1
|
Shin SM, Itson-Zoske B, Xu H, Xiang H, Fan F, Hogan QH, Yu H. Sensory neuron-specific block of multifaceted sodium channels mitigates neuropathic pain behaviors of osteoarthritis. Pain Rep 2025; 10:e1288. [PMID: 40444021 PMCID: PMC12119052 DOI: 10.1097/pr9.0000000000001288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 06/02/2025] Open
Abstract
Objective Multiple voltage-gated sodium channels (NaVs) in the peripheral sensory neurons (PSNs) regulate action potentials and their dysfunction contributes to the pain pathogenesis of osteoarthritis (OA). A combined block of multiple NaV subtypes selectively in the PSNs may, therefore, represent an effective analgesic approach in OA painful neuropathy. Methods To test this hypothesis, we generated recombinant adeno-associated virus (AAV) encoding a potent NaV inhibitory peptide aptamer, termed NaViPA1, that has a multipronged feature of inhibiting tetrodotoxin-sensitive NaV1.7, 1.6, 1.1, and 1.3, characterized in our recent report. Adeno-associated virus-encoded NaViPA1 was delivered into the ipsilateral lumbar 4/5 dorsal root ganglia of rats 2 weeks after induction of knee monoiodoacetate-OA (MIA-OA) and evoked and spontaneous sensory behaviors were followed in 6 weeks. Results Expression of NaViPA1 selective in the PSNs produced significant and comparable mitigations of evoked and spontaneous pain behavior and reversal of weight-bearing asymmetry in both male and female MIA-OA rats. Whole-cell current-clamp recordings showed that AAV-mediated NaViPA1 expression normalized action potential firing of the PSNs from MIA animals, suggesting that NaViPA1 attenuated pain behavior by, at least in part, reversing neuronal hyperexcitability. Conclusion Together, these results support that (1) NaVs in peripheral sensory pathways contribute to MIA-OA pain pathogenesis and (2) NaViPA1 is a promising analgesic lead that, combined with AAV-targeted delivery to pathological sensory ganglia, may be a viable peripherally selective PSN-targeting strategy in mitigating chronic MIA-OA pain behaviors.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hao Xu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongfei Xiang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fan Fan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
2
|
Bavencoffe A, Lopez ER, Johnson KN, Tian J, Gorgun FM, Shen BQ, Domagala DM, Zhu MX, Dessauer CW, Walters ET. Widespread hyperexcitability of nociceptor somata outlasts enhanced avoidance behavior after incision injury. Pain 2025; 166:1088-1104. [PMID: 39432803 PMCID: PMC12003080 DOI: 10.1097/j.pain.0000000000003443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 09/05/2024] [Indexed: 10/23/2024]
Abstract
ABSTRACT Nociceptors with somata in dorsal root ganglia (DRGs) readily switch from an electrically silent state to a hyperactive state of tonic, nonaccommodating, low-frequency, irregular discharge of action potentials (APs). Spontaneous activity (SA) during this state is present in vivo in rats months after spinal cord injury (SCI) and has been causally linked to SCI pain. Intrinsically generated SA and, more generally, ongoing activity (OA) are induced by various neuropathic conditions in rats, mice, and humans and are retained in nociceptor somata after dissociation and culturing, providing a powerful tool for investigating its mechanisms and functions. The present study shows that long-lasting hyperexcitability that can generate OA during modest depolarization in probable nociceptors dissociated from DRGs of male and female rats is induced by plantar incision injury. OA occurred when the soma was artificially depolarized to a level within the normal range of membrane potentials where large, transient depolarizing spontaneous fluctuations (DSFs) can approach AP threshold. This hyperexcitability persisted for at least 3 weeks, whereas behavioral indicators of affective pain-hind paw guarding and increased avoidance of a noxious substrate in an operant conflict test-persisted for 1 week or less. The most consistent electrophysiological alteration associated with OA was enhancement of DSFs. An unexpected discovery after plantar incisions was hyperexcitability in neurons from thoracic DRGs that innervate dermatomes distant from the injured tissue. Potential in vivo functions of widespread, low-frequency nociceptor OA consistent with these and other findings are to contribute to hyperalgesic priming and to drive anxiety-related hypervigilance.
Collapse
Affiliation(s)
- Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Elia R. Lopez
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Kayla N. Johnson
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Falih M. Gorgun
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Breanna Q. Shen
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Drue M. Domagala
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| |
Collapse
|
3
|
Zhang J, Jiao H. Enhancing precision in pain management for inflammatory arthritis: exploring the potential role of lidocaine in pain mechanism differentiation. Pain 2025; 166:1213. [PMID: 40237774 DOI: 10.1097/j.pain.0000000000003581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Affiliation(s)
| | - Hongwei Jiao
- Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
4
|
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; 24:358-378. [PMID: 39901031 DOI: 10.1038/s41573-024-01108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
5
|
Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Gabriel KA, Palomino S, Li Y, Uhelski ML, Shiers S, Franco-Enzástiga Ú, Wangzhou A, Lesnak JB, Bandaru S, Shrivastava A, Inturi N, Albrecht PJ, Dockum M, Cervantes AM, Horton P, Funk G, North RY, Tatsui CE, Corrales G, Yousuf MS, Curatolo M, Gereau RW, Patwardhan A, Dussor G, Dougherty PM, Rice FL, Price TJ. Expansion of OSMR expression and signaling in the human dorsal root ganglion links OSM to neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645611. [PMID: 40236060 PMCID: PMC11996445 DOI: 10.1101/2025.03.26.645611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
RNA sequencing studies on human dorsal root ganglion (hDRG) from patients suffering from neuropathic pain show upregulation of OSM, linking this IL-6 family cytokine to pain disorders. In mice, however, OSM signaling causes itch behaviors through a direct effect on its cognate receptor expressed uniquely by pruriceptive sensory neurons. We hypothesized that an expansion in function of OSM-OSM receptor (OSMR) in sensory disorders in humans could be explained by species differences in receptor expression and signaling. Our in situ hybridization and immunohistochemical findings demonstrate broad expression of OSMR in DRG nociceptors and afferent fibers innervating the superficial and deep skin of humans. In patch-clamp electrophysiology, OSM directly activates human sensory neurons engaging MAPK signaling to promote action potential firing. Using CRISPR editing we show that OSM activation of MAPK signaling is dependent on OSMR and not LIFR in hDRG. Bulk, single-nuclei, and single-cell RNA-seq of OSM-treated hDRG cultures reveal expansive similarities in the transcriptomic signature observed in pain DRGs from neuropathic patients, indicating that OSM alone can orchestrate transcriptomic signatures associated with pain. We conclude that OSM-OSMR signaling via MAPKs is a critical signaling factor for DRG plasticity that may underlie neuropathic pain in patients.
Collapse
|
6
|
Hanani M. How Do Peripheral Neurons and Glial Cells Participate in Pain Alleviation by Physical Activity? Cells 2025; 14:462. [PMID: 40136711 PMCID: PMC11941599 DOI: 10.3390/cells14060462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/21/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Chronic pain is a global health problem with major socioeconomic implications. Drug therapy for chronic pain is limited, prompting search for non-pharmacological treatments. One such approach is physical exercise, which has been found to be beneficial for numerous health issues. Research in recent years has yielded considerable evidence for the analgesic actions of exercise in humans and experimental animals, but the underlying mechanisms are far from clear. It was proposed that exercise influences the pain pathways by interacting with the immune system, mainly by reducing inflammatory responses, but the release of endogenous analgesic mediators is another possibility. Exercise acts on neurons and glial cells in both the central and peripheral nervous systems. This review focuses on the periphery, with emphasis on possible glia-neuron interactions. Key topics include interactions of Schwann cells with axons (myelinated and unmyelinated), satellite glial cells in sensory ganglia, enteric glial cells, and the sympathetic nervous system. An attempt is made to highlight several neurological diseases that are associated with pain and the roles that glial cells may play in exercise-induced pain alleviation. Among the diseases are fibromyalgia and Charcot-Marie-Tooth disease. The hypothesis that active skeletal muscles exert their effects on the nervous system by releasing myokines is discussed.
Collapse
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem 91240, Israel;
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
7
|
Farah A, Patel R, Poplawski P, Wastie BJ, Tseng M, Barry AM, Daifallah O, Dubb A, Paul I, Cheng HL, Feroz F, Su Y, Chan M, Zeilhofer HU, Price TJ, Bennett DL, Bannister K, Dawes JM. A role for leucine-rich, glioma inactivated 1 in regulating pain sensitivity. Brain 2025; 148:1001-1014. [PMID: 39301592 PMCID: PMC11884686 DOI: 10.1093/brain/awae302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/19/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
Neuronal hyperexcitability is a key driver of persistent pain states, including neuropathic pain. Leucine-rich, glioma inactivated 1 (LGI1) is a secreted protein known to regulate excitability within the nervous system and is the target of autoantibodies from neuropathic pain patients. Therapies that block or reduce antibody levels are effective at relieving pain in these patients, suggesting that LGI1 has an important role in clinical pain. Here we have investigated the role of LGI1 in regulating neuronal excitability and pain-related sensitivity by studying the consequences of genetic ablation in specific neuron populations using transgenic mouse models. LGI1 has been well studied at the level of the brain, but its actions in the spinal cord and peripheral nervous system are poorly understood. We show that LGI1 is highly expressed in dorsal root ganglion (DRG) and spinal cord dorsal horn neurons in both mouse and human. Using transgenic mouse models, we genetically ablated LGI1, either specifically in nociceptors (LGI1fl/Nav1.8+) or in both DRG and spinal neurons (LGI1fl/Hoxb8+). On acute pain assays, we found that loss of LGI1 resulted in mild thermal and mechanical pain-related hypersensitivity when compared with littermate controls. In LGI1fl/Hoxb8+ mice, we found loss of Kv1 currents and hyperexcitability of DRG neurons. LGI1fl/Hoxb8+ mice displayed a significant increase in nocifensive behaviours in the second phase of the formalin test (not observed in LGI1fl/Nav1.8+ mice), and extracellular recordings in LGI1fl/Hoxb8+ mice revealed hyperexcitability in spinal dorsal horn neurons, including enhanced wind-up. Using the spared nerve injury model, we found that LGI1 expression was dysregulated in the spinal cord. LGI1fl/Nav1.8+ mice showed no differences in nerve injury-induced mechanical hypersensitivity, brush-evoked allodynia or spontaneous pain behaviour compared with controls. However, LGI1fl/Hoxb8+ mice showed a significant exacerbation of mechanical hypersensitivity and allodynia. Our findings point to effects of LGI1 at the level of both the DRG and the spinal cord, including an important impact of spinal LGI1 on pathological pain. Overall, we find a novel role for LGI1 with relevance to clinical pain.
Collapse
Affiliation(s)
- Adham Farah
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Ryan Patel
- Wolfson Sensory, Pain & Regeneration Centre, Guy’s Campus, Kings College London, London SE1 1UL, UK
| | - Piotr Poplawski
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Benjamin J Wastie
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Mandy Tseng
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Allison M Barry
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, Department of Neuroscience, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Omar Daifallah
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Akash Dubb
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Ivan Paul
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Hoi lao Cheng
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Faisal Feroz
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Yuhe Su
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Marva Chan
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - Theodore J Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, Department of Neuroscience, University of Texas at Dallas, Richardson, TX 75080, USA
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Kirsty Bannister
- Wolfson Sensory, Pain & Regeneration Centre, Guy’s Campus, Kings College London, London SE1 1UL, UK
| | - John M Dawes
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
8
|
Guo W, Yang H, Wang Y, Liu T, Pan Y, Chen X, Xu Q, Zhao D, Shan Z, Cai S. Small-molecule natural product sophoricoside reduces peripheral neuropathic pain via directly blocking of NaV1.6 in dorsal root ganglion nociceptive neurons. Neuropsychopharmacology 2025; 50:662-672. [PMID: 39414988 PMCID: PMC11845512 DOI: 10.1038/s41386-024-01998-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 10/18/2024]
Abstract
Peripheral neuropathic pain poses a significant global health challenge. Current drugs for peripheral neuropathic pain often fall short in efficacy or come with severe side effects, emphasizing the critical need for the development of highly effective and well-tolerated alternatives. Sophoricoside (SOP) is a nature product-derived isoflavone that possesses various pharmacological effects on inflammatory and neuropathy diseases. Here, in this study, analgesic effect was investigated by intrathecally administration of SOP/vehicle to spared nerve injury (SNI) or paclitaxel-induced peripheral neuropathic pain (PINP) rodent models, and mechanical allodynia was measured in Von Frey tests. Ipsilateral L4-L6 dorsal root ganglia (DRG) were used for protein expression. In silico molecular docking analysis was applied for assessing compound-target binding affinity. Primary cultured DRG neurons were utilized to investigate SOP's effect on veratridine-triggered nociceptor activities and its selective inhibition of voltage-gated sodium channels subtype 1.6 (NaV1.6). The results showed SOP treatment alleviated mechanical allodynia in SNI and PINP rodent models (paw withdrawal threshold after 1 h of injection: SNI-vehicle: 1.385 ± 0.338 g; SNI-SOP: 9.963 ± 2.029 g, P < 0.001; PINP-vehicle: 5.040 ± 0.985 g; PINP-SOP: 8.287 ± 3.812 g, P = 0.004). SOP presented effects on both inhibiting veratridine-triggered nociceptor activities (oscillatory population: vehicle: 39.9 ± 7.3%; SOP: 30.7 ± 9.8%, P = 0.021) and selectively blocking NaV1.6 in DRG sensory neurons. Molecular docking analysis indicated direct binding between SOP and NaV1.6, leading to its endocytosis in DRG Sensory Neurons. In conclusion, SOP alleviated nociceptive allodynia induced by peripheral nerve injury via selectively blocking of NaV1.6 in DRG nociceptive neurons. we highlight its potential as an analgesic and elucidate its mechanism involving NaV1.6 endocytosis. This research opens avenues for exploring the analgesic effects of SOP and its potential impact on neuropathic pain therapy.
Collapse
Affiliation(s)
- Weijie Guo
- Health Science Center, Shenzhen University, Shenzhen, China
| | - Haoyi Yang
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yuwei Wang
- Health Science Center, Shenzhen University, Shenzhen, China
| | - Tao Liu
- Health Science Center, Shenzhen University, Shenzhen, China
| | - Yunping Pan
- Department of Periodontology & Oral Mucosa, Shenzhen Stomatology Hospital, Shenzhen, China
| | - Xiying Chen
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Qiuyin Xu
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Dizhou Zhao
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Zhiming Shan
- Department of Anesthesiology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Song Cai
- Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
9
|
Kastelik J, Schwerdtfeger K, Stolle A, Schäfer M, Tafelski S. [Systematic review of the effectiveness of local anaesthetics in the treatment of neuropathic pain or phantom pain]. DIE ANAESTHESIOLOGIE 2025; 74:128-135. [PMID: 39992390 PMCID: PMC11876198 DOI: 10.1007/s00101-025-01500-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/31/2024] [Accepted: 11/24/2024] [Indexed: 02/25/2025]
Abstract
BACKGROUND Chronic pain is still a relevant medical and socioeconomic problem. The treatment focuses not only on pain reduction but also on functional treatment goals. Neuropathic pain includes biological, social and psychological aspects. In September 2023, the updated S3 guidelines for the management of peripheral nerve injuries were published. Multimodal pain management strategies encompassing systemic and local pharmacological, physiotherapeutic and occupational therapeutic interventions, are part of the guidelines. A central question addressed the widely debated treatment option using perineural local anaesthetics. OBJECTIVE The aim of the study was to evaluate the effectiveness of local anaesthetic infiltration in the treatment of neuropathic pain following nerve injuries through a systematic literature review and evaluation of the evidence by a meta-analysis. MATERIAL AND METHODS After formulating a PICO (patient/population, intervention, comparison and outcomes) question (Infobox 1) within the guideline group, a selective literature analysis of controlled trials in databases (PubMed, Cochrane Central Register of Controlled Trials-CENTRAL) was conducted until 31 July 2023. The literature was assessed by two reviewers and systematic reviews were examined for additional references. The studies were assessed using the Risk of Bias Tool 2.0 of the Cochrane Collaboration for randomized trials and the evidence was classified according to the GRADE (Grading of Recommendations, Assessment, Development and Evaluation) system. RESULTS A total of 357 publications were identified in the literature search. After removing duplicates (n = 15) 327 publications were evaluated. The literature analysis showed heterogeneity with respect to the pain localization, local anaesthetics and reported outcomes. In an in-depth literature analysis one relevant study was identified and included in the evaluation of the evidence. This study enrolled and randomized 144 patients between December 2013 and October 2018 and evaluated the effectivity of the continuous infusion of local anaesthetics (lidocaine 2% with epinephrine 2.5 µg/ml as an initial bolus in both study groups followed by an infusion of ropivacaine 0.5% in the intervention group over 6 days) on the intensity of the phantom pain in comparison to the placebo group with a continuous infusion of saline over 6 days. The mean pain intensity and pain-related dysfunctions were reduced in the intervention group after 4 weeks. In the intervention group 25 patients and in the placebo group 40 patients received the crossover treatment after 4 weeks. CONCLUSION Infiltration with local anaesthetics represents a potential therapeutic option for neuropathic pain and phantom pain after amputations. A randomized, blinded, placebo-controlled study from 2021 demonstrated lower pain intensity and a reduction in pain-related functional limitations after 4 weeks of continuous perineural local anesthetic infiltration. Further studies are necessary to establish a higher level of evidence regarding the effectiveness of minimally invasive pain treatment with local anaesthetics. In particular, long-term follow-up is necessary to be able to draw conclusions with respect to the analgesic efficacy of infiltration with local anaesthetics.
Collapse
Affiliation(s)
- Joanna Kastelik
- Klinik für Anästhesiologie und Intensivmedizin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Deutschland.
- Schmerzmedizin Campus Charité Mitte, Klinik für Anästhesiologie und Intensivmedizin, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland.
| | - Karsten Schwerdtfeger
- Klinik für Neurochirurgie, Medizinische Fakultät der Universität des Saarlandes, 66421, Homburg (Saar), Deutschland
| | - Annette Stolle
- Andreas Wentzensen Forschungsinstitut, BG Klinik Ludwigshafen, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Deutschland
| | - Michael Schäfer
- Klinik für Anästhesiologie und Intensivmedizin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Deutschland
| | - Sascha Tafelski
- Klinik für Anästhesiologie und Intensivmedizin, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Deutschland.
- Schmerzmedizin Campus Charité Mitte, Klinik für Anästhesiologie und Intensivmedizin, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland.
| |
Collapse
|
10
|
Attal N, Branders S, Pereira A, Bouhassira D. Prediction of the response to repetitive transcranial magnetic stimulation of the motor cortex in peripheral neuropathic pain and validation of a new algorithm. Pain 2025; 166:34-41. [PMID: 38875120 DOI: 10.1097/j.pain.0000000000003297] [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: 02/06/2024] [Accepted: 05/03/2024] [Indexed: 06/16/2024]
Abstract
CLINICAL TRIAL REGISTRATION NCT02010281.
Collapse
Affiliation(s)
- Nadine Attal
- INSERM U987, APHP, UVSQ Paris SACLAY University, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | | | | | - Didier Bouhassira
- INSERM U987, APHP, UVSQ Paris SACLAY University, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| |
Collapse
|
11
|
Stratton H, Lee G, Dolatyari M, Ghetti A, Cotta T, Mitchell S, Yue X, Ibrahim M, Dumaire N, Salih L, Moutal A, François-Moutal L, Martin L, Navratilova E, Porreca F. Nociceptors are functionally male or female: from mouse to monkey to man. Brain 2024; 147:4280-4291. [PMID: 38829801 PMCID: PMC11629683 DOI: 10.1093/brain/awae179] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
The prevalence of many pain conditions often differs between sexes. In addition to such quantitative distinctions, sexual dimorphism may also be qualitative reflecting differences in mechanisms that promote pain in men and women. A major factor that influences the likelihood of pain perception is the threshold for activation of nociceptors. Peripheral nociceptor sensitization has been demonstrated to be clinically relevant in many pain conditions. Whether peripheral nociceptor sensitization can occur in a sexually dimorphic fashion, however, has not been extensively studied. To address this fundamental knowledge gap, we used patch clamp electrophysiology to evaluate the excitability of dorsal root ganglion neurons from male or female rodents, non-human primates, and humans following exposure to putative sensitizing agents. Previous studies from our laboratory, and others, have shown that prolactin promotes female-selective pain responses in rodents. Consistent with these observations, dorsal root ganglion neurons from female, but not male, mice were selectively sensitized by exposure to prolactin. The sensitizing action of prolactin was also confirmed in dorsal root ganglion neurons from a female macaque monkey. Critically, neurons recovered from female, but not male, human donors were also selectively sensitized by prolactin. In the course of studies of sleep and pain, we unexpectedly observed that an orexin antagonist could normalize pain responses in male animals. We found that orexin B produced sensitization of male, but not female, mouse, macaque, and human dorsal root ganglion neurons. Consistent with functional responses, increased prolactin receptor and orexin receptor 2 expression was observed in female and male mouse dorsal root ganglia, respectively. Immunohistochemical interrogation of cultured human sensory neurons and whole dorsal root ganglia also suggested increased prolactin receptor expression in females and orexin receptor 2 expression in males. These data reveal a functional double dissociation of nociceptor sensitization by sex, which is conserved across species and is likely directly relevant to human pain conditions. To our knowledge, this is the first demonstration of functional sexual dimorphism in human sensory neurons. Patient sex is currently not a common consideration for the choice of pain therapy. Precision medicine, based on patient sex could improve therapeutic outcomes by selectively targeting mechanisms promoting pain in women or men. Additional implications of these findings are that the design of clinical trials for pain therapies should consider the proportions of male or female patients enrolled. Lastly, re-examination of selected past failed clinical trials with subgroup analysis by sex may be warranted.
Collapse
Affiliation(s)
- Harrison Stratton
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | - Grace Lee
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | - Mahdi Dolatyari
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | | | | | - Stefanie Mitchell
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | - Xu Yue
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | - Mohab Ibrahim
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
- Department of Anesthesiology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | - Nicolas Dumaire
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, MO 63104, USA
| | - Lyuba Salih
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, MO 63104, USA
| | - Aubin Moutal
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, MO 63104, USA
| | | | - Laurent Martin
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
- Department of Anesthesiology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | - Edita Navratilova
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | - Frank Porreca
- Department of Pharmacology, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| |
Collapse
|
12
|
Sodmann A, Degenbeck J, Aue A, Schindehütte M, Schlott F, Arampatzi P, Bischler T, Schneider M, Brack A, Monoranu CM, Gräfenhan T, Bohnert M, Pham M, Antoniadis G, Blum R, Rittner HL. Human dorsal root ganglia are either preserved or completely lost after deafferentation by brachial plexus injury. Br J Anaesth 2024; 133:1250-1262. [PMID: 39393999 PMCID: PMC11589459 DOI: 10.1016/j.bja.2024.09.004] [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: 05/29/2024] [Revised: 08/05/2024] [Accepted: 09/13/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Plexus injury results in lifelong suffering from flaccid paralysis, sensory loss, and intractable pain. For this clinical problem, regenerative medicine concepts set high expectations. However, it is largely unknown how dorsal root ganglia (DRG) are affected by accidental deafferentation. METHODS Here, we phenotyped DRG of a clinically and MRI-characterised cohort of 13 patients with plexus injury. Avulsed DRG were collected during reconstructive nerve surgery. For control, we used DRG from forensic autopsy. The cellular composition of the DRG was analysed in histopathological slices with multicolour high-resolution immunohistochemistry, tile microscopy, and deep-learning-based bioimage analysis. We then sequenced the bulk RNA of corresponding DRG slices. RESULTS In about half of the patients we found loss of the typical DRG units consisting of neurones and satellite glial cells. The DRG cells were replaced by mesodermal/connective tissue. In the remaining patients, the cellular units were well preserved. Preoperative plexus MRI neurography was not able to distinguish the two types. Patients with 'neuronal preservation' had less maximum pain than patients with 'neuronal loss'. Arm function improved after nerve reconstruction, but severe pain persisted. Transcriptome analysis of preserved DRGs revealed expression of subtype-specific sensory neurone marker genes, but downregulation of neuronal attributes. Furthermore, they showed signs of ongoing inflammation and connective tissue remodelling. CONCLUSIONS Patients with plexus injury separate into two groups with either neuronal preservation or neuronal loss. The former could benefit from anti-inflammatory therapy. For the latter, studies should explore mechanisms of neuronal loss especially for regenerative approaches. CLINICAL TRIAL REGISTRATION DRKS00017266.
Collapse
Affiliation(s)
- Annemarie Sodmann
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany; Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Johannes Degenbeck
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Annemarie Aue
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Magnus Schindehütte
- Institute of Neuroradiology, University Hospital of Würzburg, Würzburg, Germany
| | - Felicitas Schlott
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany; Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Panagiota Arampatzi
- Core Unit Systems Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Thorsten Bischler
- Core Unit Systems Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Max Schneider
- Department of Neurosurgery, Peripheral Nerve Surgery Unit, University of Ulm, Günzburg, Germany
| | - Alexander Brack
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Camelia M Monoranu
- Institute of Pathology, Section Neuropathology, University of Würzburg, Würzburg, Germany
| | - Tom Gräfenhan
- Core Unit Systems Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Michael Bohnert
- Institute of Forensic Medicine, University of Würzburg, Würzburg, Germany
| | - Mirko Pham
- Institute of Neuroradiology, University Hospital of Würzburg, Würzburg, Germany
| | - Gregor Antoniadis
- Department of Neurosurgery, Peripheral Nerve Surgery Unit, University of Ulm, Günzburg, Germany
| | - Robert Blum
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany.
| | - Heike L Rittner
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, Centre for Interdisciplinary Pain Medicine, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
13
|
Patel P, Thakkar K, Shah D, Shah U, Pandey N, Patel J, Patel A. Decrypting the multifaceted peripheral neuropathy based on molecular pathology and therapeutics: a comprehensive review. Arch Physiol Biochem 2024; 130:886-897. [PMID: 38588401 DOI: 10.1080/13813455.2024.2336916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/28/2023] [Accepted: 03/26/2024] [Indexed: 04/10/2024]
Abstract
CONTEXT Peripheral neuropathy (PN) is a multifaceted complication characterized by nerve damage due to oxidative stress, inflammatory mediators, and dysregulated metabolic processes. Early PN manifests as sensory changes that develop progressively in a "stocking and glove" pattern. METHODS AND MECHANISMS A thorough review of literature has been done to find the molecular pathology, clinical trials that have been conducted to screen the effects of different drugs, current treatments and novel approaches used in PN therapy. Diabetic neuropathy occurs due to altered protein kinase C activity, elevated polyol pathway activity in neurons, and Schwann cells-induced hyperglycemia. Other causes involve chemotherapy exposure, autoimmune ailments, and chronic ethanol intake. CONCLUSION Symptomatic treatments for neuropathic pain include use of tricyclic antidepressants, anticonvulsants, and acetyl-L-carnitine. Patients will have new hope if clinicians focus on novel therapies including gene therapy, neuromodulation techniques, and cannabidiol as an alternative to traditional medications, as management is still not ideal.
Collapse
Affiliation(s)
- Praysha Patel
- Ramanbhai Patel College of Pharmacy, CHARUSAT, Changa, Gujarat, India
| | - Krishna Thakkar
- Ramanbhai Patel College of Pharmacy, CHARUSAT, Changa, Gujarat, India
| | - Div Shah
- Ramanbhai Patel College of Pharmacy, CHARUSAT, Changa, Gujarat, India
| | - Umang Shah
- Ramanbhai Patel College of Pharmacy, CHARUSAT, Changa, Gujarat, India
| | - Nilesh Pandey
- Health Science Center, Louisiana State University, Shreveport, LA, USA
| | - Jayesh Patel
- Consultant, Vascular surgeon, Shree Krishna Hospital, Karamsad, Gujarat, India
| | - Alkeshkumar Patel
- Ramanbhai Patel College of Pharmacy, CHARUSAT, Changa, Gujarat, India
| |
Collapse
|
14
|
Cooper AH, Barry AM, Chrysostomidou P, Lolignier R, Wang J, Redondo Canales M, Titterton HF, Bennett DL, Weir GA. Peripheral nerve injury results in a biased loss of sensory neuron subpopulations. Pain 2024; 165:2863-2876. [PMID: 39158319 PMCID: PMC11562755 DOI: 10.1097/j.pain.0000000000003321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/11/2024] [Accepted: 05/25/2024] [Indexed: 08/20/2024]
Abstract
ABSTRACT There is a rich literature describing the loss of dorsal root ganglion (DRG) neurons following peripheral axotomy, but the vulnerability of discrete subpopulations has not yet been characterised. Furthermore, the extent or even presence of neuron loss following injury has recently been challenged. In this study, we have used a range of transgenic recombinase driver mouse lines to genetically label molecularly defined subpopulations of DRG neurons and track their survival following traumatic nerve injury. We find that spared nerve injury leads to a marked loss of cells containing DRG volume and a concomitant loss of small-diameter DRG neurons. Neuron loss occurs unequally across subpopulations and is particularly prevalent in nonpeptidergic nociceptors, marked by expression of Mrgprd. We show that this subpopulation is almost entirely lost following spared nerve injury and severely depleted (by roughly 50%) following sciatic nerve crush. Finally, we used an in vitro model of DRG neuron survival to demonstrate that nonpeptidergic nociceptor loss is likely dependent on the absence of neurotrophic support. Together, these results profile the extent to which DRG neuron subpopulations can survive axotomy, with implications for our understanding of nerve injury-induced plasticity and pain.
Collapse
Affiliation(s)
- Andrew H. Cooper
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - Allison M. Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Romane Lolignier
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - Jinyi Wang
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | | | - Heather F. Titterton
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Greg A. Weir
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
15
|
Hincker A, Reschke M, Ginosar Y, Kagan L, Kharasch ED, Siemiątkowska A, Park C, Bakos K, Ben-Abdallah A, Haroutounian S. Epidural methadone and morphine pharmacokinetics and clinical effects in healthy volunteers: A randomized, crossover-design trial. Br J Clin Pharmacol 2024; 90:2883-2896. [PMID: 39049497 PMCID: PMC11906210 DOI: 10.1111/bcp.16178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
AIMS Epidural opioids can provide effective analgesia for acute postoperative pain. Due to its unique physicochemical properties and long systemic elimination half-life, epidural methadone may provide lasting analgesia with minimal adverse effects; however, human studies are lacking. The aim of the study was to test the hypothesis that epidural methadone would exhibit greater segmental analgesia (analgesia at the dermatome of injection vs. distant dermatomes) than epidural morphine. METHODS In a prospective, randomized, double-blinded, crossover study, thirteen healthy volunteers received a 4-mg epidural bolus of methadone or morphine at L3-L4 and underwent repeated assessment of dermatomal heat pain tolerance and pressure pain threshold at lumbar (L3) and trigeminal (V2) dermatomes, pupil diameter, respiratory parameters and venous opioid concentration for 24 h. The primary outcome was selective (lumbar vs. trigeminal) segmental analgesia for heat pain, as a marker of a spinal analgesic mechanism. RESULTS The degree of segmental analgesia to heat pain tolerance was not different between morphine and methadone (P = .09), although morphine (P = .0009) but not methadone (P = .81) produced significant analgesia to heat pain at the lumbar vs. trigeminal dermatome over 0-12 h. Morphine overall provided longer lasting analgesia to heat pain vs. methadone (24 vs. 2 h, respectively). Morphine elicited greater systemic effects, including miosis (P = .009) and opioid-related adverse effects (P = .002). CONCLUSIONS These results suggest that, with equal epidural doses, both methadone and morphine produced analgesia and methadone did not produce greater segmental effects than morphine. Epidural methadone provided a more favourable adverse effect profile.
Collapse
Affiliation(s)
- Alexander Hincker
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO
- Division of Obstetric Anesthesiology, Mercy Hospital Saint Louis, Saint Louis, MO
| | - Matthew Reschke
- Department of Anesthesiology, Banner University Medical Center, Phoenix, AZ
| | - Yehuda Ginosar
- Department of Anesthesiology, Critical Care and Pain Medicine, Hadassah Ein Karem Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Leonid Kagan
- Department of Pharmaceutics and Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ
| | - Evan D. Kharasch
- Department of Anesthesiology, Duke University School of Medicine; Bermaride LLC, Durham, NC
| | - Anna Siemiątkowska
- Department of Pharmaceutics and Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Collegium Pharmaceuticum, 3 Rokietnicka Street, Poznan 60-806, Poland
| | - Celine Park
- Department of Pharmaceutics and Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ
| | - Kristopher Bakos
- Investigational Drug Service, Department of Pharmacy, Barnes-Jewish Hospital, Saint Louis, MO
| | - Arbi Ben-Abdallah
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO
- Washington University Pain Center, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
16
|
Yang T, Liu X, Cao R, Zhou X, Li W, Wu W, Yu W, Zhang X, Guo Z, Cui S. Establishment of a Magnetically Controlled Scalable Nerve Injury Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405265. [PMID: 39287118 PMCID: PMC11538664 DOI: 10.1002/advs.202405265] [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] [Received: 05/14/2024] [Revised: 08/21/2024] [Indexed: 09/19/2024]
Abstract
Animal models of peripheral nerve injury (PNI) serve as the fundamental basis for the investigations of nerve injury, regeneration, and neuropathic pain. The injury properties of such models, including the intensity and duration, significantly influence the subsequent pathological changes, pain development, and therapeutic efficacy. However, precise control over the intensity and duration of nerve injury remains challenging within existing animal models, thereby impeding accurate and comparative assessments of relevant cases. Here, a new model that provides quantitative and off-body controllable injury properties via a magnetically controlled clamp, is presented. The clamp can be implanted onto the rat sciatic nerve and exert varying degrees of compression under the control of an external magnetic field. It is demonstrated that this model can accurately simulate various degrees of pathology of human patients by adjusting the magnetic control and reveal specific pathological changes resulting from intensity heterogeneity that are challenging to detect previously. The controllability and quantifiability of this model may significantly reduce the uncertainty of central response and inter-experimenter variability, facilitating precise investigations into nerve injury, regeneration, and pain mechanisms.
Collapse
Affiliation(s)
- Tuo Yang
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityNo.126, Xiantai StreetChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceNo.126, Xiantai StreetChangchun130033China
| | - Xilin Liu
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityNo.126, Xiantai StreetChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceNo.126, Xiantai StreetChangchun130033China
| | - Rangjuan Cao
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityNo.126, Xiantai StreetChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceNo.126, Xiantai StreetChangchun130033China
| | - Xiongyao Zhou
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityNo.126, Xiantai StreetChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceNo.126, Xiantai StreetChangchun130033China
| | - Weizhen Li
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityNo.126, Xiantai StreetChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceNo.126, Xiantai StreetChangchun130033China
| | - Wenzheng Wu
- School of Mechanical and Aerospace Engineering of Jilin University5988 Renmin StreetChangchun130025China
| | - Wei Yu
- Department of Wound Repair, Plastic and Reconstructive MicrosurgeryChina‐Japan Union Hospital of Jilin UniversityNo.126, Xiantai StreetChangchun130033China
| | - Xianyu Zhang
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityNo.126, Xiantai StreetChangchun130033China
| | - Zhengxiao Guo
- Department of ChemistryThe University of Hong KongHong Kong999077China
| | - Shusen Cui
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityNo.126, Xiantai StreetChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceNo.126, Xiantai StreetChangchun130033China
| |
Collapse
|
17
|
Sun Z, Han W, Dou Z, Lu N, Wang X, Wang F, Ma S, Tian Z, Xian H, Liu W, Liu Y, Wu W, Chu W, Guo H, Wang F, Ding H, Liu Y, Tao H, Freichel M, Birnbaumer L, Li Z, Xie R, Wu S, Luo C. TRPC3/6 Channels Mediate Mechanical Pain Hypersensitivity via Enhancement of Nociceptor Excitability and of Spinal Synaptic Transmission. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404342. [PMID: 39340833 PMCID: PMC11600220 DOI: 10.1002/advs.202404342] [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] [Received: 04/23/2024] [Revised: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Patients with tissue inflammation or injury often experience aberrant mechanical pain hypersensitivity, one of leading symptoms in clinic. Despite this, the molecular mechanisms underlying mechanical distortion are poorly understood. Canonical transient receptor potential (TRPC) channels confer sensitivity to mechanical stimulation. TRPC3 and TRPC6 proteins, coassembling as heterotetrameric channels, are highly expressed in sensory neurons. However, how these channels mediate mechanical pain hypersensitivity has remained elusive. It is shown that in mice and human, TRPC3 and TRPC6 are upregulated in DRG and spinal dorsal horn under pathological states. Double knockout of TRPC3/6 blunts mechanical pain hypersensitivity, largely by decreasing nociceptor hyperexcitability and spinal synaptic potentiation via presynaptic mechanism. In corroboration with this, nociceptor-specific ablation of TRPC3/6 produces comparable pain relief. Mechanistic analysis reveals that upon peripheral inflammation, TRPC3/6 in primary sensory neurons get recruited via released bradykinin acting on B1/B2 receptors, facilitating BDNF secretion from spinal nociceptor terminals, which in turn potentiates synaptic transmission through TRPC3/6 and eventually results in mechanical pain hypersensitivity. Antagonizing TRPC3/6 in DRG relieves mechanical pain hypersensitivity in mice and nociceptor hyperexcitability in human. Thus, TRPC3/6 in nociceptors is crucially involved in pain plasticity and constitutes a promising therapeutic target against mechanical pain hypersensitivity with minor side effects.
Collapse
Affiliation(s)
- Zhi‐Chuan Sun
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
- Department of NeurosurgeryXi'an Daxing HospitalXi'an710016China
| | - Wen‐Juan Han
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Zhi‐Wei Dou
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Na Lu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
- The Assisted Reproduction CenterNorthwest Women and Children's HospitalXi'an710000China
| | - Xu Wang
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Fu‐Dong Wang
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Sui‐Bin Ma
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Zhi‐Cheng Tian
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Hang Xian
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
- Department of OrthopedicsXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Wan‐Neng Liu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Ying‐Ying Liu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Wen‐Bin Wu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Wen‐Guang Chu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Huan Guo
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Fei Wang
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Hui Ding
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Yuan‐Ying Liu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Hui‐Ren Tao
- Department of Orthopedic SurgeryThe University of Hong Kong‐Shenzhen HospitalShenzhenGuangdong518053China
| | - Marc Freichel
- Institute of PharmacologyHeidelberg University69120HeidelbergGermany
| | - Lutz Birnbaumer
- Institute of Biomedical Research (BIOMED)Catholic University of ArgentinaBuenos AiresC1107AVVArgentina
- Signal Transduction LaboratoryNational institute of Environmental Health SciencesResearch Triangle ParkNC27709United States
| | - Zhen‐Zhen Li
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Rou‐Gang Xie
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Sheng‐Xi Wu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Ceng Luo
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
- Innovation Research InstituteXijing HospitalFourth Military Medical UniversityXi'an710032China
| |
Collapse
|
18
|
Rutter-Locher Z, Norton S, Denk F, McMahon S, Taams LS, Kirkham BW, Bannister K. A randomised controlled trial of the effect of intra-articular lidocaine on pain scores in inflammatory arthritis. Pain 2024; 165:2578-2585. [PMID: 38888846 PMCID: PMC11474916 DOI: 10.1097/j.pain.0000000000003291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/02/2024] [Accepted: 04/06/2024] [Indexed: 06/20/2024]
Abstract
ABSTRACT Chronic pain in inflammatory arthritis (IA) reflects a complex interplay between active disease in a peripheral joint and central pronociceptive mechanisms. Because intra-articular lidocaine may be used to abolish joint-specific peripheral input to the central nervous system, we aimed to validate its use as a clinical tool to identify those patients with IA whose pain likely incorporates centrally mediated mechanisms. We began by investigating whether there was a placebo response of intra-articular injection in patients with IA 1:1 randomised to receive intra-articular lidocaine or control (0.9% saline). After, in a larger patient cohort not randomized to placebo vs lidocaine groups, we tested whether patients with IA could be stratified into 2 cohorts based on their response to intra-articular lidocaine according to markers of centrally mediated pain. To this end, we evaluated postlidocaine pain numerical rating scale (NRS) scores alongside baseline painDETECT, fibromyalgia criteria fulfillment, and quantitative sensory testing outcomes. Numerical rating scale scores were collected at baseline and 3-, 5-, and 10-minutes postinjection. Firstly, the placebo effect of intra-articular injection was low: compared to baseline, the mean pain NRS score 5-minutes postinjection was reduced by 3.5 points in the lidocaine group vs 1.2 points in the control group. Secondly, postlidocaine NRS scores were significantly higher in those with a high (>18) baseline painDETECT score, fibromyalgia, and low-pressure pain threshold at the trapezius ( P = 0.002, P = 0.001, P = 0.005, respectively). Persistent high pain after intra-articular lidocaine injection could be used as an indicator of pronociceptive mechanisms that are centrally mediated, informing centrally targeted analgesic strategies.
Collapse
Affiliation(s)
- Zoe Rutter-Locher
- Rheumatology Department, Guy's and St Thomas' NHS Trust, London, United Kingdom
- Department Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College University, London, United Kingdom
| | - Sam Norton
- Centre for Rheumatic Diseases, King's College London, London, United Kingdom
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, Guy's Campus, King's College London, London, United Kingdom
| | - Stephen McMahon
- Wolfson Centre for Age-Related Diseases, Guy's Campus, King's College London, London, United Kingdom
| | - Leonie S. Taams
- Department Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College University, London, United Kingdom
| | - Bruce W. Kirkham
- Rheumatology Department, Guy's and St Thomas' NHS Trust, London, United Kingdom
| | - Kirsty Bannister
- Wolfson Centre for Age-Related Diseases, Guy's Campus, King's College London, London, United Kingdom
| |
Collapse
|
19
|
Li Y, Uhelski ML, North RY, Mwirigi JM, Tatsui CE, McDonough KE, Cata JP, Corrales G, Dussor G, Price TJ, Dougherty PM. Tomivosertib reduces ectopic activity in dorsal root ganglion neurons from patients with radiculopathy. Brain 2024; 147:2991-2997. [PMID: 39046204 PMCID: PMC11370786 DOI: 10.1093/brain/awae178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 07/25/2024] Open
Abstract
Spontaneous activity in dorsal root ganglion (DRG) neurons is a key driver of neuropathic pain in patients suffering from this largely untreated disease. While many intracellular signalling mechanisms have been examined in preclinical models that drive spontaneous activity, none have been tested directly on spontaneously active human nociceptors. Using cultured DRG neurons recovered during thoracic vertebrectomy surgeries, we showed that inhibition of mitogen-activated protein kinase interacting kinase (MNK) with tomivosertib (eFT508, 25 nM) reversibly suppresses spontaneous activity in human sensory neurons that are likely nociceptors based on size and action potential characteristics associated with painful dermatomes within minutes of treatment. Tomivosertib treatment also decreased action potential amplitude and produced alterations in the magnitude of after hyperpolarizing currents, suggesting modification of Na+ and K+ channel activity as a consequence of drug treatment. Parallel to the effects on electrophysiology, eFT508 treatment led to a profound loss of eIF4E serine 209 phosphorylation in primary sensory neurons, a specific substrate of MNK, within 2 min of drug treatment. Our results create a compelling case for the future testing of MNK inhibitors in clinical trials for neuropathic pain.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Megan L Uhelski
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Y North
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juliet M Mwirigi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Claudio E Tatsui
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kathleen E McDonough
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juan P Cata
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - German Corrales
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Greg Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Patrick M Dougherty
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
20
|
Sankaranarayanan I, Kume M, Mohammed A, Mwirigi JM, Inturi NN, Munro G, Petersen KA, Tavares-Ferreira D, Price TJ. Persistent changes in nociceptor translatomes govern hyperalgesic priming in mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.606891. [PMID: 39149295 PMCID: PMC11326310 DOI: 10.1101/2024.08.07.606891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Hyperalgesic priming is a model system that has been widely used to understand plasticity in painful stimulus-detecting sensory neurons, called nociceptors. A key feature of this model system is that following priming, stimuli that do not normally cause hyperalgesia now readily provoke this state. We hypothesized that hyperalgesic priming occurs due to reorganization of translation of mRNA in nociceptors. To test this hypothesis, we used paclitaxel treatment as the priming stimulus and translating ribosome affinity purification (TRAP) to measure persistent changes in mRNA translation in Nav1.8+ nociceptors. TRAP sequencing revealed 161 genes with persistently altered mRNA translation in the primed state. We identified Gpr88 as upregulated and Metrn as downregulated. We confirmed a functional role for these genes, wherein a GPR88 agonist causes pain only in primed mice and established hyperalgesic priming is reversed by Meteorin. Our work demonstrates that altered nociceptor translatomes are causative in producing hyperalgesic priming.
Collapse
Affiliation(s)
- Ishwarya Sankaranarayanan
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Moeno Kume
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Ayaan Mohammed
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Juliet M Mwirigi
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Nikhil Nageswar Inturi
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | | | | | - Diana Tavares-Ferreira
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Theodore J Price
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| |
Collapse
|
21
|
Koui Y, Song S, Dong X, Mukouyama YS. Local keratinocyte-nociceptor interactions enhance obesity-mediated small fiber neuropathy via NGF-TrkA-PI3K signaling axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603316. [PMID: 39372742 PMCID: PMC11452191 DOI: 10.1101/2024.07.12.603316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The pathology of diabetic small fiber neuropathy, characterized by neuropathic pain and axon degeneration, develops locally within the skin during the stages of obesity and pre-diabetes. However, the initiation and progression of morphological and functional abnormalities in skin sensory nerves remains elusive. To address this, we utilized ear skin from mice with diet-induced obesity (DIO), the mouse models for obesity and pre-type 2 diabetes. We evaluated pain-associated wiping behavior and conducted ex vivo live Ca2+ imaging of the DIO ear skin to detect sensory hypersensitivity. Our findings reveal sensory hypersensitivity in skin nociceptive axons followed by axon degeneration. Further mechanistic analysis identified keratinocytes as a major source of nerve growth factor (NGF) in DIO skin, which locally sensitizes nociceptors through NGF-mediated signaling. Indeed, the local inactivation of NGF and its receptor TrkA-mediated downstream signaling, including the phosphoinositide 3-kinases (PI3K) pathway, suppresses sensory hypersensitivity in DIO skin. Thus, targeting these local interactions between keratinocytes and nociceptors offers a therapeutic strategy for managing neuropathic pain, avoiding the adverse effects associated with systemic interventions.
Collapse
Affiliation(s)
- Yuta Koui
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health; Bethesda, Maryland 20892, USA
| | - Shuxuan Song
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health; Bethesda, Maryland 20892, USA
- Biological Sciences Graduate Program, University of Maryland, College Park, Maryland 20742, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Yoh-suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health; Bethesda, Maryland 20892, USA
| |
Collapse
|
22
|
de Almeida DL, Mendes Ferreira RC, Fonseca FC, Dias Machado DP, Aguiar DD, Guimaraes FS, Duarte IDG, Romero TRL. Cannabidiol induces systemic analgesia through activation of the PI3Kγ/nNOS/NO/KATP signaling pathway in neuropathic mice. A KATP channel S-nitrosylation-dependent mechanism. Nitric Oxide 2024; 146:1-9. [PMID: 38428514 DOI: 10.1016/j.niox.2024.02.005] [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: 11/06/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Cannabidiol (CBD) is the second most abundant pharmacologically active component present in Cannabis sp. Unlike Δ-9-tetrahydrocannabinol (THC), it has no psychotomimetic effects and has recently received significant interest from the scientific community due to its potential to treat anxiety and epilepsy. CBD has excellent anti-inflammatory potential and can be used to treat some types of inflammatory and neuropathic pain. In this context, the present study aimed to evaluate the analgesic mechanism of cannabidiol administered systemically for the treatment of neuropathic pain and determine the endogenous mechanisms involved with this analgesia. METHODS Neuropathic pain was induced by sciatic nerve constriction surgery, and the nociceptive threshold was measured using the paw compression test in mice. RESULTS CBD produced dose-dependent antinociception after intraperitoneal injection. Selective inhibition of PI3Kγ dose-dependently reversed CBD-induced antinociception. Selective inhibition of nNOS enzymes reversed the antinociception induced by CBD, while selective inhibition of iNOS and eNOS did not alter this antinociception. However, the inhibition of cGMP production by guanylyl cyclase did not alter CBD-mediated antinociception, but selective blockade of ATP-sensitive K+ channels dose-dependently reversed CBD-induced antinociception. Inhibition of S-nitrosylation dose-dependently and completely reversed CBD-mediated antinociception. CONCLUSION Cannabidiol has an antinociceptive effect when administered systemically and this effect is mediated by the activation of PI3Kγ as well as by nitric oxide and subsequent direct S-nitrosylation of KATP channels on peripheral nociceptors.
Collapse
Affiliation(s)
| | | | | | | | | | - Francisco Silveira Guimaraes
- Department of Pharmacology, FMRP, Campus USP, University of São Paulo, Av. Bandeirantes 13400, Ribeirão Preto, SP, 14049-900, Brazil
| | | | | |
Collapse
|
23
|
Cornish P, Humphrey N, Cornish A, Emmerson RB. An in silico analysis of neuromodulation for pain relief: Determining the role of classical electrodynamics. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2024; 40:e3813. [PMID: 38508998 DOI: 10.1002/cnm.3813] [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: 12/07/2022] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 03/22/2024]
Abstract
There has been ongoing debate about the efficacy and mechanism of action of neuromodulation devices in pain relief applications. It has recently been suggested that both issues may be resolved if electromagnetic theory is incorporated into the understanding and application of this technology, and we therefore undertook an in silico analysis to further explore this idea. We created a CAD replication of a standard neuromodulation electrode array with a generic linear 3/6 mm 8-contact lead, developed a parameterized algorithmic model for the pulse delivered by the device and assigned material properties to biologic media to accurately reflect their electromagnetic properties. We then created a physical simulation of the device's output both in air and in the biophysical environment. The simulations confirmed the presence of an electromagnetic field (EM field). Variations in programming of the device affected the strength of the EM field by orders of magnitude. The biologic media all absorbed the EM field, an effect which was particularly pronounced in cerebrospinal fluid and muscle. We discuss the implications of all these findings in relation to the literature. We suggest that knowledge of electromagnetic theory and its application within the biophysical space is required for the optimal use of neuromodulation devices in pain relief applications.
Collapse
Affiliation(s)
- Philip Cornish
- Specialised Pain Medicine Pty Ltd, Adelaide, South Australia, Australia
| | - Nabil Humphrey
- Special Research Centre for the Subatomic Structure of Matter, University of Adelaide, Adelaide, South Australia, Australia
| | - Anne Cornish
- Specialised Pain Medicine Pty Ltd, Adelaide, South Australia, Australia
| | | |
Collapse
|
24
|
Xie YF, Yang J, Ratté S, Prescott SA. Similar excitability through different sodium channels and implications for the analgesic efficacy of selective drugs. eLife 2024; 12:RP90960. [PMID: 38687187 PMCID: PMC11060714 DOI: 10.7554/elife.90960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
Nociceptive sensory neurons convey pain-related signals to the CNS using action potentials. Loss-of-function mutations in the voltage-gated sodium channel NaV1.7 cause insensitivity to pain (presumably by reducing nociceptor excitability) but clinical trials seeking to treat pain by inhibiting NaV1.7 pharmacologically have struggled. This may reflect the variable contribution of NaV1.7 to nociceptor excitability. Contrary to claims that NaV1.7 is necessary for nociceptors to initiate action potentials, we show that nociceptors can achieve similar excitability using different combinations of NaV1.3, NaV1.7, and NaV1.8. Selectively blocking one of those NaV subtypes reduces nociceptor excitability only if the other subtypes are weakly expressed. For example, excitability relies on NaV1.8 in acutely dissociated nociceptors but responsibility shifts to NaV1.7 and NaV1.3 by the fourth day in culture. A similar shift in NaV dependence occurs in vivo after inflammation, impacting ability of the NaV1.7-selective inhibitor PF-05089771 to reduce pain in behavioral tests. Flexible use of different NaV subtypes exemplifies degeneracy - achieving similar function using different components - and compromises reliable modulation of nociceptor excitability by subtype-selective inhibitors. Identifying the dominant NaV subtype to predict drug efficacy is not trivial. Degeneracy at the cellular level must be considered when choosing drug targets at the molecular level.
Collapse
Affiliation(s)
- Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
| | - Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
- Institute of Biomedical Engineering, University of TorontoTorontoCanada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
| | - Steven A Prescott
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
- Institute of Biomedical Engineering, University of TorontoTorontoCanada
- Department of Physiology, University of TorontoTorontoCanada
| |
Collapse
|
25
|
Alexandre C, Miracca G, Holanda VD, Sharma A, Kourbanova K, Ferreira A, Bicca MA, Zeng X, Nassar VA, Lee S, Kaur S, Sarma SV, Sacré P, Scammell TE, Woolf CJ, Latremoliere A. Nociceptor spontaneous activity is responsible for fragmenting non-rapid eye movement sleep in mouse models of neuropathic pain. Sci Transl Med 2024; 16:eadg3036. [PMID: 38630850 PMCID: PMC11106840 DOI: 10.1126/scitranslmed.adg3036] [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: 12/14/2022] [Accepted: 03/27/2024] [Indexed: 04/19/2024]
Abstract
Spontaneous pain, a major complaint of patients with neuropathic pain, has eluded study because there is no reliable marker in either preclinical models or clinical studies. Here, we performed a comprehensive electroencephalogram/electromyogram analysis of sleep in several mouse models of chronic pain: neuropathic (spared nerve injury and chronic constriction injury), inflammatory (Freund's complete adjuvant and carrageenan, plantar incision) and chemical pain (capsaicin). We find that peripheral axonal injury drives fragmentation of sleep by increasing brief arousals from non-rapid eye movement sleep (NREMS) without changing total sleep amount. In contrast to neuropathic pain, inflammatory or chemical pain did not increase brief arousals. NREMS fragmentation was reduced by the analgesics gabapentin and carbamazepine, and it resolved when pain sensitivity returned to normal in a transient neuropathic pain model (sciatic nerve crush). Genetic silencing of peripheral sensory neurons or ablation of CGRP+ neurons in the parabrachial nucleus prevented sleep fragmentation, whereas pharmacological blockade of skin sensory fibers was ineffective, indicating that the neural activity driving the arousals originates ectopically in primary nociceptor neurons and is relayed through the lateral parabrachial nucleus. These findings identify NREMS fragmentation by brief arousals as an effective proxy to measure spontaneous neuropathic pain in mice.
Collapse
Affiliation(s)
- Chloe Alexandre
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Giulia Miracca
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Victor Duarte Holanda
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ashley Sharma
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kamila Kourbanova
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ashley Ferreira
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Maíra A. Bicca
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Xiangsunze Zeng
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Victoria A. Nassar
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Seungkyu Lee
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Satvinder Kaur
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Sridevi V. Sarma
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Pierre Sacré
- Department of Electrical Engineering and Computer Science, School of Engineering, University of Liège, Liège, Belgium
| | - Thomas E. Scammell
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Clifford J. Woolf
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Latremoliere
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
26
|
Smith PA. BDNF in Neuropathic Pain; the Culprit that Cannot be Apprehended. Neuroscience 2024; 543:49-64. [PMID: 38417539 DOI: 10.1016/j.neuroscience.2024.02.020] [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: 12/16/2023] [Accepted: 02/20/2024] [Indexed: 03/01/2024]
Abstract
In males but not in females, brain derived neurotrophic factor (BDNF) plays an obligatory role in the onset and maintenance of neuropathic pain. Afferent terminals of injured peripheral nerves release colony stimulating factor (CSF-1) and other mediators into the dorsal horn. These transform the phenotype of dorsal horn microglia such that they express P2X4 purinoceptors. Activation of these receptors by neuron-derived ATP promotes BDNF release. This microglial-derived BDNF increases synaptic activation of excitatory dorsal horn neurons and decreases that of inhibitory neurons. It also alters the neuronal chloride gradient such the normal inhibitory effect of GABA is converted to excitation. By as yet undefined processes, this attenuated inhibition increases NMDA receptor function. BDNF also promotes the release of pro-inflammatory cytokines from astrocytes. All of these actions culminate in the increase dorsal horn excitability that underlies many forms of neuropathic pain. Peripheral nerve injury also alters excitability of structures in the thalamus, cortex and mesolimbic system that are responsible for pain perception and for the generation of co-morbidities such as anxiety and depression. The weight of evidence from male rodents suggests that this preferential modulation of excitably of supra-spinal pain processing structures also involves the action of microglial-derived BDNF. Possible mechanisms promoting the preferential release of BDNF in pain signaling structures are discussed. In females, invading T-lymphocytes increase dorsal horn excitability but it remains to be determined whether similar processes operate in supra-spinal structures. Despite its ubiquitous role in pain aetiology neither BDNF nor TrkB receptors represent potential therapeutic targets.
Collapse
Affiliation(s)
- Peter A Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
27
|
Demarest P, Rustamov N, Swift J, Xie T, Adamek M, Cho H, Wilson E, Han Z, Belsten A, Luczak N, Brunner P, Haroutounian S, Leuthardt EC. A novel theta-controlled vibrotactile brain-computer interface to treat chronic pain: a pilot study. Sci Rep 2024; 14:3433. [PMID: 38341457 PMCID: PMC10858946 DOI: 10.1038/s41598-024-53261-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Limitations in chronic pain therapies necessitate novel interventions that are effective, accessible, and safe. Brain-computer interfaces (BCIs) provide a promising modality for targeting neuropathology underlying chronic pain by converting recorded neural activity into perceivable outputs. Recent evidence suggests that increased frontal theta power (4-7 Hz) reflects pain relief from chronic and acute pain. Further studies have suggested that vibrotactile stimulation decreases pain intensity in experimental and clinical models. This longitudinal, non-randomized, open-label pilot study's objective was to reinforce frontal theta activity in six patients with chronic upper extremity pain using a novel vibrotactile neurofeedback BCI system. Patients increased their BCI performance, reflecting thought-driven control of neurofeedback, and showed a significant decrease in pain severity (1.29 ± 0.25 MAD, p = 0.03, q = 0.05) and pain interference (1.79 ± 1.10 MAD p = 0.03, q = 0.05) scores without any adverse events. Pain relief significantly correlated with frontal theta modulation. These findings highlight the potential of BCI-mediated cortico-sensory coupling of frontal theta with vibrotactile stimulation for alleviating chronic pain.
Collapse
Affiliation(s)
- Phillip Demarest
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St Louis, MO, 63130, USA
| | - Nabi Rustamov
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - James Swift
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Tao Xie
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Markus Adamek
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Hohyun Cho
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Elizabeth Wilson
- Division of Clinical and Translational Research, Department of Anesthesiology, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Washington University Pain Center, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Zhuangyu Han
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St Louis, MO, 63130, USA
| | - Alexander Belsten
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Nicholas Luczak
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Peter Brunner
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St Louis, MO, 63130, USA
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Simon Haroutounian
- Division of Clinical and Translational Research, Department of Anesthesiology, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
- Washington University Pain Center, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA
| | - Eric C Leuthardt
- Division of Neurotechnology, Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA.
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St Louis, MO, 63130, USA.
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St Louis, MO, 63110, USA.
| |
Collapse
|
28
|
Bavencoffe AG, Lopez ER, Johnson KN, Tian J, Gorgun FM, Shen BQ, Zhu MX, Dessauer CW, Walters ET. Widespread latent hyperactivity of nociceptors outlasts enhanced avoidance behavior following incision injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.578108. [PMID: 38352319 PMCID: PMC10862851 DOI: 10.1101/2024.01.30.578108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Nociceptors with somata in dorsal root ganglia (DRGs) exhibit an unusual readiness to switch from an electrically silent state to a hyperactive state of tonic, nonaccommodating, low-frequency, irregular discharge of action potentials (APs). Ongoing activity (OA) during this state is present in vivo in rats months after spinal cord injury (SCI), and has been causally linked to SCI pain. OA induced by various neuropathic conditions in rats, mice, and humans is retained in nociceptor somata after dissociation and culturing, providing a powerful tool for investigating its mechanisms and functions. An important question is whether similar nociceptor OA is induced by painful conditions other than neuropathy. The present study shows that probable nociceptors dissociated from DRGs of rats subjected to postsurgical pain (induced by plantar incision) exhibit OA. The OA was most apparent when the soma was artificially depolarized to a level within the normal range of membrane potentials where large, transient depolarizing spontaneous fluctuations (DSFs) can approach AP threshold. This latent hyperactivity persisted for at least 3 weeks, whereas behavioral indicators of affective pain - hindpaw guarding and increased avoidance of a noxious substrate in an operant conflict test - persisted for 1 week or less. An unexpected discovery was latent OA in neurons from thoracic DRGs that innervate dermatomes distant from the injured tissue. The most consistent electrophysiological alteration associated with OA was enhancement of DSFs. Potential in vivo functions of widespread, low-frequency nociceptor OA consistent with these and other findings are to amplify hyperalgesic priming and to drive anxiety-related hypervigilance.
Collapse
Affiliation(s)
- Alexis G. Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Elia R. Lopez
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Kayla N. Johnson
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Falih M. Gorgun
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Breanna Q. Shen
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| |
Collapse
|
29
|
Smith PA. The Known Biology of Neuropathic Pain and Its Relevance to Pain Management. Can J Neurol Sci 2024; 51:32-39. [PMID: 36799022 DOI: 10.1017/cjn.2023.10] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Patients with neuropathic pain are heterogeneous in pathophysiology, etiology, and clinical presentation. Signs and symptoms are determined by the nature of the injury and factors such as genetics, sex, prior injury, age, culture, and environment. Basic science has provided general information about pain etiology by studying the consequences of peripheral injury in rodent models. This is associated with the release of inflammatory cytokines, chemokines, and growth factors that sensitize sensory nerve endings, alter gene expression, promote post-translational modification of proteins, and alter ion channel function. This leads to spontaneous activity in primary afferent neurons that is crucial for the onset and persistence of pain and the release of secondary mediators such as colony-stimulating factor 1 from primary afferent terminals. These promote the release of tertiary mediators such as brain-derived neurotrophic factor and interleukin-1β from microglia and astrocytes. Tertiary mediators facilitate the transmission of nociceptive information at the spinal, thalamic, and cortical levels. For the most part, these findings have failed to identify new therapeutic approaches. More recent basic science has better mirrored the clinical situation by addressing the pathophysiology associated with specific types of injury, refinement of methodology, and attention to various contributory factors such as sex. Improved quantification of sensory profiles in each patient and their distribution into defined clusters may improve translation between basic science and clinical practice. If such quantification can be traced back to cellular and molecular aspects of pathophysiology, this may lead to personalized medicine approaches that dictate a rational therapeutic approach for each individual.
Collapse
Affiliation(s)
- Peter A Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| |
Collapse
|
30
|
Jang K, Garraway SM. A review of dorsal root ganglia and primary sensory neuron plasticity mediating inflammatory and chronic neuropathic pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 15:100151. [PMID: 38314104 PMCID: PMC10837099 DOI: 10.1016/j.ynpai.2024.100151] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
Pain is a sensory state resulting from complex integration of peripheral nociceptive inputs and central processing. Pain consists of adaptive pain that is acute and beneficial for healing and maladaptive pain that is often persistent and pathological. Pain is indeed heterogeneous, and can be expressed as nociceptive, inflammatory, or neuropathic in nature. Neuropathic pain is an example of maladaptive pain that occurs after spinal cord injury (SCI), which triggers a wide range of neural plasticity. The nociceptive processing that underlies pain hypersensitivity is well-studied in the spinal cord. However, recent investigations show maladaptive plasticity that leads to pain, including neuropathic pain after SCI, also exists at peripheral sites, such as the dorsal root ganglia (DRG), which contains the cell bodies of sensory neurons. This review discusses the important role DRGs play in nociceptive processing that underlies inflammatory and neuropathic pain. Specifically, it highlights nociceptor hyperexcitability as critical to increased pain states. Furthermore, it reviews prior literature on glutamate and glutamate receptors, voltage-gated sodium channels (VGSC), and brain-derived neurotrophic factor (BDNF) signaling in the DRG as important contributors to inflammatory and neuropathic pain. We previously reviewed BDNF's role as a bidirectional neuromodulator of spinal plasticity. Here, we shift focus to the periphery and discuss BDNF-TrkB expression on nociceptors, non-nociceptor sensory neurons, and non-neuronal cells in the periphery as a potential contributor to induction and persistence of pain after SCI. Overall, this review presents a comprehensive evaluation of large bodies of work that individually focus on pain, DRG, BDNF, and SCI, to understand their interaction in nociceptive processing.
Collapse
Affiliation(s)
- Kyeongran Jang
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Sandra M. Garraway
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
31
|
Kerr PL, Gregg JM. The Roles of Endogenous Opioids in Placebo and Nocebo Effects: From Pain to Performance to Prozac. ADVANCES IN NEUROBIOLOGY 2024; 35:183-220. [PMID: 38874724 DOI: 10.1007/978-3-031-45493-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Placebo and nocebo effects have been well documented for nearly two centuries. However, research has only relatively recently begun to explicate the neurobiological underpinnings of these phenomena. Similarly, research on the broader social implications of placebo/nocebo effects, especially within healthcare delivery settings, is in a nascent stage. Biological and psychosocial outcomes of placebo/nocebo effects are of equal relevance. A common pathway for such outcomes is the endogenous opioid system. This chapter describes the history of placebo/nocebo in medicine; delineates the current state of the literature related to placebo/nocebo in relation to pain modulation; summarizes research findings related to human performance in sports and exercise; discusses the implications of placebo/nocebo effects among diverse patient populations; and describes placebo/nocebo influences in research related to psychopharmacology, including the relevance of endogenous opioids to new lines of research on antidepressant pharmacotherapies.
Collapse
Affiliation(s)
- Patrick L Kerr
- West Virginia University School of Medicine-Charleston, Charleston, WV, USA.
| | - John M Gregg
- Department of Surgery, VTCSOM, Blacksburg, VA, USA
| |
Collapse
|
32
|
Echeverria-Villalobos M, Tortorici V, Brito BE, Ryskamp D, Uribe A, Weaver T. The role of neuroinflammation in the transition of acute to chronic pain and the opioid-induced hyperalgesia and tolerance. Front Pharmacol 2023; 14:1297931. [PMID: 38161698 PMCID: PMC10755684 DOI: 10.3389/fphar.2023.1297931] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Current evidence suggests that activation of glial and immune cells leads to increased production of proinflammatory mediators, creating a neuroinflammatory state. Neuroinflammation has been proven to be a fundamental mechanism in the genesis of acute pain and its transition to neuropathic and chronic pain. A noxious event that stimulates peripheral afferent nerve fibers may also activate pronociceptive receptors situated at the dorsal root ganglion and dorsal horn of the spinal cord, as well as peripheral glial cells, setting off the so-called peripheral sensitization and spreading neuroinflammation to the brain. Once activated, microglia produce cytokines, chemokines, and neuropeptides that can increase the sensitivity and firing properties of second-order neurons, upregulating the signaling of nociceptive information to the cerebral cortex. This process, known as central sensitization, is crucial for chronification of acute pain. Immune-neuronal interactions are also implicated in the lesser-known complex regulatory relationship between pain and opioids. Current evidence suggests that activated immune and glial cells can alter neuronal function, induce, and maintain pathological pain, and disrupt the analgesic effects of opioid drugs by contributing to the development of tolerance and dependence, even causing paradoxical hyperalgesia. Such alterations may occur when the neuronal environment is impacted by trauma, inflammation, and immune-derived molecules, or when opioids induce proinflammatory glial activation. Hence, understanding these intricate interactions may help in managing pain signaling and opioid efficacy beyond the classical pharmacological approach.
Collapse
Affiliation(s)
| | - Victor Tortorici
- Neuroscience Laboratory, Faculty of Science, Department of Behavioral Sciences, Universidad Metropolitana, Caracas, Venezuela
- Neurophysiology Laboratory, Center of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| | - Beatriz E. Brito
- Immunopathology Laboratory, Center of Experimental Medicine, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| | - David Ryskamp
- College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Alberto Uribe
- Anesthesiology Department, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Tristan Weaver
- Anesthesiology Department, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
33
|
Perez-Sanchez J, Middleton SJ, Pattison LA, Hilton H, Awadelkareem MA, Zuberi SR, Renke MB, Hu H, Yang X, Clark AJ, Smith ESJ, Bennett DL. A humanized chemogenetic system inhibits murine pain-related behavior and hyperactivity in human sensory neurons. Sci Transl Med 2023; 15:eadh3839. [PMID: 37792955 PMCID: PMC7615191 DOI: 10.1126/scitranslmed.adh3839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Hyperexcitability in sensory neurons is known to underlie many of the maladaptive changes associated with persistent pain. Chemogenetics has shown promise as a means to suppress such excitability, yet chemogenetic approaches suitable for human applications are needed. PSAM4-GlyR is a modular system based on the human α7 nicotinic acetylcholine and glycine receptors, which responds to inert chemical ligands and the clinically approved drug varenicline. Here, we demonstrated the efficacy of this channel in silencing both mouse and human sensory neurons by the activation of large shunting conductances after agonist administration. Virally mediated expression of PSAM4-GlyR in mouse sensory neurons produced behavioral hyposensitivity upon agonist administration, which was recovered upon agonist washout. Stable expression of the channel led to similar reversible suppression of pain-related behavior even after 10 months of viral delivery. Mechanical and spontaneous pain readouts were also ameliorated by PSAM4-GlyR activation in acute and joint pain inflammation mouse models. Furthermore, suppression of mechanical hypersensitivity generated by a spared nerve injury model of neuropathic pain was also observed upon activation of the channel. Effective silencing of behavioral hypersensitivity was reproduced in a human model of hyperexcitability and clinical pain: PSAM4-GlyR activation decreased the excitability of human-induced pluripotent stem cell-derived sensory neurons and spontaneous activity due to a gain-of-function NaV1.7 mutation causing inherited erythromelalgia. Our results demonstrate the contribution of sensory neuron hyperexcitability to neuropathic pain and the translational potential of an effective, stable, and reversible humanized chemogenetic system for the treatment of pain.
Collapse
Affiliation(s)
- Jimena Perez-Sanchez
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Steven J. Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Luke A. Pattison
- Department of Pharmacology, University of Cambridge; Cambridge CB2 1PD, UK
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge; Cambridge CB2 1PD, UK
| | | | - Sana R. Zuberi
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Maria B. Renke
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Huimin Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Xun Yang
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Alex J. Clark
- Blizard Institute, Barts and the London School of Medicine and Dentistry; London E1 2AT, UK
| | | | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| |
Collapse
|
34
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
35
|
Ivasiuk A, Matvieienko M, Kononenko NI, Duzhyy DE, Korogod SM, Voitenko N, Belan P. Diabetes-Induced Amplification of Nociceptive DRG Neuron Output by Upregulation of Somatic T-Type Ca 2+ Channels. Biomolecules 2023; 13:1320. [PMID: 37759720 PMCID: PMC10526307 DOI: 10.3390/biom13091320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
The development of pain symptoms in peripheral diabetic neuropathy (PDN) is associated with the upregulation of T-type Ca2+ channels (T-channels) in the soma of nociceptive DRG neurons. Moreover, a block of these channels in DRG neurons effectively reversed mechanical and thermal hyperalgesia in animal diabetic models, indicating that T-channel functioning in these neurons is causally linked to PDN. However, no particular mechanisms relating the upregulation of T-channels in the soma of nociceptive DRG neurons to the pathological pain processing in PDN have been suggested. Here we have electrophysiologically identified voltage-gated currents expressed in nociceptive DRG neurons and developed a computation model of the neurons, including peripheral and central axons. Simulations showed substantially stronger sensitivity of neuronal excitability to diabetes-induced T-channel upregulation at the normal body temperature compared to the ambient one. We also found that upregulation of somatic T-channels, observed in these neurons under diabetic conditions, amplifies a single action potential invading the soma from the periphery into a burst of multiple action potentials further propagated to the end of the central axon. We have concluded that the somatic T-channel-dependent amplification of the peripheral nociceptive input to the spinal cord demonstrated in this work may underlie abnormal nociception at different stages of diabetes development.
Collapse
Affiliation(s)
- Arsentii Ivasiuk
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Maksym Matvieienko
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Nikolai I. Kononenko
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Dmytro E. Duzhyy
- Department of Sensory Signaling, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine;
| | - Sergiy M. Korogod
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Nana Voitenko
- Department of Biomedicine and Neuroscience, Kyiv Academic University of NAS of Ukraine, 03142 Kyiv, Ukraine
- Research Center, Dobrobut Academy Medical School, 03022 Kyiv, Ukraine
| | - Pavel Belan
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
- Department of Biomedicine and Neuroscience, Kyiv Academic University of NAS of Ukraine, 03142 Kyiv, Ukraine
| |
Collapse
|
36
|
Li Y, Uhelski ML, North RY, Mwirigi JM, Tatsui CE, Cata JP, Corrales G, Price TJ, Dougherty PM. MNK inhibitor eFT508 (Tomivosertib) suppresses ectopic activity in human dorsal root ganglion neurons from dermatomes with radicular neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.544811. [PMID: 37398249 PMCID: PMC10312735 DOI: 10.1101/2023.06.13.544811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Spontaneous activity in dorsal root ganglion (DRG) neurons is a key driver of neuropathic pain in preclinical models and in patients suffering from this largely untreated disease. While many intracellular signaling mechanisms have been examined in preclinical models that drive this spontaneous activity (SA), none of these have been tested directly on spontaneously active human nociceptors. Using cultured DRG neurons recovered during thoracic vertebrectomy surgeries, we show that inhibition of mitogen activated protein kinase interacting kinase (MNK) with eFT508 (25 nM) reverses SA in human sensory neurons associated with painful dermatomes. MNK inhibition in spontaneously active nociceptors decreased action potential amplitude and produced alterations in the magnitude of afterhyperpolarizing currents suggesting modification of Na+ and K+ channel activity downstream of MNK inhibition. The effects of MNK inhibition on SA took minutes to emerge and were reversible over time with eFT508 washout. MNK inhibition with eFT508 led to a profound loss of eIF4E Serine 209 phosphorylation, a specific target of the kinase, within 2 min of drug treatment, consistent with the rapid action of the drug on SA in electrophysiology experiments. Our results create a compelling case for the future testing of MNK inhibitors in clinical trials for neuropathic pain.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, 77030
| | - Megan L. Uhelski
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, 77030
| | - Robert Y. North
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, 77030
| | - Juliet M. Mwirigi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
- Center for Advanced Pain Studies, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
| | - Claudio E. Tatsui
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, 77030
| | - Juan P. Cata
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, 77030
| | - German Corrales
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, 77030
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
- Center for Advanced Pain Studies, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
| | - Patrick M. Dougherty
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, 77030
| |
Collapse
|
37
|
Mitchell ME, Cook LC, Shiers S, Tavares-Ferreira D, Akopian AN, Dussor G, Price TJ. Characterization of Fragile X Mental Retardation Protein expression in human nociceptors and their axonal projections to the spinal dorsal horn. J Comp Neurol 2023; 531:814-835. [PMID: 36808110 PMCID: PMC10038933 DOI: 10.1002/cne.25463] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023]
Abstract
Fragile X Mental Retardation Protein (FMRP) regulates activity-dependent RNA localization and local translation to modulate synaptic plasticity throughout the central nervous system. Mutations in the FMR1 gene that hinder or ablate FMRP function cause Fragile X Syndrome (FXS), a disorder associated with sensory processing dysfunction. FXS premutations are associated with increased FMRP expression and neurological impairments including sex dimorphic presentations of chronic pain. In mice, FMRP ablation causes dysregulated dorsal root ganglion (DRG) neuron excitability and synaptic vesicle exocytosis, spinal circuit activity, and decreased translation-dependent nociceptive sensitization. Activity-dependent, local translation is a key mechanism for enhancing primary nociceptor excitability that promotes pain in animals and humans. These works indicate that FMRP likely regulates nociception and pain at the level of the primary nociceptor or spinal cord. Therefore, we sought to better understand FMRP expression in the human DRG and spinal cord using immunostaining in organ donor tissues. We find that FMRP is highly expressed in DRG and spinal neuron subsets with substantia gelatinosa exhibiting the most abundant immunoreactivity in spinal synaptic fields. Here, it is expressed in nociceptor axons. FMRP puncta colocalized with Nav1.7 and TRPV1 receptor signals suggesting a pool of axoplasmic FMRP localizes to plasma membrane-associated loci in these branches. Interestingly, FMRP puncta exhibited notable colocalization with calcitonin gene-related peptide (CGRP) immunoreactivity selectively in female spinal cord. Our results support a regulatory role for FMRP in human nociceptor axons of the dorsal horn and implicate it in the sex dimorphic actions of CGRP signaling in nociceptive sensitization and chronic pain.
Collapse
Affiliation(s)
- Molly E Mitchell
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Lauren C Cook
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Stephanie Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Diana Tavares-Ferreira
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Armen N Akopian
- Department of Endodontics, UT Health San Antonio, San Antonio, Texas, USA
| | - Gregory Dussor
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Theodore J Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| |
Collapse
|
38
|
Tan E, Wang MQW, Leong S, Chou H, Too CW. Neurological pain relief interventional radiology procedures. Clin Radiol 2023; 78:254-264. [PMID: 35773096 DOI: 10.1016/j.crad.2022.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/29/2022] [Indexed: 11/24/2022]
Abstract
Neurological interventions have taken on a significant role in interventional radiology (IR) practice. Indications fall under three main categories: (1) intraprocedural pain management, (2) cancer pain palliation, and (3) chronic non-cancer pain control. Short-term regional anaesthesia can be achieved with local anaesthetics, while longer-term pain control can be attained with radiofrequency neuromodulation (pulsed or otherwise) or thermal/chemical neurolysis. This review article summarises the therapeutic options, applications, and techniques of commonly used peripheral nerve and plexus interventions in IR.
Collapse
Affiliation(s)
- E Tan
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
| | - M Q W Wang
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
| | - S Leong
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
| | - H Chou
- Department of Diagnostic Radiology, Khoo Teck Puat Hospital, 90 Yishun Central, Singapore, 768828, Singapore
| | - C W Too
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore.
| |
Collapse
|
39
|
Walters ET, Crook RJ, Neely GG, Price TJ, Smith ESJ. Persistent nociceptor hyperactivity as a painful evolutionary adaptation. Trends Neurosci 2023; 46:211-227. [PMID: 36610893 PMCID: PMC9974896 DOI: 10.1016/j.tins.2022.12.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/05/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023]
Abstract
Chronic pain caused by injury or disease of the nervous system (neuropathic pain) has been linked to persistent electrical hyperactivity of the sensory neurons (nociceptors) specialized to detect damaging stimuli and/or inflammation. This pain and hyperactivity are considered maladaptive because both can persist long after injured tissues have healed and inflammation has resolved. While the assumption of maladaptiveness is appropriate in many diseases, accumulating evidence from diverse species, including humans, challenges the assumption that neuropathic pain and persistent nociceptor hyperactivity are always maladaptive. We review studies indicating that persistent nociceptor hyperactivity has undergone evolutionary selection in widespread, albeit selected, animal groups as a physiological response that can increase survival long after bodily injury, using both highly conserved and divergent underlying mechanisms.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Robyn J Crook
- Department of Biology, San Francisco State University, San Francisco, CA 94132, USA
| | - G Gregory Neely
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| |
Collapse
|
40
|
Ray PR, Shiers S, Caruso JP, Tavares-Ferreira D, Sankaranarayanan I, Uhelski ML, Li Y, North RY, Tatsui C, Dussor G, Burton MD, Dougherty PM, Price TJ. RNA profiling of human dorsal root ganglia reveals sex differences in mechanisms promoting neuropathic pain. Brain 2023; 146:749-766. [PMID: 35867896 PMCID: PMC10169414 DOI: 10.1093/brain/awac266] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/16/2022] [Accepted: 06/22/2022] [Indexed: 11/12/2022] Open
Abstract
Neuropathic pain is a leading cause of high-impact pain, is often disabling and is poorly managed by current therapeutics. Here we focused on a unique group of neuropathic pain patients undergoing thoracic vertebrectomy where the dorsal root ganglia is removed as part of the surgery allowing for molecular characterization and identification of mechanistic drivers of neuropathic pain independently of preclinical models. Our goal was to quantify whole transcriptome RNA abundances using RNA-seq in pain-associated human dorsal root ganglia from these patients, allowing comprehensive identification of molecular changes in these samples by contrasting them with non-pain-associated dorsal root ganglia. We sequenced 70 human dorsal root ganglia, and among these 50 met inclusion criteria for sufficient neuronal mRNA signal for downstream analysis. Our expression analysis revealed profound sex differences in differentially expressed genes including increase of IL1B, TNF, CXCL14 and OSM in male and CCL1, CCL21, PENK and TLR3 in female dorsal root ganglia associated with neuropathic pain. Coexpression modules revealed enrichment in members of JUN-FOS signalling in males and centromere protein coding genes in females. Neuro-immune signalling pathways revealed distinct cytokine signalling pathways associated with neuropathic pain in males (OSM, LIF, SOCS1) and females (CCL1, CCL19, CCL21). We validated cellular expression profiles of a subset of these findings using RNAscope in situ hybridization. Our findings give direct support for sex differences in underlying mechanisms of neuropathic pain in patient populations.
Collapse
Affiliation(s)
- Pradipta R Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - James P Caruso
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA.,Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Megan L Uhelski
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Yan Li
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Y North
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Claudio Tatsui
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Michael D Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Patrick M Dougherty
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
41
|
Resurgent neuropathic discharge: an obstacle to the therapeutic use of neuroma resection? Pain 2023; 164:349-361. [PMID: 35639421 DOI: 10.1097/j.pain.0000000000002704] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023]
Abstract
ABSTRACT Ectopic discharge ("ectopia") in damaged afferent axons is a major contributor to chronic neuropathic pain. Clinical opinion discourages surgical resection of nerves proximal to the original injury site for fear of resurgence of ectopia and exacerbated pain. We tested this concept in a well-established animal neuroma model. Teased-fiber recordings were made of ectopic spontaneous discharge originating in the experimental nerve-end neuroma and associated dorsal root ganglia in rats that underwent either a single transection (with ligation) of the sciatic nerve or 2 consecutive transections separated by 7, 14, 21, or 30 days. Ectopia emerged in afferent A and C fibers after a single cut with kinetics anticipated from previous studies. When resection was performed during the early period of intense A-fiber activity, a brief period of resurgence was observed. However, resection of neuromas of more than 14 days was followed by low levels of activity with no indication of resurgence. This remained the case in trials out to 60 days after the first cut. Similarly, we saw no indication of resurgent ectopia originating in axotomized dorsal root ganglion neuronal somata and no behavioral reflection of resurgence. In summary, we failed to validate the concern that proximal resection of a problematic nerve would lead to intense resurgent ectopic discharge and pain. As the well-entrenched concept of resurgence is based more on case reports and anecdotes than on solid evidence, it may be justified to relax the stricture against resecting neuromas as a therapeutic strategy, at least within the framework of controlled clinical trials.
Collapse
|
42
|
Alsaloum M, Labau JIR, Liu S, Effraim PR, Waxman SG. Stem cell-derived sensory neurons modelling inherited erythromelalgia: normalization of excitability. Brain 2023; 146:359-371. [PMID: 35088838 DOI: 10.1093/brain/awac031] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 01/11/2023] Open
Abstract
Effective treatment of pain remains an unmet healthcare need that requires new and effective therapeutic approaches. NaV1.7 has been genetically and functionally validated as a mediator of pain. Preclinical studies of NaV1.7-selective blockers have shown limited success and translation to clinical studies has been limited. The degree of NaV1.7 channel blockade necessary to attenuate neuronal excitability and ameliorate pain is an unanswered question important for drug discovery. Here, we utilize dynamic clamp electrophysiology and induced pluripotent stem cell-derived sensory neurons (iPSC-SNs) to answer this question for inherited erythromelalgia, a pain disorder caused by gain-of-function mutations in Nav1.7. We show that dynamic clamp can produce hyperexcitability in iPSC-SNs associated with two different inherited erythromelalgia mutations, NaV1.7-S241T and NaV1.7-I848T. We further show that blockade of approximately 50% of NaV1.7 currents can reverse neuronal hyperexcitability to baseline levels.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT 06510, USA.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA
| | - Julie I R Labau
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Shujun Liu
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Philip R Effraim
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
43
|
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
|
44
|
Yang H, Shan Z, Guo W, Wang Y, Cai S, Li F, Huang Q, Liu JA, Cheung CW, Cai S. Reversal of Peripheral Neuropathic Pain by the Small-Molecule Natural Product Narirutin via Block of Na v1.7 Voltage-Gated Sodium Channel. Int J Mol Sci 2022; 23:ijms232314842. [PMID: 36499167 PMCID: PMC9738487 DOI: 10.3390/ijms232314842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/12/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Neuropathic pain is a refractory chronic disease affecting millions of people worldwide. Given that present painkillers have poor efficacy or severe side effects, developing novel analgesics is badly needed. The multiplex structure of active ingredients isolated from natural products provides a new source for phytochemical compound synthesis. Here, we identified a natural product, Narirutin, a flavonoid compound isolated from the Citrus unshiu, showing antinociceptive effects in rodent models of neuropathic pain. Using calcium imaging, whole-cell electrophysiology, western blotting, and immunofluorescence, we uncovered a molecular target for Narirutin's antinociceptive actions. We found that Narirutin (i) inhibits Veratridine-triggered nociceptor activities in L4-L6 rat dorsal root ganglion (DRG) neurons, (ii) blocks voltage-gated sodium (NaV) channels subtype 1.7 in both small-diameter DRG nociceptive neurons and human embryonic kidney (HEK) 293 cell line, (iii) does not affect tetrodotoxin-resistant (TTX-R) NaV channels, and (iv) blunts the upregulation of Nav1.7 in calcitonin gene-related peptide (CGRP)-labeled DRG sensory neurons after spared nerve injury (SNI) surgery. Identifying Nav1.7 as a molecular target of Narirutin may further clarify the analgesic mechanism of natural flavonoid compounds and provide an optimal idea to produce novel selective and efficient analgesic drugs.
Collapse
Affiliation(s)
- Haoyi Yang
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Zhiming Shan
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Department of Anesthesiology, Shenzhen People’s Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen 518020, China
- Shenzhen Engineering Research Center of Anesthesiology, Shenzhen 518020, China
| | - Weijie Guo
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Yuwei Wang
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Shuxian Cai
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Fuyi Li
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Qiaojie Huang
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Jessica Aijia Liu
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
| | - Chi Wai Cheung
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Correspondence: (C.W.C.); (S.C.)
| | - Song Cai
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
- Correspondence: (C.W.C.); (S.C.)
| |
Collapse
|
45
|
Zhu YF, Kan P, Singh G. Differences and Similarities in Spontaneous Activity Between Animal Models of Cancer-Induced Pain and Neuropathic Pain. J Pain Res 2022; 15:3179-3187. [PMID: 36258759 PMCID: PMC9572504 DOI: 10.2147/jpr.s383373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022] Open
Abstract
Background Clinical data on cancer-induced pain (CIP) demonstrate widespread changes in sensory function. It is characterized in humans not only by stimulus-invoked pain, but also by spontaneous pain. In our previous studies in an animal model of CIP, we observed changes in intrinsic membrane properties and excitability of dorsal root ganglion (DRG) sensory neurons corresponding to mechanical allodynia and hyperalgesia, of which abnormal activities of Aβ-fiber sensory neurons are consistent in a rat model of peripheral neuropathic pain (NEP). Objective To investigate whether there are related peripheral neural mechanisms between the CIP and NEP models of spontaneous pain, we compared the electrophysiological properties of DRG sensory neurons at 2–3 weeks after CIP and NEP model induction. Methods CIP models were induced with metastasis tumour-1 rat breast cancer cells implanted into the distal epiphysis of the femur. NEP models were induced with a polyethylene cuff implanted around the sciatic nerve. Spontaneous pain in animals is measured by spontaneous foot lifting (SFL). After measurement of SFL, the animals were prepared for electrophysiological recordings of spontaneous activity (SA) in DRG neurons in vivo. Results Our data showed that SFL and SA occurred in both models. The proportion of SFL and SA of C-fiber sensory neurons in CIP was more significantly increased than in NEP models. There was no difference in duration of SFL and the rate of SA between the two models. The duration of SFL is related to the rate of SA in C-fiber in both models. Conclusion Thus, SFL may result from SA activity in C-fiber neurons in CIP and NEP rats. The differences and similarities in spontaneous pain between CIP and NEP rats is related to the proportion and rate of SA in C-fibers, respectively.
Collapse
Affiliation(s)
- Yong Fang Zhu
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Peter Kan
- Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Gurmit Singh
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, ON, Canada,Correspondence: Gurmit Singh, Email
| |
Collapse
|
46
|
North RY, Odem MA, Li Y, Tatsui CE, Cassidy RM, Dougherty PM, Walters ET. Electrophysiological Alterations Driving Pain-Associated Spontaneous Activity in Human Sensory Neuron Somata Parallel Alterations Described in Spontaneously Active Rodent Nociceptors. THE JOURNAL OF PAIN 2022; 23:1343-1357. [PMID: 35292377 PMCID: PMC9357108 DOI: 10.1016/j.jpain.2022.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 06/10/2023]
Abstract
Neuropathic pain in rodents can be driven by ectopic spontaneous activity (SA) generated by sensory neurons in dorsal root ganglia (DRG). The recent demonstration that SA in dissociated human DRG neurons is associated with reported neuropathic pain in patients enables a detailed comparison of pain-linked electrophysiological alterations driving SA in human DRG neurons to alterations that distinguish SA in nociceptors from SA in low-threshold mechanoreceptors (LTMRs) in rodent neuropathy models. Analysis of recordings from dissociated somata of patient-derived DRG neurons showed that SA and corresponding pain in both sexes were significantly associated with the three functional electrophysiological alterations sufficient to generate SA in the absence of extrinsic depolarizing inputs. These include enhancement of depolarizing spontaneous fluctuations of membrane potential (DSFs), which were analyzed quantitatively for the first time in human DRG neurons. The functional alterations were indistinguishable from SA-driving alterations reported for nociceptors in rodent chronic pain models. Irregular, low-frequency DSFs in human DRG neurons closely resemble DSFs described in rodent nociceptors while differing substantially from the high-frequency sinusoidal oscillations described in rodent LTMRs. These findings suggest that conserved physiological mechanisms of SA in human nociceptor somata can drive neuropathic pain despite documented cellular differences between human and rodent DRG neurons. PERSPECTIVE: Electrophysiological alterations in human sensory neurons associated with patient-reported neuropathic pain include all three of the functional alterations that logically can promote spontaneous activity. The similarity of distinctively altered spontaneous depolarizations in human DRG neurons and rodent nociceptors suggests that spontaneously active human nociceptors can persistently promote neuropathic pain in patients.
Collapse
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Max A Odem
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, Texas
| | - Yan Li
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Claudio Esteves Tatsui
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ryan M Cassidy
- M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Patrick M Dougherty
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas..
| |
Collapse
|
47
|
Goodwin G, McMurray S, Stevens EB, Denk F, McMahon SB. Examination of the contribution of Nav1.7 to axonal propagation in nociceptors. Pain 2022; 163:e869-e881. [PMID: 34561392 DOI: 10.1097/j.pain.0000000000002490] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Nav1.7 is a promising drug target for the treatment of pain. However, there is a mismatch between the analgesia produced by Nav1.7 loss-of-function and the peripherally restricted Nav1.7 inhibitors, which may reflect a lack of understanding of the function of Nav1.7 in the transmission of nociceptive information. In the periphery, the role of Nav1.7 in transduction at nociceptive peripheral terminals has been comprehensively examined, but its role in axonal propagation in these neurons is less clearly defined. In this study, we examined the contribution of Nav1.7 to axonal propagation in nociceptors using sodium channel blockers in in vivo electrophysiological and calcium imaging recordings in mice. Using the sodium channel blocker tetrodotoxin (TTX) (1-10 µM) to inhibit Nav1.7 and other tetrodotoxin-sensitive sodium channels along the sciatic nerve, we first showed that around two-thirds of nociceptive L4 dorsal root ganglion neurons innervating the skin, but a lower proportion innervating the muscle (45%), are blocked by TTX. By contrast, nearly all large-sized cutaneous afferents (95%-100%) were blocked by axonal TTX. Many cutaneous nociceptors resistant to TTX were polymodal (57%) and capsaicin sensitive (57%). Next, we applied PF-05198007 (300 nM-1 µM) to the sciatic nerve between stimulating and recording sites to selectively block axonal Nav1.7 channels. One hundred to three hundred nanomolar PF-05198007 blocked propagation in 63% of C-fiber sensory neurons, whereas similar concentrations produced minimal block (5%) in rapidly conducting A-fiber neurons. We conclude that Nav1.7 is essential for axonal propagation in around two-thirds of nociceptive cutaneous C-fiber neurons and a lower proportion (≤45%) of nociceptive neurons innervating muscle.
Collapse
Affiliation(s)
- George Goodwin
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, United Kingdom
| | | | | | - Franziska Denk
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, United Kingdom
| | - Stephen B McMahon
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, United Kingdom
| |
Collapse
|
48
|
Peripheral Trigeminal Nerve Blocks for Chronic Orbital Pain: Clinical Features and Outcomes. Ophthalmic Plast Reconstr Surg 2022; 38:369-376. [DOI: 10.1097/iop.0000000000002120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
49
|
Abdus-Saboor I, Luo W. Measuring Mouse Somatosensory Reflexive Behaviors with High-speed Videography, Statistical Modeling, and Machine Learning. NEUROMETHODS 2022; 178:441-456. [PMID: 35783537 PMCID: PMC9249079 DOI: 10.1007/978-1-0716-2039-7_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Objectively measuring and interpreting an animal's sensory experience remains a challenging task. This is particularly true when using preclinical rodent models to study pain mechanisms and screen for potential new pain treatment reagents. How to determine their pain states in a precise and unbiased manner is a hurdle that the field will need to overcome. Here, we describe our efforts to measure mouse somatosensory reflexive behaviors with greatly improved precision by high-speed video imaging. We describe how coupling sub-second ethograms of reflexive behaviors with a statistical reduction method and supervised machine learning can be used to create a more objective quantitative mouse "pain scale." Our goal is to provide the readers with a protocol of how to integrate some of the new tools described here with currently used mechanical somatosensory assays, while discussing the advantages and limitations of this new approach.
Collapse
Affiliation(s)
- Ishmail Abdus-Saboor
- Department of Biology, University of Pennsylvania, 3740 Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Wenqin Luo
- Department of Neuroscience, University of Pennsylvania, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA
| |
Collapse
|
50
|
Ishida K, Tanaka S, Shen D, Matsui S, Fuseya S, Shindo T, Kawamata M. Calcitonin gene-related peptide is not involved in neuropathic pain induced by partial sciatic nerve ligation in mice. Neurosci Lett 2022; 778:136615. [DOI: 10.1016/j.neulet.2022.136615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022]
|