yu Chang
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Jing Chen
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Sheng Lai
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Dar-Ren Chen
- Endoscopic and Oncoplastic Breast Surgery Center, Changhua Christian Hospital, Changhua, Taiwan
- Department of Surgery, Division of General Surgery, Changhua Christian Hospital, Changhua, Taiwan
- *Correspondence: Lin-Li Chang, ; Dar-Ren Chen,
| | - Lin-Li Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- *Correspondence: Lin-Li Chang, ; Dar-Ren Chen,
| |
Collapse
|
21
|
Small-Fiber-Neuropathien. DGNEUROLOGIE 2022; 5. [PMCID: PMC9559077 DOI: 10.1007/s42451-022-00488-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Die Small-Fiber-Neuropathie (SFN) ist eine Erkrankung des peripheren Nervensystems aufgrund einer isolierten oder vorwiegenden Schädigung dünn myelinisierter Aδ-Fasern und/oder unmyelinisierter C‑Fasern. Für die sichere Diagnose einer SFN ist neben der klinischen Symptomatik mit Schmerzen und Sensibilitätsstörungen, typischerweise mit distal betonter Ausbreitung, der apparative Nachweis einer Rarefizierung oder einer Funktionsstörung der dünn myelinisierten Aδ-Fasern und/oder der unmyelinisierten C‑Fasern gefordert. Im vorliegenden Beitrag wird eine Übersicht über die diagnostischen Verfahren zum Nachweis einer SFN sowie über mögliche Ursachen und Behandlungsoptionen gegeben.
Collapse
|
22
|
Stucky CL, Mikesell AR. Cutaneous pain in disorders affecting peripheral nerves. Neurosci Lett 2021; 765:136233. [PMID: 34506882 PMCID: PMC8579816 DOI: 10.1016/j.neulet.2021.136233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 08/16/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023]
Abstract
Our ability to quickly detect and respond to harmful environmental stimuli is vital for our safety and survival. This inherent acute pain detection is a "gift" because it both protects our body from harm and allows healing of damaged tissues [1]. Damage to tissues from trauma or disease can result in distorted or amplified nociceptor signaling and sensitization of the spinal cord and brain (Central Nervous System; CNS) pathways to normal input from light touch mechanoreceptors. Together, these processes can result in nagging to unbearable chronic pain and extreme sensitivity to light skin touch (allodynia). Unlike acute protective pain, chronic pain and allodynia serve no useful purpose and can severely reduce the quality of life of an affected person. Chronic pain can arise from impairment to peripheral neurons, a phenomenon called "peripheral neuropathic pain." Peripheral neuropathic pain can be caused by many insults that directly affect peripheral sensory neurons, including mechanical trauma, metabolic imbalance (e.g., diabetes), autoimmune diseases, chemotherapeutic agents, viral infections (e.g., shingles). These insults cause "acquired" neuropathies such as small-fiber neuropathies, diabetic neuropathy, chemotherapy-induced peripheral neuropathy, and post herpetic neuralgia. Peripheral neuropathic pain can also be caused by genetic factors and result in hereditary neuropathies that include Charcot-Marie-Tooth disease, rare channelopathies and Fabry disease. Many acquired and hereditary neuropathies affect the skin, our largest organ and protector of nearly our entire body. Here we review how cutaneous nociception (pain perceived from the skin) is altered following diseases that affect peripheral nerves that innervate the skin. We provide an overview of how noxious stimuli are detected and encoded by molecular transducers on subtypes of cutaneous afferent endings and conveyed to the CNS. Next, we discuss several acquired and hereditary diseases and disorders that cause painful or insensate (lack of sensation) cutaneous peripheral neuropathies, the symptoms and percepts patients experience, and how cutaneous afferents and other peripheral cell types are altered in function in these disorders. We highlight exciting new research areas that implicate non-neuronal skin cells, particularly keratinocytes, in cutaneous nociception and peripheral neuropathies. Finally, we conclude with ideas for innovative new directions, areas of unmet need, and potential opportunities for novel cutaneous therapeutics that may avoid CNS side effects, as well as ideas for improved translation of mechanisms identified in preclinical models to patients.
Collapse
Affiliation(s)
- Cheryl L Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States.
| | - Alexander R Mikesell
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| |
Collapse
|
23
|
Boeddinghaus R, Whyte A. Imaging of Trigeminal Neuralgia and Other Facial Pain. Neuroimaging Clin N Am 2021; 31:485-508. [PMID: 34689929 DOI: 10.1016/j.nic.2021.05.008] [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: 11/16/2022]
Abstract
We review and illustrate the radiology of facial pain, emphasizing trigeminal neuralgia, relevant anatomy, current classification, concepts about etiology, and the role of imaging and its influence on the choice of treatment. We discuss glossopharyngeal neuralgia, other neuropathic causes of facial pain, postinflammatory and neoplastic causes, and nociceptive (end-organ) causes of facial pain, as well as referred otalgia. Other conditions that may present with facial pain, including trigeminal autonomic cephalgias and giant cell arteritis, are reviewed briefly. We discuss the elements of a comprehensive MR imaging protocol to enable detection of these diverse causes of facial pain.
Collapse
Affiliation(s)
- Rudolf Boeddinghaus
- Perth Radiological Clinic, 127 Hamersley Road, Subiaco, Western Australia 6008, Australia; Department of Surgery, University of Western Australia, 35 Stirling Highway, Perth, Western Australia 6009, Australia.
| | - Andy Whyte
- Department of Medicine and Radiology, University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia; Department of Dentistry, University of Western Australia, 35 Stirling Highway, Perth, Western Australia 6009, Australia
| |
Collapse
|
24
|
Dai Z, Xu X, Chen Y, Lin C, Lin F, Liu R. Effects of High-Voltage Pulsed Radiofrequency on the Ultrastructure and Nav1.7 Level of the Dorsal Root Ganglion in Rats With Spared Nerve Injury. Neuromodulation 2021; 25:980-988. [PMID: 34487572 DOI: 10.1111/ner.13527] [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: 04/03/2021] [Revised: 07/18/2021] [Accepted: 08/03/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To investigate the analgesic effect of high-voltage pulsed radiofrequency (HV-PRF) on the dorsal root ganglion (DRG) for neuropathic pain induced by spared nerve injury (SNI) in rats, especially the influence of this treatment on the DRG ultrastructure and voltage-gated sodium channel 1.7 (Nav1.7) level in the DRG. MATERIALS AND METHODS One hundred fifty adult male Sprague-Dawley rats were randomly divided into five groups: Sham, SNI, Free-PRF, standard-voltage PRF (SV-PRF), and HV-PRF. The 45V-PRF and 85V-PRF procedures applied to the left L5 DRG were performed in SV-PRF group and the HV-PRF group respectively on day 7 after SNI, whereas no PRF was concurrently delivered in Free-PRF group. The paw mechanical withdrawal threshold (PMWT) was detected before SNI (baseline) and on days 1, 3, 7, 8, 10, 14, and 21. The changes of left L5 DRG ultrastructure were analyzed with transmission electron microscopy on days 14 and 21. The expression levels of Nav1.7 in left L5 DRG were detected by immunofluorescence and Western blot. RESULTS Compared with the Free-PRF group, PMWT in the SV-PRF group and HV-PRF group were both significantly increased after PRF (all p < 0.05). Meanwhile, the PMWT was significantly higher in the HV-PRF group than that in the SV-PRF group on days 14 and 21 all (p < 0.05). There were statistically significant differences between the SV-PRF and Free-PRF groups (p < 0.05). Similarly, statistically significant difference was found between the HV-PRF and Free-PRF groups (p < 0.05). Especially, comparison of the SV-PRF group and the HV-PRF group revealed statistically significant difference (p < 0.05). The Nav1.7 levels were significantly down-regulated in the SV-PRF group and HV-PRF groups compared to that in the Free-PRF group (all p < 0.01). A significantly lower Nav1.7 level was also found in the HV-PRF group compared to that in the SV-PRF group (p < 0.05). CONCLUSIONS The HV-PRF produces a better analgesic effect than SV-PRF applied to the DRG in SNI rats. The underlying mechanisms may be associated with improving the histopathological prognosis and the downregulation of Nav1.7 levels in the DRG.
Collapse
Affiliation(s)
- Zhisen Dai
- Department of Pain Management, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xueru Xu
- Department of Pain Management, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yanqin Chen
- Department of Pain Management, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chun Lin
- Laboratory of Pain Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Fan Lin
- Fujian Key Laboratory of Geriatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Rongguo Liu
- Department of Pain Management, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
25
|
Bali KK, Gandla J, Rangel DR, Castaldi L, Mouritzen P, Agarwal N, Schmelz M, Heppenstall P, Kuner R. A genome-wide screen reveals microRNAs in peripheral sensory neurons driving painful diabetic neuropathy. Pain 2021; 162:1334-1351. [PMID: 33492037 DOI: 10.1097/j.pain.0000000000002159] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/23/2020] [Indexed: 12/25/2022]
Abstract
ABSTRACT Diabetes is a leading cause of peripheral neuropathy (diabetic peripheral neuropathy, DPN), and uncontrolled long-lasting hyperglycemia leads to severe complications. A major proportion of diabetics develop excruciating pain with a variable course. Mechanisms leading to painful DPN are not completely understood and treatment options limited. We hypothesized that epigenetic modulation at the level of microRNA (miRNA) expression triggered by metabolic imbalance and nerve damage regulates the course of pain development. We used clinically relevant preclinical models, genome-wide screening, in silico analyses, cellular assays, miRNA fluorescent in situ hybridization, in vivo molecular manipulations, and behavioral analyses in the current study. We identified miRNAs and their targets that critically impact on nociceptive hypersensitivity in painful DPN. Our analyses identify miR-33 and miR-380 expressed in nociceptive neurons as critical denominators of diabetic pain and miR-124-1 as a mediator of physiological nociception. Our comprehensive analyses on the putative mRNA targets for miR-33 or miR-124-1 identified a set of mRNAs that are regulated after miR-33 or miR-124-1 overexpression in dorsal root ganglia in vivo. Our results shed light on the regulation of DPN pathophysiology and implicate specific miRNAs as novel therapeutic targets for treating painful DPN.
Collapse
Affiliation(s)
- Kiran Kumar Bali
- Department of Molecular Pharmacology, Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany . Dr. Bali is now with the Department of Experimental Pain Research, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jagadeesh Gandla
- Department of Molecular Pharmacology, Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany . Dr. Bali is now with the Department of Experimental Pain Research, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Rojas Rangel
- Department of Molecular Pharmacology, Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany . Dr. Bali is now with the Department of Experimental Pain Research, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | - Nitin Agarwal
- Department of Molecular Pharmacology, Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany . Dr. Bali is now with the Department of Experimental Pain Research, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Martin Schmelz
- Department of Experimental Pain Research, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Rohini Kuner
- Department of Molecular Pharmacology, Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany . Dr. Bali is now with the Department of Experimental Pain Research, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
26
|
Sachau J, Kersebaum D, Baron R, Dickenson AH. Unusual Pain Disorders - What Can Be Learned from Them? J Pain Res 2021; 13:3539-3554. [PMID: 33758536 PMCID: PMC7980038 DOI: 10.2147/jpr.s287603] [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: 10/30/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Pain is common in many different disorders and leads to a significant reduction in quality of life in the affected patients. Current treatment options are limited and often result in insufficient pain relief, partly due to the incomplete understanding of the underlying pathophysiological mechanisms. The identification of these pathomechanisms is therefore a central object of current research. There are also a number of rare pain diseases, that are generally little known and often undiagnosed, but whose correct diagnosis and examination can help to improve the management of pain disorders in general. In some of these unusual pain disorders like sodium-channelopathies or sensory modulation disorder the underlying pathophysiological mechanisms have only recently been unravelled. These mechanisms might serve as pharmacological targets that may also play a role in subgroups of other, more common pain diseases. In other unusual pain disorders, the identification of pathomechanisms has already led to the development of new drugs. A completely new therapeutic approach, the gene silencing, can even stop progression in hereditary transthyretin amyloidosis and porphyria, ie in pain diseases that would otherwise be rapidly fatal if left untreated. Thus, pain therapists and researchers should be aware of these rare and unusual pain disorders as they offer the unique opportunity to study mechanisms, identify new druggable targets and finally because early diagnosis might save many patient lives.
Collapse
Affiliation(s)
- Juliane Sachau
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - Dilara Kersebaum
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - Anthony H Dickenson
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| |
Collapse
|
27
|
Qaswal AB, Ababneh O, Khreesha L, Al-Ani A, Suleihat A, Abbad M. Mathematical Modeling of Ion Quantum Tunneling Reveals Novel Properties of Voltage-Gated Channels and Quantum Aspects of Their Pathophysiology in Excitability-Related Disorders. PATHOPHYSIOLOGY 2021; 28:116-154. [PMID: 35366274 PMCID: PMC8830480 DOI: 10.3390/pathophysiology28010010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/08/2023] Open
Abstract
Voltage-gated channels are crucial in action potential initiation and propagation and there are many diseases and disorders related to them. Additionally, the classical mechanics are the main mechanics used to describe the function of the voltage-gated channels and their related abnormalities. However, the quantum mechanics should be considered to unravel new aspects in the voltage-gated channels and resolve the problems and challenges that classical mechanics cannot solve. In the present study, the aim is to mathematically show that quantum mechanics can exhibit a powerful tendency to unveil novel electrical features in voltage-gated channels and be used as a promising tool to solve the problems and challenges in the pathophysiology of excitability-related diseases. The model of quantum tunneling of ions through the intracellular hydrophobic gate is used to evaluate the influence of membrane potential and gating free energy on the tunneling probability, single channel conductance, and quantum membrane conductance. This evaluation is mainly based on graphing the mathematical relationships between these variables. The obtained mathematical graphs showed that ions can achieve significant quantum membrane conductance, which can affect the resting membrane potential and the excitability of cells. In the present work, quantum mechanics reveals original electrical properties associated with voltage-gated channels and introduces new insights and implications into the pathophysiology of excitability- related disorders. In addition, the present work sets a mathematical and theoretical framework that can be utilized to conduct experimental studies in order to explore the quantum aspects of voltage-gated channels and the quantum bioelectrical property of biological membranes.
Collapse
Affiliation(s)
- Abdallah Barjas Qaswal
- Department of Internship Program, Jordan University Hospital, The University of Jordan, Amman 11942, Jordan
| | - Omar Ababneh
- Department of Anesthesia and Intensive Care, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Lubna Khreesha
- Department of Special Surgery, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Abdallah Al-Ani
- School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Ahmad Suleihat
- Department of General Surgery, School of Medicine, The University of Jordan, Amman 11942, Jordan; (A.S.); (M.A.)
| | - Mutaz Abbad
- Department of General Surgery, School of Medicine, The University of Jordan, Amman 11942, Jordan; (A.S.); (M.A.)
| |
Collapse
|
28
|
Kerth CM, Hautvast P, Körner J, Lampert A, Meents JE. Phosphorylation of a chronic pain mutation in the voltage-gated sodium channel Nav1.7 increases voltage sensitivity. J Biol Chem 2021; 296:100227. [PMID: 33361158 PMCID: PMC7948457 DOI: 10.1074/jbc.ra120.014288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 12/17/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Mutations in voltage-gated sodium channels (Navs) can cause alterations in pain sensation, such as chronic pain diseases like inherited erythromelalgia. The mutation causing inherited erythromelalgia, Nav1.7 p.I848T, is known to induce a hyperpolarized shift in the voltage dependence of activation in Nav1.7. So far, however, the mechanism to explain this increase in voltage sensitivity remains unknown. In the present study, we show that phosphorylation of the newly introduced Thr residue explains the functional change. We expressed wildtype human Nav1.7, the I848T mutant, or other mutations in HEK293T cells and performed whole-cell patch-clamp electrophysiology. As the insertion of a Thr residue potentially creates a novel phosphorylation site for Ser/Thr kinases and because Nav1.7 had been shown in Xenopus oocytes to be affected by protein kinases C and A, we used different nonselective and selective kinase inhibitors and activators to test the effect of phosphorylation on Nav1.7 in a human system. We identify protein kinase C, but not protein kinase A, to be responsible for the phosphorylation of T848 and thereby for the shift in voltage sensitivity. Introducing a negatively charged amino acid instead of the putative phosphorylation site mimics the effect on voltage gating to a lesser extent. 3D modeling using the published cryo-EM structure of human Nav1.7 showed that introduction of this negatively charged site seems to alter the interaction of this residue with the surrounding amino acids and thus to influence channel function. These results could provide new opportunities for the development of novel treatment options for patients with chronic pain.
Collapse
Affiliation(s)
- Clara M Kerth
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Petra Hautvast
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Jannis Körner
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany; Department of Anesthesiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Jannis E Meents
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
29
|
Azizi SA. Monoamines: Dopamine, Norepinephrine, and Serotonin, Beyond Modulation, "Switches" That Alter the State of Target Networks. Neuroscientist 2020; 28:121-143. [PMID: 33292070 DOI: 10.1177/1073858420974336] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
How do monoamines influence the perceptual and behavioral aspects of brain function? A library of information regarding the genetic, molecular, cellular, and function of monoamines in the nervous system and other organs has accumulated. We briefly review monoamines' anatomy and physiology and discuss their effects on the target neurons and circuits. Monoaminergic cells in the brain stem receive inputs from sensory, limbic, and prefrontal areas and project extensively to the forebrain and hindbrain. We review selected studies on molecular, cellular, and electrophysiological effects of monoamines on the brain's target areas. The idea is that monoamines, by reversibly modulating the "primary" information processing circuits, regulate and switch the functions of brain networks and can reversibly alter the "brain states," such as consciousness, emotions, and movements. Monoamines, as the drivers of normal motor and sensory brain operations, including housekeeping, play essential roles in pathogenesis of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Sayed Ausim Azizi
- Department of Neurology, Global Neuroscience Institute, Chester, PA, USA
| |
Collapse
|
30
|
Smith CA, Paskhover B, Mammis A. Molecular mechanisms of trigeminal neuralgia: A systematic review. Clin Neurol Neurosurg 2020; 200:106397. [PMID: 33338828 DOI: 10.1016/j.clineuro.2020.106397] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To conduct a systematic review of the available literature for primary research articles identifying potential gene mutations, polymorphisms and other molecular regulatory mechanisms related to trigeminal neuralgia in order to identify the genetic and molecular models of primary trigeminal neuralgia currently being investigated. METHODS PubMed and Web of Science were systematically searched to identify primary research articles discussing genetic predictors of trigeminal neuralgia and neuropathic pain that were published prior to July 2020. This review was conducted according to PRISMA guidelines. RESULTS Out of the 333 articles originally identified, a total of 14 papers were selected for study inclusion. These articles included 5 human studies, 6 mouse studies and 3 rat studies. Four articles investigated sodium channels, 1 investigated a sodium channel and nerve growth factor receptor, 2 investigated potassium channels, 1 investigated calcium channels, 1 investigated the downstream regulatory element antagonist modulator protein, 1 investigated the dynorphin-kappa opioid receptor system, 1 investigated TRPA1, 1 investigated the Nrg1/ErbB3/ErbB2 signaling complex, 1 investigated a serotonin transporter and 1 investigated potassium channels, sodium channels, calcium channels, chloride channels, TRP channels and gap junctions. CONCLUSION Researchers have identified multiple genetic and molecular targets involved with potential pathophysiologies that have a relationship to the creation of trigeminal neuralgia. At this time, there does not seem to be clear causal frontrunner, demonstrating the possibility that genetic predisposition to trigeminal neuralgia may involve multiple genes and/or downstream products, such as ion channels.
Collapse
Affiliation(s)
- Cynthia A Smith
- Rutgers New Jersey Medical School, Department of Neurological Surgery, Newark, NJ, USA.
| | - Boris Paskhover
- Rutgers New Jersey Medical School, Department of Otolaryngology - Head & Neck Surgery, Newark, NJ, USA.
| | - Antonios Mammis
- NYU Grossman School of Medicine, Department of Neurosurgery, New York, NY, USA.
| |
Collapse
|
31
|
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
|
32
|
Slepukhina MA, Ivashchenko DV, Sheina MA, Muradian AA, Blagovestnov DA, Sychev DA. Pain pharmacogenetics. Drug Metab Pers Ther 2020; 35:dmpt-2020-2939. [PMID: 32776897 DOI: 10.1515/dmpt-2020-2939] [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: 10/15/2019] [Accepted: 03/16/2020] [Indexed: 11/15/2022]
Abstract
Pain is a significant problem in medicine. The use of PGx markers to personalize postoperative analgesia can increase its effectiveness and avoid undesirable reactions. This article describes the mechanisms of nociception and antinociception and shows the pathophysiological mechanisms of pain in the human body. The main subject of this article is pharmacogenetic approach to the selection of anesthetics. Current review presents data for local and general anesthetics, opioids, and non-steroidal anti-inflammatory drugs. None of the anesthetics currently has clinical guidelines for pharmacogenetic testing. This literature review summarizes the results of original research available, to date, and draws attention to this area.
Collapse
Affiliation(s)
| | - Dmitriy V Ivashchenko
- Child Psychiatry and Psychotherapy Department, Department of Personalized Medicine, Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - Maria A Sheina
- Department of Anesthesiology and Intensive Care, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | | | - Dmitriy A Sychev
- Department of Clinical Pharmacology and Therapeutics, Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| |
Collapse
|
33
|
Mitchell R, Mikolajczak M, Kersten C, Fleetwood-Walker S. ErbB1-dependent signalling and vesicular trafficking in primary afferent nociceptors associated with hypersensitivity in neuropathic pain. Neurobiol Dis 2020; 142:104961. [DOI: 10.1016/j.nbd.2020.104961] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/26/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
|
34
|
Identification of N-acyl amino acids that are positive allosteric modulators of glycine receptors. Biochem Pharmacol 2020; 180:114117. [PMID: 32579961 DOI: 10.1016/j.bcp.2020.114117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 01/31/2023]
Abstract
Glycine receptors (GlyRs) mediate inhibitory neurotransmission within the spinal cord and play a crucial role in nociceptive signalling. This makes them primary targets for the development of novel chronic pain therapies. Endogenous lipids have previously been shown to modulate glycine receptors and produce analgesia in pain models, however little is known about what chemical features mediate these effects. In this study, we characterised lipid modulation of GlyRs by screening a library of N-acyl amino acids across all receptor subtypes and determined chemical features crucial for their activity. Acyl-glycine's with a C18 carbon tail were found to produce the greatest potentiation, and require a cis double bond within the central region of the carbon tail (ω6 - ω9) to be active. At 1 µM, C18 ω6,9 glycine potentiated glycine induced currents in α3 and α3β receptors by over 50%, and α1, α2, α1β and α2β receptors by over 100%. C18 ω9 glycine (N-oleoyl glycine) significantly enhance glycine induced peak currents and cause a dose-dependent shift in the glycine concentration response. In the presence of 3 µM C18 ω9 glycine, the EC5o of glycine at the α1 receptor was reduced from 17 µM to 10 µM. This study has identified several acyl-amino acids which are positive allosteric modulators of GlyRs and make promising lead compounds for the development of novel chronic pain therapies.
Collapse
|
35
|
The role of Nav1.7 in human nociceptors: insights from human induced pluripotent stem cell-derived sensory neurons of erythromelalgia patients. Pain 2020; 160:1327-1341. [PMID: 30720580 PMCID: PMC6554007 DOI: 10.1097/j.pain.0000000000001511] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental Digital Content is Available in the Text. Human sodium channel NaV1.7 in induced pluripotent stem cell–derived sensory neurons sets the action potential threshold but does not support subthreshold depolarizations. The chronic pain syndrome inherited erythromelalgia (IEM) is attributed to mutations in the voltage-gated sodium channel (NaV) 1.7. Still, recent studies targeting NaV1.7 in clinical trials have provided conflicting results. Here, we differentiated induced pluripotent stem cells from IEM patients with the NaV1.7/I848T mutation into sensory nociceptors. Action potentials in these IEM nociceptors displayed a decreased firing threshold, an enhanced upstroke, and afterhyperpolarization, all of which may explain the increased pain experienced by patients. Subsequently, we investigated the voltage dependence of the tetrodotoxin-sensitive NaV activation in these human sensory neurons using a specific prepulse voltage protocol. The IEM mutation induced a hyperpolarizing shift of NaV activation, which leads to activation of NaV1.7 at more negative potentials. Our results indicate that NaV1.7 is not active during subthreshold depolarizations, but that its activity defines the action potential threshold and contributes significantly to the action potential upstroke. Thus, our model system with induced pluripotent stem cell–derived sensory neurons provides a new rationale for NaV1.7 function and promises to be valuable as a translational tool to profile and develop more efficacious clinical analgesics.
Collapse
|
36
|
Khammissa RAG, Ballyram R, Fourie J, Bouckaert M, Lemmer J, Feller L. Selected pathobiological features and principles of pharmacological pain management. J Int Med Res 2020; 48:300060520903653. [PMID: 32408839 PMCID: PMC7232056 DOI: 10.1177/0300060520903653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
Pain induced by inflammation and nerve injury arises from abnormal neural activity of primary afferent nociceptors in response to tissue damage, which causes long-term elevation of the sensitivity and responsiveness of spinal cord neurons. Inflammatory pain typically resolves following resolution of inflammation; however, nerve injury-either peripheral or central-may cause persistent neuropathic pain, which frequently manifests as hyperalgesia or allodynia. Neuralgias, malignant metastatic bone disease, and diabetic neuropathy are some of the conditions associated with severe, often unremitting chronic pain that is both physically and psychologically debilitating or disabling. Therefore, optimal pain management for patients with chronic neuropathic pain requires a multimodal approach that comprises pharmacological and psychological interventions. Non-opioid analgesics (e.g., paracetamol, aspirin, or other non-steroidal anti-inflammatory drugs) are first-line agents used in the treatment of mild-to-moderate acute pain, while opioids of increasing potency are indicated for the treatment of persistent, moderate-to-severe inflammatory pain. N-methyl D-aspartate receptor antagonists, antidepressants, anticonvulsants, or a combination of these should be considered for the treatment of chronic neuropathic pain. This review discusses the various neural signals that mediate acute and chronic pain, as well as the general principles of pain management.
Collapse
Affiliation(s)
- Razia Abdool Gafaar Khammissa
- Department of Periodontology and Oral Medicine, Sefako Makgatho
University, Pretoria, South Africa
- Department of Periodontics and Oral Medicine, University of
Pretoria, Pretoria, South Africa
| | - Raoul Ballyram
- Department of Periodontology and Oral Medicine, Sefako Makgatho
University, Pretoria, South Africa
| | - Jeanine Fourie
- Department of Periodontology and Oral Medicine, Sefako Makgatho
University, Pretoria, South Africa
| | - Michael Bouckaert
- Department of Maxillofacial and Oral Surgery, Sefako Makgatho
University, Pretoria, South Africa
| | - Johan Lemmer
- Department of Periodontology and Oral Medicine, Sefako Makgatho
University, Pretoria, South Africa
| | - Liviu Feller
- Department of Periodontology and Oral Medicine, Sefako Makgatho
University, Pretoria, South Africa
| |
Collapse
|
37
|
Cornish PB, Cornish AP. On the Origin of Pain – the ‘Pain Channel’ Hypothesis. Med Hypotheses 2020; 137:109576. [DOI: 10.1016/j.mehy.2020.109576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/01/2020] [Accepted: 01/16/2020] [Indexed: 10/25/2022]
|
38
|
Kushnarev M, Pirvulescu IP, Candido KD, Knezevic NN. Neuropathic pain: preclinical and early clinical progress with voltage-gated sodium channel blockers. Expert Opin Investig Drugs 2020; 29:259-271. [PMID: 32070160 DOI: 10.1080/13543784.2020.1728254] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Neuropathic pain is a chronic condition that significantly affects the quality of life of millions of people globally. Most of the pharmacologic treatments currently in use demonstrate modest efficacy and over half of all patients do not respond to medical management. Hence, there is a need for new, efficacious drugs. Evidence points toward voltage-gated sodium channels as a key target for novel analgesics.Area covered: The role of voltage-gated sodium channels in pain pathophysiology is illuminated and the preclinical and clinical data for new sodium channel blockers and toxin-derived lead compounds are examined. The expansion of approved sodium channel blockers is discussed along with the limitations of current research, trends in drug development, and the potential of personalized medicine.Expert opinion: The transition from preclinical to clinical studies can be difficult because of the inherent inability of animal models to express the complexities of pain states. Pain pathways are notoriously intricate and may be pharmacologically modulated at a variety of targets; it is unlikely that action at a single target could completely abolish a pain response because pain is rarely unifactorial. Combination therapy may be necessary and this could further confound the discovery of novel agents.
Collapse
Affiliation(s)
- Mikhail Kushnarev
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Iulia Paula Pirvulescu
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Kenneth D Candido
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA.,Department of Anesthesiology, College of Medicine, University of Illinois, Chicago, IL, USA.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA.,Department of Anesthesiology, College of Medicine, University of Illinois, Chicago, IL, USA.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| |
Collapse
|
39
|
Li N, Liu B, Wu W, Hong Y, Zhang J, Liu Y, Zhang M, Zhang X, Duan G. Upregulation of transcription factor 4 downregulates Na V1.8 expression in DRG neurons and prevents the development of rat inflammatory and neuropathic hypersensitivity. Exp Neurol 2020; 327:113240. [PMID: 32045596 DOI: 10.1016/j.expneurol.2020.113240] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/20/2020] [Accepted: 02/07/2020] [Indexed: 10/25/2022]
Abstract
The voltage sodium channel 1.8 (NaV1.8) in the dorsal root ganglion (DRG) neurons contributes to the initiation and development of chronic inflammatory and neuropathic pain. However, an effective intervention on NaV1.8 remains to be studied in pre-clinical research and clinical trials. In this study, we aimed to investigate whether transcription factor 4 (TCF4) overexpression represses NaV1.8 expression in DRG neurons, thus preventing the development of chronic pain. Using chromatin immunoprecipitation (CHIP), we verified the interaction of TCF4 and sodium voltage-gated channel alpha subunit 10A (SCN10A) enhancer in HEK293 cells and rat DRG neurons. Using a dual luciferase reporter assay, we confirmed the transcriptional inhibition of TCF4 on SCN10A promoter in vitro. To investigate the regulation of TCF4 on Nav1.8, we then upregulated TCF4 expression by intrathecally delivering an overexpression of recombinant adeno-associated virus (rAAV) in the Complete Freund's adjuvant (CFA)-induced inflammatory pain model and spared nerve injury (SNI)-induced neuropathic pain model. By using a quantitative polymerase chain reaction (qPCR), western blot, and immunostaining, we evaluated NaV1.8 expression after a noxious stimulation and the application of the TCF4 overexpression virus. We showed that the intrathecal delivery of TCF4 overexpression virus significantly repressed the increase of NaV1.8 and prevented the development of hyperalgesia in rats. Moreover, we confirmed the efficient role of an overexpressed TCF4 in preventing the CFA- and SNI-induced neuronal hyperexcitability by calcium imaging. Our results suggest that attenuating the dysregulation of NaV1.8 by targeting TCF4 may be a novel therapeutic strategy for chronic inflammatory and neuropathic pain.
Collapse
Affiliation(s)
- Ningbo Li
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China.; Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Baowen Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenyao Wu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yishun Hong
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jin Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mi Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xianwei Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China..
| | - Guangyou Duan
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China..
| |
Collapse
|
40
|
Jin Y, Cai S, Jiang Y, Zhong K, Wen C, Ruan Y, Chew LA, Khanna R, Xu Z, Yu J. Tetramethylpyrazine Reduces Epileptogenesis Progression in Electrical Kindling Models by Modulating Hippocampal Excitatory Neurotransmission. ACS Chem Neurosci 2019; 10:4854-4863. [PMID: 31756074 DOI: 10.1021/acschemneuro.9b00575] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Antiepileptic drugs (AEDs) are the primary agents prescribed for clinical management of limbic epilepsy. However, high incidence of pharmacoresistance and a limited armory of drugs for inhibiting the pathological progression of epilepsy pose major obstacles to managing epilepsy. Here, we investigated the effect of tetramethylpyrazine (TMP), the main bioactive alkaloid isolated from the oriental medicine Ligusticum chuanxiong Hort., against the epileptogenesis progression of acute hippocampal and corneal (6 Hz) electrical kindling models of TLE. TMP dose-dependently limited the progression of seizures and reduced the after-discharge duration (ADDs) in a hippocampal mouse kindling model. Mice treated with TMP (20, 50 mg/kg, i.p.) remained in stage 1 of epileptic progression for a protracted period, requiring additional stimulation to induce stages 2-5 epileptic phenotypes. TMP (50 mg/kg) also inhibited 6 Hz corneal kindling progression. In contrast, TMP did not reverse the phenotypes induced in a generalized seizures (GS) model, or the maximal electroshock (MES) or pentylenetetrazole (PTZ)-induced models of epilepsy. Furthermore, patch clamp recordings revealed no effect of TMP (10 μM) on CA1 hippocampal neurons' intrinsic properties but suppressed the (i) frequency of spontaneous excitatory post synaptic currents (sEPSCs), (ii) paired pulse ratio (PPR), and (iii) long-term potentiation (LTP) induction in the Schaffer collateral-CA1 pathway. TMP suppressed the activity of calcium, but not sodium, channels. Taken together, these results suggest that TMP has an antiepileptogenic effect, likely through suppression of excitatory synaptic transmission by its effects on inhibition of calcium channels; these traits distinguish TMP from currently available AEDs. As mice administered TMP did not show any neurologic impairment in the object recognition and open field tests, the data support further development of TMP as a promising treatment for epilepsy.
Collapse
Affiliation(s)
| | - Song Cai
- Department of Anatomy, Histology & Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen 518055, China
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, Arizona 85724, United States
| | | | - Kai Zhong
- Hangzhou Medical College, Hangzhou 310053, China
| | | | | | - Lindsey A. Chew
- School of Medicine, Duke University, Durham, North Carolina 27710, United States
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, Arizona 85724, United States
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, United States
| | | | | |
Collapse
|
41
|
Spider Knottin Pharmacology at Voltage-Gated Sodium Channels and Their Potential to Modulate Pain Pathways. Toxins (Basel) 2019; 11:toxins11110626. [PMID: 31671792 PMCID: PMC6891507 DOI: 10.3390/toxins11110626] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are a key determinant of neuronal signalling. Neurotoxins from diverse taxa that selectively activate or inhibit NaV channels have helped unravel the role of NaV channels in diseases, including chronic pain. Spider venoms contain the most diverse array of inhibitor cystine knot (ICK) toxins (knottins). This review provides an overview on how spider knottins modulate NaV channels and describes the structural features and molecular determinants that influence their affinity and subtype selectivity. Genetic and functional evidence support a major involvement of NaV subtypes in various chronic pain conditions. The exquisite inhibitory properties of spider knottins over key NaV subtypes make them the best lead molecules for the development of novel analgesics to treat chronic pain.
Collapse
|
42
|
de Greef BTA, Hoeijmakers JGJ, Geerts M, Oakes M, Church TJE, Waxman SG, Dib-Hajj SD, Faber CG, Merkies ISJ. Lacosamide in patients with Nav1.7 mutations-related small fibre neuropathy: a randomized controlled trial. Brain 2019; 142:263-275. [PMID: 30649227 DOI: 10.1093/brain/awy329] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/02/2018] [Indexed: 11/14/2022] Open
Abstract
Symptomatic treatment of neuropathic pain in small fibre neuropathy is often disappointing. The finding of voltage-gated sodium channel mutations in small fibre neuropathy (with mutations in SCN9A, encoding for Nav1.7) being most frequently reported suggest a specific target for therapy. The anticonvulsant lacosamide acts on Nav1.3, Nav1.7, and Nav1.8. The aim of this study was to evaluate the efficacy, safety, and tolerability of lacosamide as a potential treatment for pain in Nav1.7-related small fibre neuropathy. The Lacosamide-Efficacy-'N'-Safety in SFN (LENSS) was a randomized, placebo-controlled, double-blind, crossover-design study. Subjects were recruited in the Netherlands between November 2014 and July 2016. Patients with Nav1.7-related small fibre neuropathy were randomized to start with lacosamide followed by placebo or vice versa. In both 8-week treatment phases, patients received 200 mg two times a day (BID), preceded by a titration period, and ended by a tapering period. The primary outcome was efficacy, defined as the proportion of patients with 1-point average pain score reduction compared to baseline using the Pain Intensity Numerical Rating Scale. The trial is registered with ClinicalTrials.gov, number NCT01911975. Twenty-four subjects received lacosamide, and 23 received placebo. In 58.3% of patients receiving lacosamide, mean average pain decreased by at least 1 point, compared to 21.7% in the placebo group [sensitivity analyses, odds ratio 5.65 (95% confidence interval: 1.83-17.41); P = 0.0045]. In the lacosamide group, 33.3% reported that their general condition improved versus 4.3% in the placebo group (P-value = 0.0156). Additionally, a significant decrease in daily sleep interference, and in surface pain intensity was demonstrated. No significant changes in quality of life or autonomic symptoms were found. Lacosamide was well tolerated and safe in use. This study shows that lacosamide has a significant effect on pain, general wellbeing, and sleep quality. Lacosamide was well tolerated and safe, suggesting that it can be used for pain treatment in Nav1.7-related small fibre neuropathy.
Collapse
Affiliation(s)
- Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Margot Geerts
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Mike Oakes
- ParamStat Limited, University of Sussex, Fairlight, East Sussex, UK
| | | | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ingemar S J Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands.,Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| |
Collapse
|
43
|
Loke MF, Wei H, Yeo J, Sng BL, Sia AT, Tan EC. Deep sequencing analysis to identify novel and rare variants in pain-related genes in patients with acute postoperative pain and high morphine use. J Pain Res 2019; 12:2755-2770. [PMID: 31571979 PMCID: PMC6756825 DOI: 10.2147/jpr.s213869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/01/2019] [Indexed: 01/04/2023] Open
Abstract
Purpose Most of the genetic variants that are reported to be associated with common pain phenotypes and analgesic use are common polymorphisms. The objective of our study was to identify new variants and investigate less common genetic variants that are usually not included in either small single-gene studies or high-throughput genotyping arrays. Patients and methods From a cohort of 1075 patients who underwent a scheduled total abdominal hysterectomy, 92 who had higher self-rated pain scores and used more morphine were selected for the re-sequencing of 105 genes. Results We identified over 2400 variants in 104 genes. Most were intronic with frequencies >5%. There were 181 novel variants, of which 30 were located in exons: 17 nonsynonymous, 10 synonymous, 2 non-coding RNA, and 1 stop-gain. For known variants that are rare (population frequency <1%), the frequencies of 54 exonic variants and eight intronic variants for the sequenced samples were higher than the weighted frequencies in the Genome Aggregation Database for East and South Asians (P-values ranging from 0.000 to 0.046). Overall, patients who had novel and/or rare variants used more morphine than those who only had common variants. Conclusion Our study uncovered novel variants in patients who reported higher pain and used more morphine. Compared with the general population, rare variants were more common in this group.
Collapse
Affiliation(s)
- Mun-Fai Loke
- Research Laboratory, KK Women's & Children's Hospital, Singapore, Singapore
| | - Heming Wei
- Research Laboratory, KK Women's & Children's Hospital, Singapore, Singapore
| | - Junjie Yeo
- Duke-NUS Medical School, Singapore, Singapore
| | - Ban-Leong Sng
- Department of Women's Anaesthesia, KK Women's & Children's Hospital, Singapore, Singapore
| | - Alex T Sia
- Department of Women's Anaesthesia, KK Women's & Children's Hospital, Singapore, Singapore
| | - Ene-Choo Tan
- Research Laboratory, KK Women's & Children's Hospital, Singapore, Singapore
| |
Collapse
|
44
|
Vysokov N, McMahon SB, Raouf R. The role of Na V channels in synaptic transmission after axotomy in a microfluidic culture platform. Sci Rep 2019; 9:12915. [PMID: 31501450 PMCID: PMC6733904 DOI: 10.1038/s41598-019-49214-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 08/20/2019] [Indexed: 01/07/2023] Open
Abstract
Voltage gated sodium channels are key players in aberrant pain signaling and sensitization of nociceptors after peripheral nerve injury. The extent to which sodium channel activity after injury contributes to synaptic transmission at the first pain synapse however remains unclear. To investigate the effect of axotomy on synaptic transmission between dorsal root ganglia neurons and dorsal horn neurons, we reconstructed the first pain synapse in a novel microfluidic based compartmentalized cell culture system, which recapitulates the connectivity of peripheral pain signaling. We show that following axotomy of the distal axons, inhibition of NaV1.7 and NaV1.8 sodium channels in incoming presynaptic DRG axons is no longer sufficient to block activation of these axons and the resulting synaptic transmission to dorsal horn neurons. We found that blockade of NaV1.6 activity is highly effective in reducing activation of incoming axons contributing to synaptic transmission after axotomy of DRG neurons. The microfluidic culture system described here offers an in vitro platform to recapitulate and study the first pain synapse.
Collapse
Affiliation(s)
- Nickolai Vysokov
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Ramin Raouf
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom.
| |
Collapse
|
45
|
Effraim PR, Huang J, Lampert A, Stamboulian S, Zhao P, Black JA, Dib-Hajj SD, Waxman SG. Fibroblast growth factor homologous factor 2 (FGF-13) associates with Nav1.7 in DRG neurons and alters its current properties in an isoform-dependent manner. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100029. [PMID: 31223136 PMCID: PMC6565799 DOI: 10.1016/j.ynpai.2019.100029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 11/29/2022]
Abstract
Fibroblast Growth Factor Homologous Factors (FHF) constitute a subfamily of FGF proteins with four prototypes (FHF1-4; also known as FGF11-14). FHF proteins have been shown to bind directly to the membrane-proximal segment of the C-terminus in voltage-gated sodium channels (Nav), and regulate current density, availability, and frequency-dependent inhibition of sodium currents. Members of the FHF2 subfamily, FHF2A and FHF2B, differ in the length and sequence of their N-termini, and, importantly, differentially regulate Nav1.6 gating properties. Using immunohistochemistry, we show that FHF2 isoforms are expressed in adult dorsal root ganglion (DRG) neurons where they co-localize with Nav1.6 and Nav1.7. FHF2A and FHF2B show differential localization in neuronal compartments in DRG neurons, and levels of expression of FHF2 factors are down-regulated following sciatic nerve axotomy. Because Nav1.7 in nociceptors plays a critical role in pain, we reasoned that its interaction with FHF2 isoforms might regulate its current properties. Using whole-cell patch clamp in heterologous expression systems, we show that the expression of FHF2A in HEK293 cell line stably expressing Nav1.7 channels causes no change in activation, whereas FHF2B depolarizes activation. Both FHF2 isoforms depolarize fast-inactivation. Additionally, FHF2A causes an accumulation of inactivated channels at all frequencies tested due to a slowing of recovery from inactivation, whereas FHF2B has little effect on these properties of Nav1.7. Measurements of the Nav1.7 current in DRG neurons in which FHF2 levels are knocked down confirmed the effects of FHF2A on repriming, and FHF2B on activation, however FHF2A and B did not have an effect on fast inactivation. Our data demonstrates that FHF2 does indeed regulate the current properties of Nav1.7 and does so in an isoform and cell-specific manner.
Collapse
Affiliation(s)
- Philip R. Effraim
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Jianying Huang
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Severine Stamboulian
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Joel A. Black
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
46
|
Abstract
Acute pain is adaptive, but chronic pain is a global challenge. Many chronic pain syndromes are peripheral in origin and reflect hyperactivity of peripheral pain-signaling neurons. Current treatments are ineffective or only partially effective and in some cases can be addictive, underscoring the need for better therapies. Molecular genetic studies have now linked multiple human pain disorders to voltage-gated sodium channels, including disorders characterized by insensitivity or reduced sensitivity to pain and others characterized by exaggerated pain in response to normally innocuous stimuli. Here, we review recent developments that have enhanced our understanding of pathophysiological mechanisms in human pain and advances in targeting sodium channels in peripheral neurons for the treatment of pain using novel and existing sodium channel blockers.
Collapse
Affiliation(s)
- Sulayman D Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Rehabilitation Research Center, Veterans Affairs, Connecticut Healthcare System, West Haven, Connecticut 06516, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Rehabilitation Research Center, Veterans Affairs, Connecticut Healthcare System, West Haven, Connecticut 06516, USA
| |
Collapse
|
47
|
Haberberger RV, Barry C, Dominguez N, Matusica D. Human Dorsal Root Ganglia. Front Cell Neurosci 2019; 13:271. [PMID: 31293388 PMCID: PMC6598622 DOI: 10.3389/fncel.2019.00271] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Sensory neurons with cell bodies situated in dorsal root ganglia convey information from external or internal sites of the body such as actual or potential harm, temperature or muscle length to the central nervous system. In recent years, large investigative efforts have worked toward an understanding of different types of DRG neurons at transcriptional, translational, and functional levels. These studies most commonly rely on data obtained from laboratory animals. Human DRG, however, have received far less investigative focus over the last 30 years. Nevertheless, knowledge about human sensory neurons is critical for a translational research approach and future therapeutic development. This review aims to summarize both historical and emerging information about the size and location of human DRG, and highlight advances in the understanding of the neurochemical characteristics of human DRG neurons, in particular nociceptive neurons.
Collapse
Affiliation(s)
- Rainer Viktor Haberberger
- Pain and Pulmonary Neurobiology Laboratory, Centre for Neuroscience, Anatomy and Histology, Flinders University, Adelaide, SA, Australia.,Órama Institute, Flinders University, Adelaide, SA, Australia
| | - Christine Barry
- Pain and Pulmonary Neurobiology Laboratory, Centre for Neuroscience, Anatomy and Histology, Flinders University, Adelaide, SA, Australia
| | - Nicholas Dominguez
- Pain and Pulmonary Neurobiology Laboratory, Centre for Neuroscience, Anatomy and Histology, Flinders University, Adelaide, SA, Australia
| | - Dusan Matusica
- Pain and Pulmonary Neurobiology Laboratory, Centre for Neuroscience, Anatomy and Histology, Flinders University, Adelaide, SA, Australia.,Órama Institute, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
48
|
Li M, Zhang SJ, Yang L, Fang XL, Hu HF, Zhao MY, Li L, Guo YY, Shao JP. Voltage-gated sodium channel 1.7 expression decreases in dorsal root ganglia in a spinal nerve ligation neuropathic pain model. Kaohsiung J Med Sci 2019; 35:493-500. [PMID: 31087766 DOI: 10.1002/kjm2.12088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 04/30/2019] [Indexed: 02/06/2023] Open
Abstract
The role of the voltage-gated sodium channel 1.7 (Nav1.7) is unclear in models of neuropathic pain induced by nerve injury. In the present study, we measured expression levels of Nav1.7 in two distinct neuropathic pain models: spinal nerve ligation (SNL) and chronic constriction injury (CCI). In the SNL model, both mRNA and protein levels of Nav1.7 were markedly lower in the L5 dorsal root ganglia (DRG) but were significantly higher in the L4 DRG. Nav1.7 protein levels were notably higher in both L4 and L5 DRGs under CCI conditions. We found that excessive damage of L5 nerves such as SNL reduced expression levels of Nav1.7 in the injured L5 DRG and activated the adjacent uninjured DRG, resulting in Nav1.7 level increases in the adjacent L4 DRG. We confirmed again that Nav1.7 was closely related to neuropathic pain induced by nerve injury. More importantly, our results suggest that tracing the molecular changes exclusively in the L5 DRG in SNL model may not completely explain the pain mechanism; it is necessary to study the adjacent uninjured L4 DRG.
Collapse
Affiliation(s)
- Ming Li
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Shao-Jin Zhang
- School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Lin Yang
- School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Xiao-Lei Fang
- School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Hao-Fan Hu
- School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Ming-Yue Zhao
- School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Lei Li
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Yan-Yan Guo
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Jin-Ping Shao
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
49
|
Pediatric Erythromelalgia and SCN9A Mutations: Systematic Review and Single-Center Case Series. J Pediatr 2019; 206:217-224.e9. [PMID: 30416015 DOI: 10.1016/j.jpeds.2018.10.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/07/2018] [Accepted: 10/11/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To evaluate the clinical features of erythromelalgia in childhood associated with gain-of-function SCN9A mutations that increase activity of the Nav1.7 voltage-gated sodium channel, we conducted a systematic review of pediatric presentations of erythromelalgia related to SCN9A mutations, and compared pediatric clinical presentations of symptomatic erythromelalgia, with or without SCN9A mutations. STUDY DESIGN PubMed, Embase, and PsycINFO Databases were searched for reports of inherited erythromelalgia in childhood. Clinical features, management, and genotype were extracted. Case notes of pediatric patients with erythromelalgia from the Great Ormond Street Hospital Pain Service were reviewed for clinical features, patient-reported outcomes, and treatments. Children aged over 10 years were recruited for quantitative sensory testing. RESULTS Twenty-eight publications described erythromelalgia associated with 15 different SCN9A gene variants in 25 children. Pain was severe and often refractory to multiple treatments, including nonspecific sodium channel blockers. Skin damage or other complications of cold immersion for symptomatic relief were common (60%). SCN9A mutations resulting in greater hyperpolarizing shifts in Nav1.7 sodium channels correlated with symptom onset at younger ages (P = .016). Variability in reporting, and potential publication bias toward severe cases, limit any estimations of overall prevalence. In our case series, symptoms were similar but comorbidities were more common in children with SCN9A mutations. Quantitative sensory testing revealed marked dynamic warm allodynia. CONCLUSIONS Inherited erythromelalgia in children is associated with difficult-to-manage pain and significant morbidity. Standardized reporting of outcome and management in larger series will strengthen identification of genotype-phenotype relationships. More effective long-term therapies are a significant unmet clinical need.
Collapse
|
50
|
A Holistic Approach to Pain Management in the Rheumatic Diseases. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2019. [DOI: 10.1007/s40674-019-00116-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|