1
|
Khan MG, Hussain SHA, Alkhayl FFA, Ahsan M, Ridha-Salman H. Cannabinoids in neuropathic pain treatment: pharmacological insights and clinical outcomes from recent trials. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04134-7. [PMID: 40261351 DOI: 10.1007/s00210-025-04134-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Neuropathic pain, a complex and often devastating condition, poses significant challenges for its effective management. Despite promising research on various cannabis formulations and delivery methods for neuropathic pain, significant gaps remain in our knowledge. While inhaled cannabis shows analgesic effects and alternative delivery methods may improve bioavailability, oral formulations have yielded mixed results, often limited by small sample sizes and placebo effects. Therefore, further research is essential to optimize cannabis formulations, identify responder profiles to tailor treatments effectively, and, most critically, confirm the long-term safety and efficacy of cannabis-based therapies in managing NP. This review article aims to provide a comprehensive analysis of the therapeutic potential of cannabis-based medicines, with a particular focus on cannabinoids. This review, though not systematic, examines 11 clinical studies, specifically Randomised Clinical Trials) published from 2014 to 2024, highlighting the efficacy of numerous cannabis formulations, in alleviating neuropathic pain. Key findings show that cannabinoids can reduce pain perception, improve patient quality of life, and mitigate other symptoms associated with neuropathic pain. The synergistic effects of tetrahydrocannabinol and cannabidiol are discussed, emphasizing their ability to enhance analgesic effects, while potentially reducing the psychoactive side effects of tetrahydrocannabinol. This review emphasizes the importance of the personalized approach to improve therapeutic outcomes. Limitations of the existing research focusing on cannabis for neuropathic pain are limited by heterogeneity, lack of standardization, small sample sizes, and reliance on subjective outcomes, impacting the reliability and generalizability of findings. However, this exhaustive review aims to inform clinicians and researchers about the evolving role of cannabis in contemporary pain management strategies, illustrating the diverse pharmacological profiles of cannabinoids and their potential as adjunct therapies for neuropathic pain management.
Collapse
Affiliation(s)
- Mohammad Gayoor Khan
- Department of Pharmacology, Daksh Institute of Pharmaceutical Science, Chhatarpur Madhya Pradesh, General Secretary Society of Clinical Research and Medical Professionals, Hyderabad, 500059, India
| | - Sajid Hussain Altaf Hussain
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, 11952, Majmaah, Saudi Arabia.
| | - Faris F Aba Alkhayl
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Marya Ahsan
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Hayder Ridha-Salman
- College of Pharmacy, Department of Pharmacology, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
2
|
Yogi A, Banderali U, Moreno MJ, Martina M. Preclinical Animal Models to Investigate the Role of Na v1.7 Ion Channels in Pain. Life (Basel) 2025; 15:640. [PMID: 40283194 PMCID: PMC12028925 DOI: 10.3390/life15040640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Chronic pain is a maladaptive neurological disease that remains a major global healthcare problem. Voltage-gated sodium channels (Navs) are major drivers of the excitability of sensory neurons, and the Nav subtype 1.7 (Nav1.7) has been shown to be critical for the transmission of pain-related signaling. This is highlighted by demonstrations that gain-of-function mutations in the Nav1.7 gene SCN9A result in various pain pathologies, whereas loss-of-function mutations cause complete insensitivity to pain. A substantial body of evidence demonstrates that chronic neuropathy and inflammation result in an upregulation of Nav1.7, suggesting that this channel contributes to pain transmission and sensation. As such, Nav1.7 is an attractive human-validated target for the treatment of pain. Nonetheless, a lack of subtype selectivity, insufficient efficacy, and adverse reactions are some of the issues that have hindered Nav1.7-targeted drug development. This review summarizes the pain behavior profiles mediated by Nav1.7 reported in multiple preclinical models, outlining the current knowledge of the biophysical, physiological, and distribution properties required for a Nav1.7 inhibitor to produce analgesia.
Collapse
Affiliation(s)
- Alvaro Yogi
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada; (U.B.); (M.J.M.); (M.M.)
| | | | | | | |
Collapse
|
3
|
Alves-Simões M, Teege L, Tomni C, Lürkens M, Schmidt A, Iseppon F, Millet Q, Kühs S, Katona I, Weis J, Heinemann SH, Hübner CA, Wood J, Leipold E, Kurth I, Haag N. Na V 1.8/Na V 1.9 double deletion mildly affects acute pain responses in mice. Pain 2025; 166:773-792. [PMID: 39382328 PMCID: PMC11921451 DOI: 10.1097/j.pain.0000000000003411] [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: 11/20/2023] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT The 2 tetrodotoxin-resistant (TTXr) voltage-gated sodium channel subtypes Na V 1.8 and Na V 1.9 are important for peripheral pain signaling. As determinants of sensory neuron excitability, they are essential for the initial transduction of sensory stimuli, the electrogenesis of the action potential, and the release of neurotransmitters from sensory neuron terminals. Na V 1.8 and Na V 1.9, which are encoded by SCN10A and SCN11A , respectively, are predominantly expressed in pain-sensitive (nociceptive) neurons localized in the dorsal root ganglia (DRG) along the spinal cord and in the trigeminal ganglia. Mutations in these genes cause various pain disorders in humans. Gain-of-function missense variants in SCN10A result in small fiber neuropathy, while distinct SCN11A mutations cause, i. a., congenital insensitivity to pain, episodic pain, painful neuropathy, and cold-induced pain. To determine the impact of loss-of-function of both channels, we generated Na V 1.8/Na V 1.9 double knockout (DKO) mice using clustered regularly interspaced short palindromic repeats/Cas-mediated gene editing to achieve simultaneous gene disruption. Successful knockout of both channels was verified by whole-cell recordings demonstrating the absence of Na V 1.8- and Na V 1.9-mediated Na + currents in Na V 1.8/Na V 1.9 DKO DRG neurons. Global RNA sequencing identified significant deregulation of C-LTMR marker genes as well as of pain-modulating neuropeptides in Na V 1.8/Na V 1.9 DKO DRG neurons, which fits to the overall only moderately impaired acute pain behavior observed in DKO mice. Besides addressing the function of both sodium channels in pain perception, we further demonstrate that the null-background is a very valuable tool for investigations on the functional properties of individual human disease-causing variants in Na V 1.8 or Na V 1.9 in their native physiological environment.
Collapse
Affiliation(s)
- Marta Alves-Simões
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London, United Kingdom
| | - Laura Teege
- Center of Brain, Behavior and Metabolism & Clinic for Anesthesiology and Intensive Care, University of Lübeck, Lübeck, Germany
| | - Cecilia Tomni
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Martha Lürkens
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Annika Schmidt
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London, United Kingdom
| | - Queensta Millet
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London, United Kingdom
| | - Samuel Kühs
- Center of Brain, Behavior and Metabolism & Clinic for Anesthesiology and Intensive Care, University of Lübeck, Lübeck, Germany
| | - Istvan Katona
- Institute of Neuropathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Joachim Weis
- Institute of Neuropathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Stefan H. Heinemann
- Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital, Jena, Germany
| | - Christian A. Hübner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - John Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London, United Kingdom
| | - Enrico Leipold
- Center of Brain, Behavior and Metabolism & Clinic for Anesthesiology and Intensive Care, University of Lübeck, Lübeck, Germany
| | - Ingo Kurth
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Natja Haag
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
4
|
Duan L, Wang Q, Chen J, Fan Z, Zhang W, Yan J. Unraveling the PVT Glu-mPFC Glu circuit: A new frontier in chronic pain management for bone cancer pain. Brain Res Bull 2025; 222:111235. [PMID: 39922506 DOI: 10.1016/j.brainresbull.2025.111235] [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: 10/10/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
Bone cancer pain (BCP) is a type of ongoing or breakthrough pain caused by a primary bone tumor or bone metastasis. BCP impairs patients' quality of life. Depending upon clinical observations, the administration of centrally acting analgesic has been associated with the alleviation of pain symptoms BCP patients. Central nervous system sensitization performs a crucial role in pain-regulating perception in BCP. Nevertheless, the precise neural circuitry and mechanism of action remain enigmatic. In the present study, we observed the activation of glutamatergic neurons in the Prelimbic cortex (mPFC) and paraventricular thalamus (PVT) in BCP mice. Experimental validation using viral tracers confirmed the existence of a projection pathway between the PVT and mPFC. Inhibition of the input from PVT glutamatergic neurons to mPFC glutamatergic neurons alleviates chronic pain in BCP, whereas activation of the PVTGlu-mPFCGlu projection induces chronic pain in mice. These findings imply a pivotal role for the PVTGlu-mPFCGlu circuit in the regulation of chronic pain in BCP.
Collapse
Affiliation(s)
- Liqun Duan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China; Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qianliang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Jianpeng Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Zelin Fan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Wenzhi Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Jun Yan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China.
| |
Collapse
|
5
|
Karcz M, Abd-Elsayed A, Chakravarthy K, Aman MM, Strand N, Malinowski MN, Latif U, Dickerson D, Suvar T, Lubenow T, Peskin E, D’Souza R, Cornidez E, Dudas A, Lam C, Farrell II M, Sim GY, Sebai M, Garcia R, Bracero L, Ibrahim Y, Mahmood SJ, Lawandy M, Jimenez D, Shahgholi L, Sochacki K, Ramadan ME, Tieppo Francio V, Sayed D, Deer T. Pathophysiology of Pain and Mechanisms of Neuromodulation: A Narrative Review (A Neuron Project). J Pain Res 2024; 17:3757-3790. [PMID: 39583192 PMCID: PMC11581984 DOI: 10.2147/jpr.s475351] [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] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
Pain serves as a vital innate defense mechanism that can significantly impact an individual's quality of life. Understanding the physiological effects of pain well plays an important role in developing novel pain treatments. Nociceptor neurons play a key role in pain and inflammation. Interactions between nociceptors and the immune system occur both at the site of injury and within the central nervous system. Modulating chemical mediators and nociceptor activity offers promising new approaches to pain management. Essentially, the sensory nervous system is essential for modulating the body's protective response, making it critical to understand these interactions to discover new pain treatment strategies. New innovations in neuromodulation have led to alternatives to opioids individuals with chronic pain with consequent improvement in disease-based treatment and nerve targeting. New neural targets from cellular and structural perspectives have revolutionized the field of neuromodulation. This narrative review aims to elucidate the mechanisms of pain transmission and processing, examine the characteristics and properties of nociceptors, and explore how the immune system influences pain perception. It further provides an updated overview of the physiology of pain and neuromodulatory mechanisms essential for managing acute and chronic pain. We assess the current understanding of different pain types, focusing on key molecules involved in each type and their physiological effects. Additionally, we compare painful and painless neuropathies and discuss the neuroimmune interactions involved in pain manifestation.
Collapse
Affiliation(s)
- Marcin Karcz
- The Spine and Nerve Centers of the Virginias, Charleston, WV, USA
| | - Alaa Abd-Elsayed
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA
| | | | - Mansoor M Aman
- Aurora Pain Management, Aurora Health Care, Oshkosh, WI, USA
| | - Natalie Strand
- Anesthesiology and Perioperative Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Mark N Malinowski
- OhioHealth Neurological Physicians, OhioHealth Inc, Columbus, OH, USA
| | - Usman Latif
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - David Dickerson
- Department of Pain Medicine, Northshore University Health System, Skokie, IL, USA
| | - Tolga Suvar
- Department of Anesthesiology and Pain Medicine, Rush University Medical Center, Oak Park, IL, USA
| | - Timothy Lubenow
- Department of Anesthesiology and Pain Medicine, Rush University Medical Center, Oak Park, IL, USA
| | - Evan Peskin
- Department of Pain Management, Insight Institute of Neurosurgery & Neuroscience, Flint, MI, USA
| | - Ryan D’Souza
- Anesthesiology and Perioperative Medicine, Mayo Clinic, Phoenix, AZ, USA
| | | | - Andrew Dudas
- Mays and Schnapp Neurospine and Pain, Memphis, TN, USA
| | - Christopher Lam
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michael Farrell II
- Department of Pain Management, Erie County Medical Center, Buffalo, NY, USA
| | - Geum Yeon Sim
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Mohamad Sebai
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rosa Garcia
- Department of Physical Medicine & Rehabilitation, Larkin Hospital Health System, Miami, FL, USA
| | - Lucas Bracero
- The Spine and Nerve Centers of the Virginias, Charleston, WV, USA
| | - Yussr Ibrahim
- Department of Pain Management at Northern Light Health – Eastern Maine Medical Center, Bangor, ME, USA
| | - Syed Jafar Mahmood
- Department of Pain Medicine, University of California Davis Health System, Sacramento, CA, USA
| | - Marco Lawandy
- Department of Physical Medicine & Rehabilitation, Montefiore Medical Center, Bronx, NY, USA
| | - Daniel Jimenez
- Department of Physical Medicine & Rehabilitation, Michigan State University, Lansing, MI, USA
| | - Leili Shahgholi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kamil Sochacki
- Department of Anesthesiology and Perioperative Medicine, Rutgers Robert Wood Johnson, New Brunswick, NJ, USA
| | - Mohamed Ehab Ramadan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vinicius Tieppo Francio
- Division of Pain Medicine, Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Dawood Sayed
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Timothy Deer
- The Spine and Nerve Centers of the Virginias, Charleston, WV, USA
| |
Collapse
|
6
|
Suhandi C, Wilar G, Narsa AC, Mohammed AFA, El-Rayyes A, Muchtaridi M, Shamsuddin S, Safuan S, Wathoni N. Updating the Pharmacological Effects of α-Mangostin Compound and Unraveling Its Mechanism of Action: A Computational Study Review. Drug Des Devel Ther 2024; 18:4723-4748. [PMID: 39469723 PMCID: PMC11514645 DOI: 10.2147/dddt.s478388] [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: 05/15/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024] Open
Abstract
α-Mangostin, initially identified in 1855, is a xanthone derivative compound predominantly located in the pericarp of the mangosteen fruit (Garcinia mangostana L). This compound is known for its beneficial properties as an antioxidant and anti-inflammatory agent, still holding promise for potential benefits in other related pathologies. In the investigative process, computational studies have proven highly valuable in providing evidence and initial screening before progressing to preclinical and clinical studies. This review aims to present the pharmacological findings and mechanisms of action of α-mangostin based on computational studies. The compilation of this review is founded on the analysis of relevant articles obtained from PubMed, Scopus, and ScienceDirect databases. The study commences with an elucidation of the physicochemical characteristics, drug-likeness, pharmacokinetics, and toxicity profile of α-mangostin, which demonstrates that α-mangostin complies with the Lipinski's Rule of Five, exhibits favorable profiles of absorption, distribution, metabolism, and excretion, and presents low toxicity. Subsequent investigations have revealed that computational studies employing various software tools including ArgusLab, AutoDock, AutoDock Vina, Glide, HEX, and MOE, have been pivotal to comprehend the pharmacology of α-mangostin. Beyond its well established roles as an antioxidant and anti-inflammatory agent, α-mangostin is now recognized for its pharmacological effects in Alzheimer's disease, diabetes, cancer, chronic kidney disease, chronic periodontitis, infectious diseases, and rheumatoid arthritis. Moreover, α-mangostin is projected to have applications in pain management and as a potent mosquito larvicide. All of these findings are based on the attainment of adequate binding affinity to specific target receptors associated with each respective pathological condition. Consequently, it is anticipated that these findings will serve as a foundation for future scientific endeavours, encompassing both in vitro and in vivo studies, as well as clinical investigations, to better understand the pharmacological effects of α-mangostin.
Collapse
Affiliation(s)
- Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Angga Cipta Narsa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Mulawarman University, Samarinda, 71157, Indonesia
| | | | - Ali El-Rayyes
- Department of Chemistry, College of Science, Northern Border University, Arar, Saudi Arabia
| | - Muchtaridi Muchtaridi
- Department of Analytical Pharmacy and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Sabreena Safuan
- Department of Biomedicine, School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| |
Collapse
|
7
|
Dai SP, Yang CC, Chin Y, Sun WH. T cell death-associated gene 8-mediated distinct signaling pathways modulate the early and late phases of neuropathic pain. iScience 2024; 27:110955. [PMID: 39381739 PMCID: PMC11460492 DOI: 10.1016/j.isci.2024.110955] [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: 11/06/2023] [Revised: 05/01/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
Peripheral nerve injury alters the transduction of nociceptive signaling. The coordination of neurons, glia, and immune cells results in persistent pain and inflammation. T cell death-associated gene 8 (TDAG8), located at nociceptors and immune cells, is involved in inflammatory pain and arthritis-induced pain. Here, we employed TDAG8-deficient mice, pharmacological approaches, and calcium/sodium imaging to elucidate how TDAG8-mediated signaling modulates neuron activities in a mouse model of chronic constriction injury-induced neuropathic pain. We demonstrated that TDAG8 participated alone in mechanical allodynia induced by constriction injury. (1) TDAG8-Nav1.8 signaling in small-diameter isolectin B4-positive [IB4(+)] neurons initiates mechanical allodynia; it also modulated substance P release from IB4(-) neurons to facilitate the development of early mechanical allodynia. (2) TDAG8-mediated signaling increased medium-to large-diameter IB4(-) neuron activity to maintain late mechanical allodynia; it also modulated substance P release in soma to reduce satellite glial number and Nav1.7 expression, thus attenuating chronic mechanical allodynia.
Collapse
Affiliation(s)
- Shih-Ping Dai
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Chieh Yang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yin Chin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Hsin Sun
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| |
Collapse
|
8
|
Lewis CM, Griffith TN. Ion channels of cold transduction and transmission. J Gen Physiol 2024; 156:e202313529. [PMID: 39051992 PMCID: PMC11273221 DOI: 10.1085/jgp.202313529] [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: 12/15/2023] [Revised: 06/04/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Thermosensation requires the activation of a unique collection of ion channels and receptors that work in concert to transmit thermal information. It is widely accepted that transient receptor potential melastatin 8 (TRPM8) activation is required for normal cold sensing; however, recent studies have illuminated major roles for other ion channels in this important somatic sensation. In addition to TRPM8, other TRP channels have been reported to contribute to cold transduction mechanisms in diverse sensory neuron populations, with both leak- and voltage-gated channels being identified for their role in the transmission of cold signals. Whether the same channels that contribute to physiological cold sensing also mediate noxious cold signaling remains unclear; however, recent work has found a conserved role for the kainite receptor, GluK2, in noxious cold sensing across species. Additionally, cold-sensing neurons likely engage in functional crosstalk with nociceptors to give rise to cold pain. This Review will provide an update on our understanding of the relationship between various ion channels in the transduction and transmission of cold and highlight areas where further investigation is required.
Collapse
Affiliation(s)
- Cheyanne M Lewis
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Theanne N Griffith
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
9
|
Haroun R, Gossage SJ, Iseppon F, Fudge A, Caxaria S, Arcangeletti M, Leese C, Davletov B, Cox JJ, Sikandar S, Welsh F, Chessell IP, Wood JN. Novel therapies for cancer-induced bone pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100167. [PMID: 39399223 PMCID: PMC11470602 DOI: 10.1016/j.ynpai.2024.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 10/15/2024]
Abstract
Cancer pain is a growing problem, especially with the substantial increase in cancer survival. Reports indicate that bone metastasis, whose primary symptom is bone pain, occurs in 65-75% of patients with advanced breast or prostate cancer. We optimized a preclinical in vivo model of cancer-induced bone pain (CIBP) involving the injection of Lewis Lung Carcinoma cells into the intramedullary space of the femur of C57BL/6 mice or transgenic mice on a C57BL/6 background. Mice gradually reduce the use of the affected limb, leading to altered weight bearing. Symptoms of secondary cutaneous heat sensitivity also manifest themselves. Following optimization, three potential analgesic treatments were assessed; 1) single ion channel targets (targeting the voltage-gated sodium channels NaV1.7, NaV1.8, or acid-sensing ion channels), 2) silencing µ-opioid receptor-expressing neurons by modified botulinum compounds, and 3) targeting two inflammatory mediators simultaneously (nerve growth factor (NGF) and tumor necrosis factor (TNF)). Unlike global NaV1.8 knockout mice which do not show any reduction in CIBP-related behavior, embryonic conditional NaV1.7 knockout mice in sensory neurons exhibit a mild reduction in CIBP-linked behavior. Modified botulinum compounds also failed to cause a detectable analgesic effect. In contrast, inhibition of NGF and/or TNF resulted in a significant reduction in CIBP-driven weight-bearing alterations and prevented the development of secondary cutaneous heat hyperalgesia. Our results support the inhibition of these inflammatory mediators, and more strongly their dual inhibition to treat CIBP, given the superiority of combination therapies in extending the time needed to reach limb use score zero in our CIBP model.
Collapse
Affiliation(s)
- Rayan Haroun
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London WC1E 6BT, United Kingdom
| | - Samuel J. Gossage
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London WC1E 6BT, United Kingdom
| | - Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London WC1E 6BT, United Kingdom
| | - Alexander Fudge
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London WC1E 6BT, United Kingdom
| | - Sara Caxaria
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Manuel Arcangeletti
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London WC1E 6BT, United Kingdom
| | - Charlotte Leese
- Department of Biomedical Science, University of Sheffield, South Yorkshire S10 2TN, United Kingdom
| | - Bazbek Davletov
- Department of Biomedical Science, University of Sheffield, South Yorkshire S10 2TN, United Kingdom
| | - James J. Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London WC1E 6BT, United Kingdom
| | - Shafaq Sikandar
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Fraser Welsh
- AstraZeneca BioPharmaceuticals R&D, Neuroscience, Discovery Centre, Biomedical campus, 1 Francis Crick Ave, Cambridge CB2 0AA, United Kingdom
| | - Iain P. Chessell
- AstraZeneca BioPharmaceuticals R&D, Neuroscience, Discovery Centre, Biomedical campus, 1 Francis Crick Ave, Cambridge CB2 0AA, United Kingdom
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London WC1E 6BT, United Kingdom
| |
Collapse
|
10
|
Cao B, Xu Q, Shi Y, Zhao R, Li H, Zheng J, Liu F, Wan Y, Wei B. Pathology of pain and its implications for therapeutic interventions. Signal Transduct Target Ther 2024; 9:155. [PMID: 38851750 PMCID: PMC11162504 DOI: 10.1038/s41392-024-01845-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] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/10/2024] Open
Abstract
Pain is estimated to affect more than 20% of the global population, imposing incalculable health and economic burdens. Effective pain management is crucial for individuals suffering from pain. However, the current methods for pain assessment and treatment fall short of clinical needs. Benefiting from advances in neuroscience and biotechnology, the neuronal circuits and molecular mechanisms critically involved in pain modulation have been elucidated. These research achievements have incited progress in identifying new diagnostic and therapeutic targets. In this review, we first introduce fundamental knowledge about pain, setting the stage for the subsequent contents. The review next delves into the molecular mechanisms underlying pain disorders, including gene mutation, epigenetic modification, posttranslational modification, inflammasome, signaling pathways and microbiota. To better present a comprehensive view of pain research, two prominent issues, sexual dimorphism and pain comorbidities, are discussed in detail based on current findings. The status quo of pain evaluation and manipulation is summarized. A series of improved and innovative pain management strategies, such as gene therapy, monoclonal antibody, brain-computer interface and microbial intervention, are making strides towards clinical application. We highlight existing limitations and future directions for enhancing the quality of preclinical and clinical research. Efforts to decipher the complexities of pain pathology will be instrumental in translating scientific discoveries into clinical practice, thereby improving pain management from bench to bedside.
Collapse
Affiliation(s)
- Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qixuan Xu
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Yajiao Shi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China
| | - Ruiyang Zhao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Hanghang Li
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China.
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China.
| | - Bo Wei
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
11
|
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
|
12
|
Alsaadi H, Peller J, Ghasemlou N, Kawaja MD. Immunohistochemical phenotype of sensory neurons associated with sympathetic plexuses in the trigeminal ganglia of adult nerve growth factor transgenic mice. J Comp Neurol 2024; 532:e25563. [PMID: 37986234 DOI: 10.1002/cne.25563] [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: 11/22/2023]
Abstract
Following peripheral nerve injury, postganglionic sympathetic axons sprout into the affected sensory ganglia and form perineuronal sympathetic plexuses with somata of sensory neurons. This sympathosensory coupling contributes to the onset and persistence of injury-induced chronic pain. We have documented the presence of similar sympathetic plexuses in the trigeminal ganglia of adult mice that ectopically overexpress nerve growth factor (NGF), in the absence of nerve injury. In this study, we sought to further define the phenotype(s) of these trigeminal sensory neurons having sympathetic plexuses in our transgenic mice. Using quantitative immunofluorescence staining analyses, we show that the invading sympathetic axons specifically target sensory somata immunopositive for several biomarkers: NGF high-affinity receptor tyrosine kinase A (trkA), calcitonin gene-related peptide (CGRP), neurofilament heavy chain (NFH), and P2X purinoceptor 3 (P2X3). Based on these phenotypic characteristics, the majority of the sensory somata surrounded by sympathetic plexuses are likely to be NGF-responsive nociceptors (i.e., trkA expressing) that are peptidergic (i.e., CGRP expressing), myelinated (i.e., NFH expressing), and ATP sensitive (i.e., P2X3 expressing). Our data also show that very few sympathetic plexuses surround sensory somata expressing other nociceptive (pain) biomarkers, including substance P and acid-sensing ion channel 3. No sympathetic plexuses are associated with sensory somata that display isolectin B4 binding. Though the cellular mechanisms that trigger the formation of sympathetic plexus (with and without nerve injury) remain unknown, our new observations yield an unexpected specificity with which invading sympathetic axons appear to target a precise subtype of nociceptors. This selectivity likely contributes to pain development and maintenance associated with sympathosensory coupling.
Collapse
Affiliation(s)
- Hanin Alsaadi
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Jacob Peller
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Michael D Kawaja
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
13
|
Makau CM, Towett PK, Kanui TI, Abelson KSP. Effects of inhibition of Nav1.3, Nav1.7, and Nav1.8 channels on pain-related behavior in Speke's hinge-back tortoise (Kinixys spekii). J Neurosci Res 2024; 102:e25274. [PMID: 38284848 DOI: 10.1002/jnr.25274] [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: 06/01/2023] [Revised: 10/10/2023] [Accepted: 10/25/2023] [Indexed: 01/30/2024]
Abstract
Comparative studies using reptiles as experimental animals in pain research could expand our knowledge on the evolution and adaptation of pain mechanisms. Currently, there are no data reported on the involvement of voltage-gated sodium ion channels on nociception in reptiles. The aim of this study was to investigate the involvement of Nav1.3, Nav1.7, and Nav1.8 ion channels in nociception in Speke's hinge-back tortoise. ICA 121341 (selective blocker for Nav1.1/Nav1.3), NAV 26 (selective blocker for Nav1.7), and A803467 (selective blocker for Nav1.8) were used to investigate the involvement of Nav1.3, Nav1.7, and Nav1.8, respectively. The chemicals were administered intracoelomically thirty minutes before the start of nociceptive tests. ICA 121341 did not cause a significant decrease in the time spent in pain-related behavior in all the nociceptive tests. NAV 26 and A8034667 caused a statistically significant decrease in the mean time spent in pain-related behavior in the formalin and capsaicin tests. Only A803467 caused a statistically significant increase in the mean latency to pain-related behavior in the hot plate test. NAV 26 and A803467 had no observable side effects. In conclusion, Nav1.7 and Nav1.8 are involved in the processing of chemically induced inflammatory pain in Speke's hinge back tortoise. In addition, Nav1.8 are also significantly involved in the development of thermal-induced pain-related behavior in this species of reptile. However, our results do not support the involvement of Nav1.3 on the development of chemical or thermal induced pain-related behavior in the Speke's hinge back tortoise.
Collapse
Affiliation(s)
- Christopher M Makau
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Philemon K Towett
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Titus I Kanui
- School of Agriculture and Veterinary Sciences, South Eastern Kenya University, Kitui, Kenya
| | - Klas S P Abelson
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Haroun R, Gossage SJ, Luiz AP, Arcangeletti M, Sikandar S, Zhao J, Cox JJ, Wood JN. Chemogenetic Silencing of Na V1.8-Positive Sensory Neurons Reverses Chronic Neuropathic and Bone Cancer Pain in FLEx PSAM 4-GlyR Mice. eNeuro 2023; 10:ENEURO.0151-23.2023. [PMID: 37679042 PMCID: PMC10523839 DOI: 10.1523/eneuro.0151-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Drive from peripheral neurons is essential in almost all pain states, but pharmacological silencing of these neurons to effect analgesia has proved problematic. Reversible gene therapy using long-lived chemogenetic approaches is an appealing option. We used the genetically activated chloride channel PSAM4-GlyR to examine pain pathways in mice. Using recombinant AAV9-based delivery to sensory neurons, we found a reversal of acute pain behavior and diminished neuronal activity using in vitro and in vivo GCaMP imaging on activation of PSAM4-GlyR with varenicline. A significant reduction in inflammatory heat hyperalgesia and oxaliplatin-induced cold allodynia was also observed. Importantly, there was no impairment of motor coordination, but innocuous von Frey sensation was inhibited. We generated a transgenic mouse that expresses a CAG-driven FLExed PSAM4-GlyR downstream of the Rosa26 locus that requires Cre recombinase to enable the expression of PSAM4-GlyR and tdTomato. We used NaV1.8 Cre to examine the role of predominantly nociceptive NaV1.8+ neurons in cancer-induced bone pain (CIBP) and neuropathic pain caused by chronic constriction injury (CCI). Varenicline activation of PSAM4-GlyR in NaV1.8-positive neurons reversed CCI-driven mechanical, thermal, and cold sensitivity. Additionally, varenicline treatment of mice with CIBP expressing PSAM4-GlyR in NaV1.8+ sensory neurons reversed cancer pain as assessed by weight-bearing. Moreover, when these mice were subjected to acute pain assays, an elevation in withdrawal thresholds to noxious mechanical and thermal stimuli was detected, but innocuous mechanical sensations remained unaffected. These studies confirm the utility of PSAM4-GlyR chemogenetic silencing in chronic pain states for mechanistic analysis and potential future therapeutic use.
Collapse
Affiliation(s)
- Rayan Haroun
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - Samuel J Gossage
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - Ana Paula Luiz
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - Manuel Arcangeletti
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - Shafaq Sikandar
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Jing Zhao
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - James J Cox
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| | - John N Wood
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
15
|
Brackx W, de Cássia Collaço R, Theys M, Cruyssen JV, Bosmans F. Understanding the physiological role of Na V1.9: Challenges and opportunities for pain modulation. Pharmacol Ther 2023; 245:108416. [PMID: 37061202 DOI: 10.1016/j.pharmthera.2023.108416] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
Voltage-activated Na+ (NaV) channels are crucial contributors to rapid electrical signaling in the human body. As such, they are among the most targeted membrane proteins by clinical therapeutics and natural toxins. Several of the nine mammalian NaV channel subtypes play a documented role in pain or other sensory processes such as itch, touch, and smell. While causal relationships between these subtypes and biological function have been extensively described, the physiological role of NaV1.9 is less understood. Yet, mutations in NaV1.9 can cause striking disease phenotypes related to sensory perception such as loss or gain of pain and chronic itch. Here, we explore our current knowledge of the mechanisms by which NaV1.9 may contribute to pain and elaborate on the challenges associated with establishing links between experimental conditions and human disease. This review also discusses the lack of comprehensive insights into NaV1.9-specific pharmacology, an unfortunate situation since modulatory compounds may have tremendous potential in the clinic to treat pain or as precision tools to examine the extent of NaV1.9 participation in sensory perception processes.
Collapse
Affiliation(s)
- Wayra Brackx
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Rita de Cássia Collaço
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Margaux Theys
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Jolien Vander Cruyssen
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Frank Bosmans
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium.
| |
Collapse
|
16
|
Tellegen AR, Rudnik-Jansen I, Utomo L, Versteeg S, Beukers M, Maarschalkerweerd R, van Zuilen D, van Klaveren NJ, Houben K, Teske E, van Weeren PR, Karssemakers-Degen N, Mihov G, Thies J, Eijkelkamp N, Creemers LB, Meij BP, Tryfonidou MA. Sustained release of locally delivered celecoxib provides pain relief for osteoarthritis: a proof of concept in dog patients. Osteoarthritis Cartilage 2023; 31:351-362. [PMID: 36473675 DOI: 10.1016/j.joca.2022.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Drug delivery platforms that allow for gradual drug release after intra-articular administration have become of much interest as a treatment strategy for osteoarthritis (OA). The aim of this study was to investigate the safety and efficacy of an intra-articular sustained release formulation containing celecoxib (CXB), a cyclooxygenase-2 (COX-2) selective inhibitor. METHODS Amino acid-based polyesteramide microspheres (PEAMs), a biodegradable and non-toxic platform, were loaded with CXB and employed in two in vivo models of arthritis: an acute inflammatory arthritis model in rats (n = 12), and a randomized controlled study in chronic OA dog patients (n = 30). In parallel, the bioactivity of sustained release of CXB was evaluated in monolayer cultures of primary dog chondrocytes under inflammatory conditions. RESULTS Sustained release of CXB did not alleviate acute arthritis signs in the rat arthritis model, based on pain measurements and synovitis severity. However, in OA dog patients, sustained release of CXB improved limb function as objective parameter of pain and quality of life based on gait analysis and owner questionnaires. It also decreased pain medication dependency over a 2-month period and caused no adverse effects. Prostaglandin E2 levels, a marker for inflammation, were lower in the synovial fluid of CXB-treated dog OA patients and in CXB-treated cultured dog chondrocytes. CONCLUSION These results show that local sustained release of CXB is less suitable to treat acute inflammation in arthritic joints, while safe and effective in treating pain in chronic OA in dogs.
Collapse
Affiliation(s)
- A R Tellegen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - I Rudnik-Jansen
- Department of Orthopedics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - L Utomo
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - S Versteeg
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - M Beukers
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - R Maarschalkerweerd
- Department of Orthopedics, Medisch Centrum voor Dieren, Amsterdam, the Netherlands
| | - D van Zuilen
- Department of Orthopedics, Medisch Centrum voor Dieren, Amsterdam, the Netherlands
| | - N J van Klaveren
- Department of Orthopedics, Medisch Centrum voor Dieren, Amsterdam, the Netherlands
| | - K Houben
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - E Teske
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - P R van Weeren
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | | - G Mihov
- DSM Biomedical, Geleen, the Netherlands
| | - J Thies
- DSM Biomedical, Geleen, the Netherlands
| | - N Eijkelkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - L B Creemers
- Department of Orthopedics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - B P Meij
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - M A Tryfonidou
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
17
|
Haroun R, Wood JN, Sikandar S. Mechanisms of cancer pain. FRONTIERS IN PAIN RESEARCH 2023; 3:1030899. [PMID: 36688083 PMCID: PMC9845956 DOI: 10.3389/fpain.2022.1030899] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/14/2022] [Indexed: 01/05/2023] Open
Abstract
Personalised and targeted interventions have revolutionised cancer treatment and dramatically improved survival rates in recent decades. Nonetheless, effective pain management remains a problem for patients diagnosed with cancer, who continue to suffer from the painful side effects of cancer itself, as well as treatments for the disease. This problem of cancer pain will continue to grow with an ageing population and the rapid advent of more effective therapeutics to treat the disease. Current pain management guidelines from the World Health Organisation are generalised for different pain severities, but fail to address the heterogeneity of mechanisms in patients with varying cancer types, stages of disease and treatment plans. Pain is the most common complaint leading to emergency unit visits by patients with cancer and over one-third of patients that have been diagnosed with cancer will experience under-treated pain. This review summarises preclinical models of cancer pain states, with a particular focus on cancer-induced bone pain and chemotherapy-associated pain. We provide an overview of how preclinical models can recapitulate aspects of pain and sensory dysfunction that is observed in patients with persistent cancer-induced bone pain or neuropathic pain following chemotherapy. Peripheral and central nervous system mechanisms of cancer pain are discussed, along with key cellular and molecular mediators that have been highlighted in animal models of cancer pain. These include interactions between neuronal cells, cancer cells and non-neuronal cells in the tumour microenvironment. Therapeutic targets beyond opioid-based management are reviewed for the treatment of cancer pain.
Collapse
Affiliation(s)
- Rayan Haroun
- Division of Medicine, Wolfson Institute of Biomedical Research, University College London, London, United Kingdom
| | - John N Wood
- Division of Medicine, Wolfson Institute of Biomedical Research, University College London, London, United Kingdom
| | - Shafaq Sikandar
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
18
|
Na V1.7 Channel Blocker [Ala 5, Phe 6, Leu 26, Arg 28]GpTx-1 Attenuates CFA-induced Inflammatory Hypersensitivity in Rats via Endogenous Enkephalin Mechanism. THE JOURNAL OF PAIN 2022; 24:840-859. [PMID: 36586660 DOI: 10.1016/j.jpain.2022.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Venom-derived NaV1.7 channel blockers have promising prospects in pain management. The 34-residue tarantula peptide GpTx-1 is a potent NaV1.7 channel blocker. Its powerful analog [Ala5, Phe6, Leu26, Arg28]GpTx-1 (GpTx-1-71) displayed excellent NaV1.7 selectivity and analgesic properties in mice. The current study aimed to elucidate the anti-hyperalgesic activities of GpTx-1-71 in inflammatory pain and reveal the underlying mechanisms. Our results demonstrated that intrathecal and intraplantar injections of GpTx-1-71 dose-dependently attenuated CFA-induced inflammatory hypersensitivity in rats. Moreover, GpTx-1-71-induced anti-hyperalgesia was significantly reduced by opioid receptor antagonists and the enkephalin antibody and diminished in proenkephalin (Penk) gene knockout animals. Consistently, GpTx-1-71 treatment increased the enkephalin level in the spinal dorsal horn and promoted the Penk transcription and enkephalin release in primary dorsal root ganglion (DRG) neurons, wherein sodium played a crucial role in these processes. Mass spectrometry analysis revealed that GpTx-1-71 mainly promoted the secretion of Met-enkephalin but not Leu-enkephalin from DRG neurons. In addition, the combination of subtherapeutic Met-enkephalin and GpTx-1-71 produced synergistic anti-hyperalgesia in CFA-induced inflammatory hypersensitivity. These findings suggest that the endogenous enkephalin pathway is essential for GpTx-1-71-induced spinal and peripheral analgesia in inflammatory pain. PERSPECTIVE: This article presents a possible pharmacological mechanism underlying NaV1.7 blocker-induced analgesia in inflammatory pain, which helps us to better understand and develop venom-based painkillers for incurable pain.
Collapse
|
19
|
Reid AR, Côté PD, McDougall JJ. Long-Term Blockade of Nociceptive Na v1.7 Channels Is Analgesic in Rat Models of Knee Arthritis. Biomolecules 2022; 12:1571. [PMID: 36358921 PMCID: PMC9687684 DOI: 10.3390/biom12111571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
The voltage gated sodium channels (Nav) 1.7, 1.8, and 1.9 are primarily located on nociceptors where they are involved in signalling neuropathic pain. This study examined the effect of Nav1.7 blockade on joint pain using either the small molecule inhibitor PF05089771 or an antibody directed towards the intracellular domain of the ion channel. Male Wistar rats were assigned to one of three experimental groups consisting of either intra-articular injection of 3 mg sodium monoiodoacetate (MIA-joint degeneration group), intra-articular injection of 100 μg lysophosphatidic acid (LPA-joint neuropathy group), or transection of the medial meniscus (MMT-posttraumatic osteoarthritis group). G-ratio calculations were performed to determine potential demyelination and immunohistochemistry was used to measure Nav1.7 expression on joint afferent cell bodies. Pain behaviour was evaluated over 3 h by von Frey hair algesiometry and hindlimb weight bearing before and after local administration of PF05089771 (0.1 mg/50 µL). Chronic pain behaviour was assessed over 28 days following peripheral treatment with a Nav1.7 antibody (Ab) in conjunction with the transmembrane carrier peptide Pep1. Demyelination and increased Nav1.7 channel expression were observed in MIA and LPA rats, but not with MMT. Acute secondary allodynia was diminished by PF05089771 while a single injection of Nav1.7 Ab-Pep1 reduced pain up to 28 days. This analgesia only occurred in MIA and LPA animals. Hindlimb incapacitance was not affected by any treatment. These data indicate that joint pain associated with neural demyelination can be alleviated somewhat by Nav1.7 channel blockade. Biologics that inactivate Nav1.7 channels have the potential to reduce arthritis pain over a protracted period of time.
Collapse
Affiliation(s)
- Allison R. Reid
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada
| | - Patrice D. Côté
- Department of Biology, Dalhousie University, 1355 Oxford, Halifax, NS B3H 4R2, Canada
| | - Jason J. McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
20
|
Liu XG. Normalization of Neuroinflammation: A New Strategy for Treatment of Persistent Pain and Memory/Emotional Deficits in Chronic Pain. J Inflamm Res 2022; 15:5201-5233. [PMID: 36110505 PMCID: PMC9469940 DOI: 10.2147/jir.s379093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic pain, which affects around 1/3 of the world population and is often comorbid with memory deficit and mood depression, is a leading source of suffering and disability. Studies in past decades have shown that hyperexcitability of primary sensory neurons resulting from abnormal expression of ion channels and central sensitization mediated pathological synaptic plasticity, such as long-term potentiation in spinal dorsal horn, underlie the persistent pain. The memory/emotional deficits are associated with impaired synaptic connectivity in hippocampus. Dysregulation of numerous endogenous proteins including receptors and intracellular signaling molecules is involved in the pathological processes. However, increasing knowledge contributes little to clinical treatment. Emerging evidence has demonstrated that the neuroinflammation, characterized by overproduction of pro-inflammatory cytokines and glial activation, is reliably detected in humans and animals with chronic pain, and is sufficient to induce persistent pain and memory/emotional deficits. The abnormal expression of ion channels and pathological synaptic plasticity in spinal dorsal horn and in hippocampus are resulting from neuroinflammation. The neuroinflammation is initiated and maintained by the interactions of circulating monocytes, glial cells and neurons. Obviously, unlike infectious diseases and cancer, which are caused by pathogens or malignant cells, chronic pain is resulting from alterations of cells and molecules which have numerous physiological functions. Therefore, normalization (counterbalance) but not simple inhibition of the neuroinflammation is the right strategy for treating neuronal disorders. Currently, no such agent is available in clinic. While experimental studies have demonstrated that intracellular Mg2+ deficiency is a common feature of chronic pain in animal models and supplement Mg2+ are capable of normalizing the neuroinflammation, activation of upregulated proteins that promote recovery, such as translocator protein (18k Da) or liver X receptors, has a similar effect. In this article, relevant experimental and clinical evidence is reviewed and discussed.
Collapse
Affiliation(s)
- Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
21
|
Kitano Y, Shinozuka T. Inhibition of Na V1.7: the possibility of ideal analgesics. RSC Med Chem 2022; 13:895-920. [PMID: 36092147 PMCID: PMC9384491 DOI: 10.1039/d2md00081d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 08/03/2023] Open
Abstract
The selective inhibition of NaV1.7 is a promising strategy for developing novel analgesic agents with fewer adverse effects. Although the potent selective inhibition of NaV1.7 has been recently achieved, multiple NaV1.7 inhibitors failed in clinical development. In this review, the relationship between preclinical in vivo efficacy and NaV1.7 coverage among three types of voltage-gated sodium channel (VGSC) inhibitors, namely conventional VGSC inhibitors, sulphonamides and acyl sulphonamides, is discussed. By demonstrating the PK/PD discrepancy of preclinical studies versus in vivo models and clinical results, the potential reasons behind the disconnect between preclinical results and clinical outcomes are discussed together with strategies for developing ideal analgesic agents.
Collapse
Affiliation(s)
- Yutaka Kitano
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| | - Tsuyoshi Shinozuka
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| |
Collapse
|
22
|
Neuroimmune Mechanisms Underlying Neuropathic Pain: The Potential Role of TNF-α-Necroptosis Pathway. Int J Mol Sci 2022; 23:ijms23137191. [PMID: 35806192 PMCID: PMC9266916 DOI: 10.3390/ijms23137191] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
The neuroimmune mechanism underlying neuropathic pain has been extensively studied. Tumor necrosis factor-alpha (TNF-α), a key pro-inflammatory cytokine that drives cytokine storm and stimulates a cascade of other cytokines in pain-related pathways, induces and modulates neuropathic pain by facilitating peripheral (primary afferents) and central (spinal cord) sensitization. Functionally, TNF-α controls the balance between cell survival and death by inducing an inflammatory response and two programmed cell death mechanisms (apoptosis and necroptosis). Necroptosis, a novel form of programmed cell death, is receiving increasing attraction and may trigger neuroinflammation to promote neuropathic pain. Chronic pain is often accompanied by adverse pain-associated emotional reactions and cognitive disorders. Overproduction of TNF-α in supraspinal structures such as the anterior cingulate cortex (ACC) and hippocampus plays an important role in pain-associated emotional disorders and memory deficits and also participates in the modulation of pain transduction. At present, studies reporting on the role of the TNF-α–necroptosis pathway in pain-related disorders are lacking. This review indicates the important research prospects of this pathway in pain modulation based on its role in anxiety, depression and memory deficits associated with other neurodegenerative diseases. In addition, we have summarized studies related to the underlying mechanisms of neuropathic pain mediated by TNF-α and discussed the role of the TNF-α–necroptosis pathway in detail, which may represent an avenue for future therapeutic intervention.
Collapse
|
23
|
Lischka A, Lassuthova P, Çakar A, Record CJ, Van Lent J, Baets J, Dohrn MF, Senderek J, Lampert A, Bennett DL, Wood JN, Timmerman V, Hornemann T, Auer-Grumbach M, Parman Y, Hübner CA, Elbracht M, Eggermann K, Geoffrey Woods C, Cox JJ, Reilly MM, Kurth I. Genetic pain loss disorders. Nat Rev Dis Primers 2022; 8:41. [PMID: 35710757 DOI: 10.1038/s41572-022-00365-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2022] [Indexed: 01/05/2023]
Abstract
Genetic pain loss includes congenital insensitivity to pain (CIP), hereditary sensory neuropathies and, if autonomic nerves are involved, hereditary sensory and autonomic neuropathy (HSAN). This heterogeneous group of disorders highlights the essential role of nociception in protecting against tissue damage. Patients with genetic pain loss have recurrent injuries, burns and poorly healing wounds as disease hallmarks. CIP and HSAN are caused by pathogenic genetic variants in >20 genes that lead to developmental defects, neurodegeneration or altered neuronal excitability of peripheral damage-sensing neurons. These genetic variants lead to hyperactivity of sodium channels, disturbed haem metabolism, altered clathrin-mediated transport and impaired gene regulatory mechanisms affecting epigenetic marks, long non-coding RNAs and repetitive elements. Therapies for pain loss disorders are mainly symptomatic but the first targeted therapies are being tested. Conversely, chronic pain remains one of the greatest unresolved medical challenges, and the genes and mechanisms associated with pain loss offer new targets for analgesics. Given the progress that has been made, the coming years are promising both in terms of targeted treatments for pain loss disorders and the development of innovative pain medicines based on knowledge of these genetic diseases.
Collapse
Affiliation(s)
- Annette Lischka
- Institute of Human Genetics, Medical Faculty, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Petra Lassuthova
- Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - Arman Çakar
- Neuromuscular Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Christopher J Record
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Jonas Van Lent
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Antwerp, Belgium
| | - Jonathan Baets
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Antwerp, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Maike F Dohrn
- Department of Neurology, Medical Faculty, Uniklinik RWTH Aachen University, Aachen, Germany
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Jan Senderek
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | - Angelika Lampert
- Institute of Physiology, Medical Faculty, Uniklinik RWTH Aachen University, Aachen, Germany
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, Oxford University, Oxford, UK
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Antwerp, Belgium
| | - Thorsten Hornemann
- Department of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michaela Auer-Grumbach
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Yesim Parman
- Neuromuscular Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | | | - Miriam Elbracht
- Institute of Human Genetics, Medical Faculty, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Katja Eggermann
- Institute of Human Genetics, Medical Faculty, Uniklinik RWTH Aachen University, Aachen, Germany
| | - C Geoffrey Woods
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Mary M Reilly
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty, Uniklinik RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
24
|
Xue Y, Kremer M, Muniz Moreno MDM, Chidiac C, Lorentz R, Birling MC, Barrot M, Herault Y, Gaveriaux-Ruff C. The Human SCN9AR185H Point Mutation Induces Pain Hypersensitivity and Spontaneous Pain in Mice. Front Mol Neurosci 2022; 15:913990. [PMID: 35769334 PMCID: PMC9234669 DOI: 10.3389/fnmol.2022.913990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
The voltage-gated sodium channel Nav1.7 is encoded by SCN9A gene and plays a critical role in pain sensitivity. Several SCN9A gain-of-function (GOF) mutations have been found in patients with small fiber neuropathy (SFN) having chronic pain, including the R185H mutation. However, for most of these variants, their involvement in pain phenotype still needs to be experimentally elucidated. In order to delineate the impact of R185H mutation on pain sensitivity, we have established the Scn9aR185H mutant mouse model using the CRISPR/Cas9 technology. The Scn9aR185H mutant mice show no cellular alteration in the dorsal root ganglia (DRG) containing cell bodies of sensory neurons and no alteration of growth or global health state. Heterozygous and homozygous animals of both sexes were investigated for pain sensitivity. The mutant mice were more sensitive than the wild-type mice in the tail flick and hot plate tests, acetone, and von Frey tests for sensitivity to heat, cold, and touch, respectively, although with sexual dimorphic effects. The newly developed bioinformatic pipeline, Gdaphen is based on general linear model (GLM) and random forest (RF) classifiers as well as a multifactor analysis of mixed data and shows the qualitative and quantitative variables contributing the most to the pain phenotype. Using Gdaphen, tail flick, Hargreaves, hot plate, acetone, cold plate, and von Frey tests, sex and genotype were found to be contributing most to the pain phenotype. Importantly, the mutant animals displayed spontaneous pain as assessed in the conditioned place preference (CPP) assay. Altogether, our results indicate that Scn9aR185H mice show a pain phenotype, suggesting that the SCN9AR185H mutation identified in patients with SFN having chronic pain contributes to their symptoms. Therefore, we provide genetic evidence for the fact that this mutation in Nav1.7 channel plays an important role in nociception and in the pain experienced by patients with SFN who have this mutation. These findings should aid in exploring further pain treatments based on the Nav1.7 channel.
Collapse
Affiliation(s)
- Yaping Xue
- Centre National de la Recherche Scientifique (CNRS), Institut de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Mélanie Kremer
- Centre National de la Recherche Scientifique (CNRS), Institut des Neurosciences Cellulaires et Intégratives (INCI), Université de Strasbourg, Strasbourg, France
| | - Maria del Mar Muniz Moreno
- Centre National de la Recherche Scientifique (CNRS), Institut de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Celeste Chidiac
- Centre National de la Recherche Scientifique (CNRS), Institut de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Romain Lorentz
- Centre National de la Recherche Scientifique (CNRS), Institut de la Santé et de la Recherche Médicale (INSERM), CELPHEDIA-PHENOMIN-Institut Clinique de la Souris, (PHENOMIN-ICS), Université de Strasbourg, Illkirch, France
| | - Marie-Christine Birling
- Centre National de la Recherche Scientifique (CNRS), Institut de la Santé et de la Recherche Médicale (INSERM), CELPHEDIA-PHENOMIN-Institut Clinique de la Souris, (PHENOMIN-ICS), Université de Strasbourg, Illkirch, France
| | - Michel Barrot
- Centre National de la Recherche Scientifique (CNRS), Institut des Neurosciences Cellulaires et Intégratives (INCI), Université de Strasbourg, Strasbourg, France
| | - Yann Herault
- Centre National de la Recherche Scientifique (CNRS), Institut de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), Institut de la Santé et de la Recherche Médicale (INSERM), CELPHEDIA-PHENOMIN-Institut Clinique de la Souris, (PHENOMIN-ICS), Université de Strasbourg, Illkirch, France
- *Correspondence: Yann Herault,
| | - Claire Gaveriaux-Ruff
- Centre National de la Recherche Scientifique (CNRS), Institut de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS) UMR 7242, Université de Strasbourg, Illkirch, France
- Claire Gaveriaux-Ruff,
| |
Collapse
|
25
|
Li S, Ding M, Wu Y, Xue S, Ji Y, Zhang P, Zhang Z, Cao Z, Zhang F. Histamine Sensitization of the Voltage-Gated Sodium Channel Nav1.7 Contributes to Histaminergic Itch in Mice. ACS Chem Neurosci 2022; 13:700-710. [PMID: 35157443 DOI: 10.1021/acschemneuro.2c00012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Itch, a common clinical symptom of many skin diseases, severely impairs the life quality of patients. Nav1.7, a subtype of voltage-gated sodium channels mainly expressed in primary sensory neurons, is responsible for the amplification of threshold currents that trigger action potential (AP) generation. Gain-of-function mutation of Nav1.7 leads to paroxysmal itch, while pharmacological inhibition of Nav1.7 alleviates histamine-dependent itch. However, the crosstalk between histamine and Nav1.7 that leads to itch is unclear. In the present study, we demonstrated that in the dorsal root ganglion (DRG) neurons from histamine-dependent itch model mice induced by compound 48/80, tetrodotoxin-sensitive (TTX-S) but not TTX-resistant Na+ currents were activated at more hyperpolarized membrane potentials compared to those on DRG neurons from vehicle-treated mice. Meanwhile, bath application of histamine shifted the activation voltages of TTX-S Na+ currents to the hyperpolarized direction, increased the AP frequency, and reduced the current threshold required to elicit APs. Further mechanistic studies demonstrated that selective activation of H1 but not H2 and H4 receptors mimicked histamine effect on TTX-S Na+ channels in DRG neurons. The protein kinase C (PKC) inhibitor GO 8963, but not the PKA inhibitor H89, normalized histamine-sensitized TTX-S Na+ channels. We also demonstrated that histamine shifted the activation voltages of Na+ currents to the hyperpolarized direction in Chinese hamster ovary (CHO) cells expressing Nav1.7. Importantly, selective inhibition of Nav1.7 by PF-05089771 significantly relieved the scratching frequency in a histamine-dependent itch model induced by compound 48/80. Taken together, these data suggest that activation of H1 receptors by histamine sensitizes Nav1.7 channels through the PKC pathway in DRG neurons that contributes to histamine-dependent itch.
Collapse
Affiliation(s)
- Shaoheng Li
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Meihuizi Ding
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wu
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Shuwen Xue
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunyun Ji
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Pinhui Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhuang Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Fan Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| |
Collapse
|
26
|
OUP accepted manuscript. Brain 2022; 145:1632-1640. [DOI: 10.1093/brain/awac078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 01/18/2022] [Accepted: 02/12/2022] [Indexed: 11/14/2022] Open
|
27
|
Diochot S. Pain-related toxins in scorpion and spider venoms: a face to face with ion channels. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20210026. [PMID: 34925480 PMCID: PMC8667759 DOI: 10.1590/1678-9199-jvatitd-2021-0026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Pain is a common symptom induced during envenomation by spiders and scorpions.
Toxins isolated from their venom have become essential tools for studying the
functioning and physiopathological role of ion channels, as they modulate their
activity. In particular, toxins that induce pain relief effects can serve as a
molecular basis for the development of future analgesics in humans. This review
provides a summary of the different scorpion and spider toxins that directly
interact with pain-related ion channels, with inhibitory or stimulatory effects.
Some of these toxins were shown to affect pain modalities in different animal
models providing information on the role played by these channels in the pain
process. The close interaction of certain gating-modifier toxins with membrane
phospholipids close to ion channels is examined along with molecular approaches
to improve selectivity, affinity or bioavailability in vivo for
therapeutic purposes.
Collapse
Affiliation(s)
- Sylvie Diochot
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS) UMR 7275 et Université Côte d'Azur (UCA), 06560 Valbonne, France. Institut de Pharmacologie Moléculaire et Cellulaire Centre National de la Recherche Scientifique Université Côte d'Azur Valbonne France
| |
Collapse
|
28
|
Chidiac C, Xue Y, Muniz Moreno MDM, Bakr Rasheed AA, Lorentz R, Birling MC, Gaveriaux-Ruff C, Herault Y. The Human SCN10A G1662S Point Mutation Established in Mice Impacts on Mechanical, Heat, and Cool Sensitivity. Front Pharmacol 2021; 12:780132. [PMID: 34925037 PMCID: PMC8671994 DOI: 10.3389/fphar.2021.780132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
The voltage-gated sodium channel NAV1.8 is expressed in primary nociceptive neurons and is involved in pain transmission. Mutations in the SCN10A gene (encoding NAV1.8 channel) have been identified in patients with idiopathic painful small fiber neuropathy (SFN) including the SCN10AG1662S gain-of-function mutation. However, the role of this mutation in pain sensation remains unknown. We have generated the first mouse model for the G1662S mutation by using homologous recombination in embryonic stem cells. The corresponding Scn10aG1663S mouse line has been analyzed for Scn10a expression, intraepidermal nerve fiber density (IENFD), and nociception using behavioral tests for thermal and mechanical sensitivity. The Scn10aG1663S mutants had a similar Scn10a expression level in dorsal root ganglia (DRG) to their wild-type littermates and showed normal IENFD in hindpaw skin. Mutant mice were more sensitive to touch than wild types in the von Frey test. In addition, sexual dimorphism was observed for several pain tests, pointing to the relevance of performing the phenotypical assessment in both sexes. Female homozygous mutants tended to be more sensitive to cooling stimuli in the acetone test. For heat sensitivity, male homozygous mutants showed shorter latencies to radiant heat in the Hargreaves test while homozygous females had longer latencies in the tail flick test. In addition, mutant males displayed a shorter reaction latency on the 54°C hot plate. Collectively, Scn10aG1663S mutant mice show a moderate but consistent increased sensitivity in behavioral tests of nociception. This altered nociception found in Scn10aG1663S mice demonstrates that the corresponding G1662 mutation of SCN10A found in SFN patients with pain contributes to their pain symptoms.
Collapse
Affiliation(s)
- Celeste Chidiac
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Yaping Xue
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Maria Del Mar Muniz Moreno
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Ameer Abu Bakr Rasheed
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Romain Lorentz
- CNRS, INSERM, PHENOMIN-Institut Clinique de la Souris, Université de Strasbourg, Illkirch, France
| | - Marie-Christine Birling
- CNRS, INSERM, PHENOMIN-Institut Clinique de la Souris, Université de Strasbourg, Illkirch, France
| | - Claire Gaveriaux-Ruff
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Yann Herault
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France.,CNRS, INSERM, PHENOMIN-Institut Clinique de la Souris, Université de Strasbourg, Illkirch, France
| |
Collapse
|
29
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
30
|
Abdalla HB, Napimoga MH, Trindade-da-Silva CA, Guimarães M, Lopes M, Dos Santos PCV, Buarque E Silva WA, Andrade E Silva F, Clemente-Napimoga JT. Occlusal Trauma Induces Neuroimmune Crosstalk for a Pain State. J Dent Res 2021; 101:339-347. [PMID: 34596449 DOI: 10.1177/00220345211039482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Temporomandibular joint (TMJ) disorder caused by occlusal trauma is one of the most controversial topics in dentistry. Experimental traumatic occlusion (ETO) induced by metal crowns cemented to mandibular first molars in rats causes a long-lasting nociceptive response. This study aimed to elucidate whether ETO generates an increase in inflammatory mediators in the TMJ. In addition, the impact of ETO on trigeminal ganglia, neurotransmitter release, and satellite glial cell (SGC) activation was investigated. ELISA revealed enhanced inflammatory mediators, including TNF-α, IL-1β, IL-6, CX3CL1, and ADAM-17 by Western blotting, in periarticular TMJ tissue after 28 d of ETO. In the trigeminal ganglia, ETO groups increased the release of the neurotransmitters substance P and glutamate. Overexpression of the AMPA receptor and upregulation of NMDA were observed in the 0.4- and 0.7-mm ETO groups, respectively, highlighting enhanced neuronal excitation. Increased IL-1β and COX-2 mRNA levels in the 0.7-mm ETO group confirmed trigeminal ganglia SGC activation. Immunofluorescence and electrophoresis of SGC revealed increased pERK expression in the 0.7-mm ETO group. ERK phosphorylation was shown to be nociceptive specific, with its upregulation occurring in cases of chronic inflammatory pain. Increased PKA mRNA levels were observed in the 0.4-mm ETO group, while CREB mRNA levels were upregulated for both ETO groups. Electrophoresis showed overexpression of sodium channel Nav 1.7 in the 0.7-mm ETO group, while immunofluorescence revealed that Nav 1.7 is expressed in sensory trigeminal ganglia cells. The results of this study suggest that occlusal trauma induces neuroimmune crosstalk, with synthesis of proinflammatory/pronociceptive mediators, which increases neuronal activity in trigeminal ganglia via the activation of an inflammatory response cascade to develop a persistent neuroinflammatory state that leads to central sensitization.
Collapse
Affiliation(s)
- H B Abdalla
- Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Laboratoy of Neuroimmune Interface of Pain Research, Campinas, SP, Brazil
| | - M H Napimoga
- Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Laboratoy of Neuroimmune Interface of Pain Research, Campinas, SP, Brazil
| | - C A Trindade-da-Silva
- Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Laboratoy of Neuroimmune Interface of Pain Research, Campinas, SP, Brazil
| | - M Guimarães
- Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Laboratoy of Neuroimmune Interface of Pain Research, Campinas, SP, Brazil
| | - M Lopes
- Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Laboratoy of Neuroimmune Interface of Pain Research, Campinas, SP, Brazil
| | - P C V Dos Santos
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas-UNICAMP, Piracicaba, São Paulo, Brazil
| | - W A Buarque E Silva
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas-UNICAMP, Piracicaba, São Paulo, Brazil
| | - F Andrade E Silva
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas-UNICAMP, Piracicaba, São Paulo, Brazil
| | - J T Clemente-Napimoga
- Faculdade São Leopoldo Mandic, Instituto e Centro de Pesquisas São Leopoldo Mandic, Laboratoy of Neuroimmune Interface of Pain Research, Campinas, SP, Brazil
| |
Collapse
|
31
|
Wang GJ, Zhang X, Huang LD, Xiao Y. Involvement of the Sodium Channel Nav1.7 in Paclitaxel-induced Peripheral Neuropathy through ERK1/2 Signaling in Rats. Curr Neurovasc Res 2021; 17:267-274. [PMID: 32407275 DOI: 10.2174/1567202617666200514113441] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/02/2020] [Accepted: 03/07/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Paclitaxel treatment is a major cause of chemotherapy-induced peripheral neuropathy. The sodium channel Nav1.7 plays a critical role in pain perception. However, whether Nav1.7 in the dorsal root ganglion (DRG) is involved in paclitaxel-induced peripheral neuropathy remains unclear. Thus, our study aimed to evaluate whether Nav1.7 participates in the pathogenesis of paclitaxel-induced neuropathy. METHODS Paclitaxel-induced peripheral neuropathy was generated by intraperitoneal administration of paclitaxel on four alternate days. RESULTS The results showed that DRG mRNA and protein expression levels of Nav1.7 were upregulated between days 7 and 21 after the administration of paclitaxel. Besides, paclitaxel upregulated extracellular signal-regulated kinase (ERK1/2) phosphorylation in DRG. Intrathecal injection of U0126 (a MEK inhibitor) blocking ERK1/2 phosphorylation blunted up-regulation of Nav1.7 in the DRG and correspondingly attenuated hyperalgesia. CONCLUSION These results indicated that the sodium channel Nav1.7 in the DRG exerted an important function in paclitaxel-induced neuropathy, which was associated with ERK phosphorylation in neurons.
Collapse
Affiliation(s)
- Guang Jie Wang
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Xi Zhang
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Li-De Huang
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yun Xiao
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| |
Collapse
|
32
|
Middleton SJ, Perez-Sanchez J, Dawes JM. The structure of sensory afferent compartments in health and disease. J Anat 2021; 241:1186-1210. [PMID: 34528255 PMCID: PMC9558153 DOI: 10.1111/joa.13544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/12/2021] [Accepted: 08/30/2021] [Indexed: 12/20/2022] Open
Abstract
Primary sensory neurons are a heterogeneous population of cells able to respond to both innocuous and noxious stimuli. Like most neurons they are highly compartmentalised, allowing them to detect, convey and transfer sensory information. These compartments include specialised sensory endings in the skin, the nodes of Ranvier in myelinated axons, the cell soma and their central terminals in the spinal cord. In this review, we will highlight the importance of these compartments to primary afferent function, describe how these structures are compromised following nerve damage and how this relates to neuropathic pain.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - John M Dawes
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Tetrodotoxin: A New Strategy to Treat Visceral Pain? Toxins (Basel) 2021; 13:toxins13070496. [PMID: 34357968 PMCID: PMC8310099 DOI: 10.3390/toxins13070496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
Visceral pain is one of the most common symptoms associated with functional gastrointestinal (GI) disorders. Although the origin of these symptoms has not been clearly defined, the implication of both the central and peripheral nervous systems in visceral hypersensitivity is well established. The role of several pathways in visceral nociception has been explored, as well as the influence of specific receptors on afferent neurons, such as voltage-gated sodium channels (VGSCs). VGSCs initiate action potentials and dysfunction of these channels has recently been associated with painful GI conditions. Current treatments for visceral pain generally involve opioid based drugs, which are associated with important side-effects and a loss of effectiveness or tolerance. Hence, efforts have been intensified to find new, more effective and longer-lasting therapies. The implication of VGSCs in visceral hypersensitivity has drawn attention to tetrodotoxin (TTX), a relatively selective sodium channel blocker, as a possible and promising molecule to treat visceral pain and related diseases. As such, here we will review the latest information regarding this toxin that is relevant to the treatment of visceral pain and the possible advantages that it may offer relative to other treatments, alone or in combination.
Collapse
|
34
|
Oh J. Evaluation and diagnosis of neuropathic pain. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2021. [DOI: 10.5124/jkma.2021.64.7.477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background: Thorough evaluation and an accurate diagnosis of neuropathic pain are essential for effective treatment. The therapeutic approach and choice of medication for neuropathic pain are different from those for other kinds of nociceptive pain. Therefore, this study aimed to present the current evaluation and diagnostic methods for neuropathic pain.Current Concepts: Grading of the certainty of the presence of neuropathic pain according to the results of clinical history, neurological examination, and confirmatory tests improves the diagnosis of neuropathic pain. The Leeds Assessment of Neuropathic Symptoms and Signs, Neuropathic Pain Questionnaire, Douleur Neuropathique en 4 Questions, and PainDETECT are mainly used for neuropathic pain screening. During physical examination, sensory nerve function tests are more critical than other nervous system examination items, including the test of the sense of touch with a cotton swab and the sense of vibration with a tuning fork. In addition, pain sensation using pins and temperature sensation using cold metal are tested to check for nociceptive pathway abnormalities. Diagnostic tests include imaging tests, nerve conduction tests, and other neurophysiological tests, such as quantitative sensory function tests, autonomic nerve function tests, and blood tests.Discussion and Conclusion: To diagnose neuropathic pain, physicians should first determine whether patient symptoms match the characteristics of neuropathic pain. If there is a possibility of neuropathic pain, physicians should perform a neurological screening examination and a proper diagnostic test to identify the cause of pain.
Collapse
|
35
|
Sun J, Li L, Yang L, Duan G, Ma T, Li N, Liu Y, Yao J, Liu JY, Zhang X. Novel SCN9A missense mutations contribute to congenital insensitivity to pain: Unexpected correlation between electrophysiological characterization and clinical phenotype. Mol Pain 2021; 16:1744806920923881. [PMID: 32420800 PMCID: PMC7235659 DOI: 10.1177/1744806920923881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Congenital insensitivity to pain (OMIM 243000) is an extremely rare disorder caused by loss-of-function mutations in SCN9A encoding Nav1.7. Although the SCN9A mutations and phenotypes of painlessness and anosmia/hyposmia in patients are previously well documented, the complex relationship between genotype and phenotype of congenital insensitivity to pain remains unclear. Here, we report a congenital insensitivity to pain patient with novel SCN9A mutations. Functional significance of novel SCN9A mutations was assessed in HEK293 cells expressing Nav1.7, the results showed that p.Arg99His significantly decreased current density and reduced total Nav1.7 protein levels, whereas p.Trp917Gly almost abolished Nav1.7 sodium current without affecting its protein expression. These revealed that mutations in Nav1.7 in this congenital insensitivity to pain patient still retained partial channel function, but the patient showed completely painlessness, the unexpected genotypic-phenotypic relationship of SCN9A mutations in our patient may challenge the previous findings “Nav1.7 total loss-of-function leads to painlessness.” Additionally, these findings are helpful for understanding the critical amino acid for maintaining function of Nav1.7, thus contributing to the development of Nav1.7-targeted analgesics.
Collapse
Affiliation(s)
- Jiaoli Sun
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lulu Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Luyao Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Guangyou Duan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Tingbin Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ningbo Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yi Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jing Yao
- College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Jing Yu Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xianwei Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
36
|
Bouali-Benazzouz R, Landry M, Benazzouz A, Fossat P. Neuropathic pain modeling: Focus on synaptic and ion channel mechanisms. Prog Neurobiol 2021; 201:102030. [PMID: 33711402 DOI: 10.1016/j.pneurobio.2021.102030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/22/2021] [Indexed: 12/28/2022]
Abstract
Animal models of pain consist of modeling a pain-like state and measuring the consequent behavior. The first animal models of neuropathic pain (NP) were developed in rodents with a total lesion of the sciatic nerve. Later, other models targeting central or peripheral branches of nerves were developed to identify novel mechanisms that contribute to persistent pain conditions in NP. Objective assessment of pain in these different animal models represents a significant challenge for pre-clinical research. Multiple behavioral approaches are used to investigate and to validate pain phenotypes including withdrawal reflex to evoked stimuli, vocalizations, spontaneous pain, but also emotional and affective behaviors. Furthermore, animal models were very useful in investigating the mechanisms of NP. This review will focus on a detailed description of rodent models of NP and provide an overview of the assessment of the sensory and emotional components of pain. A detailed inventory will be made to examine spinal mechanisms involved in NP-induced hyperexcitability and underlying the current pharmacological approaches used in clinics with the possibility to present new avenues for future treatment. The success of pre-clinical studies in this area of research depends on the choice of the relevant model and the appropriate test based on the objectives of the study.
Collapse
Affiliation(s)
- Rabia Bouali-Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.
| | - Marc Landry
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Abdelhamid Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Pascal Fossat
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
37
|
Foxo1 selectively regulates static mechanical pain by interacting with Nav1.7. Pain 2021; 162:490-502. [PMID: 32868747 DOI: 10.1097/j.pain.0000000000002055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
ABSTRACT Mechanical allodynia is a debilitating condition for millions of patients with chronic pain. Mechanical allodynia can manifest in distinct forms, including brush-evoked dynamic and filament-evoked static allodynia. In the nervous system, the forkhead protein Foxo1 plays a critical role in neuronal structures and functions. However, the role of Foxo1 in the somatosensory signal remains unclear. Here, we found that Foxo1 selectively regulated static mechanical pain. Foxo1 knockdown decreased sensitivity to static mechanical stimuli in normal rats and attenuated static mechanical allodynia in rat models for neuropathic, inflammatory, and chemotherapy pain. Conversely, Foxo1 overexpression selectively enhanced sensitivity to static mechanical stimuli and provoked static mechanical allodynia. Furthermore, Foxo1 interacted with voltage-gated sodium Nav1.7 channels and increased the Nav1.7 current density by accelerating activation rather than by changing the expression of Nav1.7 in dorsal root ganglia neurons. In addition, the serum level of Foxo1 was found to be increased in chronic pain patients and to be positively correlated with the severity of chronic pain. Altogether, our findings suggest that serum Foxo1 level could be used as a biological marker for prediction and diagnosis of chronic pain. Moreover, selective blockade of Foxo1/Nav1.7 interaction may offer a new therapeutic approach in patients with mechanical pain.
Collapse
|
38
|
MacDonald DI, Sikandar S, Weiss J, Pyrski M, Luiz AP, Millet Q, Emery EC, Mancini F, Iannetti GD, Alles SRA, Arcangeletti M, Zhao J, Cox JJ, Brownstone RM, Zufall F, Wood JN. A central mechanism of analgesia in mice and humans lacking the sodium channel Na V1.7. Neuron 2021; 109:1497-1512.e6. [PMID: 33823138 PMCID: PMC8110947 DOI: 10.1016/j.neuron.2021.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/05/2020] [Accepted: 03/08/2021] [Indexed: 11/18/2022]
Abstract
Deletion of SCN9A encoding the voltage-gated sodium channel NaV1.7 in humans leads to profound pain insensitivity and anosmia. Conditional deletion of NaV1.7 in sensory neurons of mice also abolishes pain, suggesting that the locus of analgesia is the nociceptor. Here we demonstrate, using in vivo calcium imaging and extracellular recording, that NaV1.7 knockout mice have essentially normal nociceptor activity. However, synaptic transmission from nociceptor central terminals in the spinal cord is greatly reduced by an opioid-dependent mechanism. Analgesia is also reversed substantially by central but not peripheral application of opioid antagonists. In contrast, the lack of neurotransmitter release from olfactory sensory neurons is opioid independent. Male and female humans with NaV1.7-null mutations show naloxone-reversible analgesia. Thus, inhibition of neurotransmitter release is the principal mechanism of anosmia and analgesia in mouse and human Nav1.7-null mutants.
Collapse
Affiliation(s)
- Donald Iain MacDonald
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| | - Shafaq Sikandar
- Centre for Experimental Medicine & Rheumatology, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jan Weiss
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Martina Pyrski
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Ana P Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Queensta Millet
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Edward C Emery
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Flavia Mancini
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Gian D Iannetti
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK; Neuroscience and Behaviour Laboratory, Istituto Italiano di Tecnologia, Rome, Italy
| | - Sascha R A Alles
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Manuel Arcangeletti
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | | | - Frank Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
39
|
Goodwin G, McMahon SB. The physiological function of different voltage-gated sodium channels in pain. Nat Rev Neurosci 2021; 22:263-274. [PMID: 33782571 DOI: 10.1038/s41583-021-00444-w] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 02/01/2023]
Abstract
Evidence from human genetic pain disorders shows that voltage-gated sodium channel α-subtypes Nav1.7, Nav1.8 and Nav1.9 are important in the peripheral signalling of pain. Nav1.7 is of particular interest because individuals with Nav1.7 loss-of-function mutations are congenitally insensitive to acute and chronic pain, and there is considerable hope that phenocopying these effects with a pharmacological antagonist will produce a new class of analgesic drug. However, studies in these rare individuals do not reveal how and where voltage-gated sodium channels contribute to pain signalling, which is of critical importance for drug development. More than a decade of research utilizing rodent genetic models and pharmacological tools to study voltage-gated sodium channels in pain has begun to unravel the role of different subtypes. Here, we review the contribution of individual channel subtypes in three key physiological processes necessary for transmission of sensory information to the CNS: transduction of stimuli at peripheral nerve terminals, axonal transmission of action potentials and neurotransmitter release from central terminals. These data suggest that drugs seeking to recapitulate the analgesic effects of loss of function of Nav1.7 will need to be brain-penetrant - which most of those developed to date are not.
Collapse
Affiliation(s)
- George Goodwin
- Pain and Neurorestoration Group, King's College London, London, UK.
| | | |
Collapse
|
40
|
Mohammed ZA, Kaloyanova K, Nassar MA. An unbiased and efficient assessment of excitability of sensory neurons for analgesic drug discovery. Pain 2021; 161:1100-1108. [PMID: 31929383 PMCID: PMC7170445 DOI: 10.1097/j.pain.0000000000001802] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Distinct veratridine-induced calcium responses in nociceptors and non-nociceptors allow for unbiased and efficient assessment of drugs' action on both populations separately but simultaneously. Alleviating chronic pain is challenging, due to lack of drugs that effectively inhibit nociceptors without off-target effects on motor or central neurons. Dorsal root ganglia (DRG) contain nociceptive and non-nociceptive neurons. Drug screening on cultured DRG neurons, rather than cell lines, allows for the identification of drugs most potent on nociceptors with no effects on non-nociceptors (as a proxy for unwanted side effects on central nervous system and motor neurons). However, screening using DRG neurons is currently a low-throughput process, and there is a need for assays to speed this process for analgesic drug discovery. We previously showed that veratridine elicits distinct response profiles in sensory neurons. Here, we show evidence that a veratridine-based calcium assay allows for an unbiased and efficient assessment of a drug effect on nociceptors (targeted neurons) and non-nociceptors (nontargeted neurons). We confirmed the link between the oscillatory profile and nociceptors, and the slow-decay profile and non-nociceptors using 3 transgenic mouse lines of known pain phenotypes. We used the assay to show that blockers for Nav1.7 and Nav1.8 channels, which are validated targets for analgesics, affect non-nociceptors at concentrations needed to effectively inhibit nociceptors. However, a combination of low doses of both blockers had an additive effect on nociceptors without a significant effect on non-nociceptors, indicating that the assay can also be used to screen for combinations of existing or novel drugs for the greatest selective inhibition of nociceptors.
Collapse
Affiliation(s)
- Zainab A Mohammed
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | | | | |
Collapse
|
41
|
Nair M, Jagadeeshan S, Katselis G, Luan X, Momeni Z, Henao-Romero N, Chumala P, Tam JS, Yamamoto Y, Ianowski JP, Campanucci VA. Lipopolysaccharides induce a RAGE-mediated sensitization of sensory neurons and fluid hypersecretion in the upper airways. Sci Rep 2021; 11:8336. [PMID: 33863932 PMCID: PMC8052339 DOI: 10.1038/s41598-021-86069-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Thoracic dorsal root ganglia (tDRG) contribute to fluid secretion in the upper airways. Inflammation potentiates DRG responses, but the mechanisms remain under investigation. The receptor for advanced glycation end-products (RAGE) underlies potentiation of DRG responses in pain pathologies; however, its role in other sensory modalities is less understood. We hypothesize that RAGE contributes to electrophysiological and biochemical changes in tDRGs during inflammation. We used tDRGs and tracheas from wild types (WT), RAGE knock-out (RAGE-KO), and with the RAGE antagonist FPS-ZM1, and exposed them to lipopolysaccharides (LPS). We studied: capsaicin (CAP)-evoked currents and action potentials (AP), tracheal submucosal gland secretion, RAGE expression and downstream pathways. In WT neurons, LPS increased CAP-evoked currents and AP generation, and it caused submucosal gland hypersecretion in tracheas from WT mice exposed to LPS. In contrast, LPS had no effect on tDRG excitability or gland secretion in RAGE-KO mice or mice treated with FPS-ZM1. LPS upregulated full-length RAGE (encoded by Tv1-RAGE) and downregulated a soluble (sRAGE) splice variant (encoded by MmusRAGEv4) in tDRG neurons. These data suggest that sensitization of tDRG neurons contributes to hypersecretion in the upper airways during inflammation. And at least two RAGE variants may be involved in these effects of LPS.
Collapse
Affiliation(s)
- Manoj Nair
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Santosh Jagadeeshan
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - George Katselis
- Department of Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Xiaojie Luan
- Department of Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Zeinab Momeni
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Nicolas Henao-Romero
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Paulos Chumala
- Department of Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Julian S Tam
- Department of Medicine, Division of Respirology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, 920-8640, Japan
| | - Juan P Ianowski
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Verónica A Campanucci
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
42
|
Xue Y, Chidiac C, Herault Y, Gaveriaux-Ruff C. Pain behavior in SCN9A (Nav1.7) and SCN10A (Nav1.8) mutant rodent models. Neurosci Lett 2021; 753:135844. [PMID: 33775738 DOI: 10.1016/j.neulet.2021.135844] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 11/18/2022]
Abstract
The two voltage gated sodium channels Nav1.7 and Nav1.8 are expressed in the peripheral nervous system and involved in various pain conditions including inflammatory and neuropathic pain. Rodent models bearing deletions or mutations of the corresponding genes, Scn9a and Scn10a, were created in order to understand the role of these channels in the pathophysiological mechanism underlying pain symptoms. This review summarizes the pain behavior profiles reported in Scn9a and Scn10a rodent models. The complete loss-of-function or knockout (KO) of Scn9a or Scn10a and the conditional KO (cKO) of Scn9a in specific cell populations were shown to decrease sensitivity to various pain stimuli. The Possum mutant mice bearing a dominant hypermorphic mutation in Scn10a revealed higher sensitivity to noxious stimuli. Several gain-of-function mutations were identified in patients with painful small fiber neuropathy. Future knowledge obtained from preclinical models bearing these mutations will allow understanding how these mutations affect pain. In addition, the review gives perspectives for creating models that better mimic patients' pain symptoms in view to developing novel analgesic strategies.
Collapse
Affiliation(s)
- Yaping Xue
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) Translational Medicine and Neurogenetics Department, Illkirch, France
| | - Celeste Chidiac
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) Translational Medicine and Neurogenetics Department, Illkirch, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) Translational Medicine and Neurogenetics Department, Illkirch, France.
| | - Claire Gaveriaux-Ruff
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) Translational Medicine and Neurogenetics Department, Illkirch, France
| |
Collapse
|
43
|
Wang D, Lu J, Xu X, Yuan Y, Zhang Y, Xu J, Chen H, Liu J, Shen Y, Zhang H. Satellite Glial Cells Give Rise to Nociceptive Sensory Neurons. Stem Cell Rev Rep 2021; 17:999-1013. [PMID: 33389681 DOI: 10.1007/s12015-020-10102-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/27/2022]
Abstract
Dorsal root ganglia (DRG) sensory neurons can transmit information about noxious stimulus to cerebral cortex via spinal cord, and play an important role in the pain pathway. Alterations of the pain pathway lead to CIPA (congenital insensitivity to pain with anhidrosis) or chronic pain. Accumulating evidence demonstrates that nerve damage leads to the regeneration of neurons in DRG, which may contribute to pain modulation in feedback. Therefore, exploring the regeneration process of DRG neurons would provide a new understanding to the persistent pathological stimulation and contribute to reshape the somatosensory function. It has been reported that a subpopulation of satellite glial cells (SGCs) express Nestin and p75, and could differentiate into glial cells and neurons, suggesting that SGCs may have differentiation plasticity. Our results in the present study show that DRG-derived SGCs (DRG-SGCs) highly express neural crest cell markers Nestin, Sox2, Sox10, and p75, and differentiate into nociceptive sensory neurons in the presence of histone deacetylase inhibitor VPA, Wnt pathway activator CHIR99021, Notch pathway inhibitor RO4929097, and FGF pathway inhibitor SU5402. The nociceptive sensory neurons express multiple functionally-related genes (SCN9A, SCN10A, SP, Trpv1, and TrpA1) and are able to generate action potentials and voltage-gated Na+ currents. Moreover, we found that these cells exhibited rapid calcium transients in response to capsaicin through binding to the Trpv1 vanilloid receptor, confirming that the DRG-SGC-derived cells are nociceptive sensory neurons. Further, we show that Wnt signaling promotes the differentiation of DRG-SGCs into nociceptive sensory neurons by regulating the expression of specific transcription factor Runx1, while Notch and FGF signaling pathways are involved in the expression of SCN9A. These results demonstrate that DRG-SGCs have stem cell characteristics and can efficiently differentiate into functional nociceptive sensory neurons, shedding light on the clinical treatment of sensory neuron-related diseases.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Junhou Lu
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Xiaojing Xu
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Ye Yuan
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Yu Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jianwei Xu
- National Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Huanhuan Chen
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Jinming Liu
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Yixin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Huanxiang Zhang
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
44
|
de Clauser L, Luiz AP, Santana-Varela S, Wood JN, Sikandar S. Sensitization of Cutaneous Primary Afferents in Bone Cancer Revealed by In Vivo Calcium Imaging. Cancers (Basel) 2020; 12:cancers12123491. [PMID: 33255209 PMCID: PMC7760605 DOI: 10.3390/cancers12123491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Cancer-induced bone pain severely impairs the quality of life of cancer patients, many of whom suffer from inadequate pain relief. The development of new analgesic therapies depends on the identification of the cells and mechanisms involved in cancer-induced bone pain. Bone marrow innervating sensory neurons have been proposed to contribute to this debilitating disease, but their role remains unexplored. Here we used in vivo calcium imaging to determine the functional role of bone innervating and skin innervating neurons in contributing to pain at an advanced stage of bone cancer. Our results indicate increased excitability of skin innervating neurons, while those innervating bone are unaffected. Our data suggests skin-innervating neurons become hyperexcitable in cancer-induced bone pain and are a potential target for pain relief. Abstract Cancer-induced bone pain (CIBP) is a complex condition, comprising components of inflammatory and neuropathic processes, but changes in the physiological response profiles of bone-innervating and cutaneous afferents remain poorly understood. We used a combination of retrograde labelling and in vivo calcium imaging of bone marrow-innervating dorsal root ganglia (DRG) neurons to determine the contribution of these cells in the maintenance of CIBP. We found a majority of femoral bone afferent cell bodies in L3 dorsal root ganglia (DRG) that also express the sodium channel subtype Nav1.8—a marker of nociceptive neurons—and lack expression of parvalbumin—a marker for proprioceptive primary afferents. Surprisingly, the response properties of bone marrow afferents to both increased intraosseous pressure and acid were unchanged by the presence of cancer. On the other hand, we found increased excitability and polymodality of cutaneous afferents innervating the ipsilateral paw in cancer bearing animals, as well as a behavioural phenotype that suggests changes at the level of the DRG contribute to secondary hypersensitivity. This study demonstrates that cutaneous afferents at distant sites from the tumour bearing tissue contribute to mechanical hypersensitivity, highlighting these cells as targets for analgesia.
Collapse
Affiliation(s)
- Larissa de Clauser
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London SE1 1UL, UK
| | - Ana P. Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
| | - Sonia Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
| | - Shafaq Sikandar
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
- William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Correspondence:
| |
Collapse
|
45
|
de Clauser L, Santana-Varela S, Wood JN, Sikandar S. Physiologic osteoclasts are not sufficient to induce skeletal pain in mice. Eur J Pain 2020; 25:199-212. [PMID: 32955748 PMCID: PMC8436750 DOI: 10.1002/ejp.1662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/21/2020] [Accepted: 09/13/2020] [Indexed: 12/18/2022]
Abstract
Background Increased bone resorption is driven by augmented osteoclast activity in pathological states of the bone, including osteoporosis, fracture and metastatic bone cancer. Pain is a frequent co‐morbidity in bone pathologies and adequate pain management is necessary for symptomatic relief. Bone cancer is associated with severe skeletal pain and dysregulated bone remodelling, while increased osteoclast activity and bone pain are also observed in osteoporosis and during fracture repair. However, the effects of altered osteoclast activity and bone resorption on nociceptive processing of bone afferents remain unclear. Methods This study investigates whether physiologic osteoclasts and resulting changes in bone resorption can induce skeletal pain. We first assessed correlation between changes in bone microarchitecture (through µCT) and skeletal pain using standardized behavioural phenotyping assays in a mouse model of metastatic bone cancer. We then investigated whether increased activity of physiologic osteoclasts, and the associated bone resorption, is sufficient to induce skeletal pain using mouse models of localized and widespread bone resorption following administration of exogenous receptor activator of nuclear factor kappa‐B ligand (RANKL). Results Our data demonstrates that mice with bone cancer exhibit progressive pain behaviours that correlate with increased bone resorption at the tumour site. Systemic RANKL injections enhance osteoclast activity and associated bone resorption, without producing any changes in motor function or pain behaviours at both early and late timepoints. Conclusion These findings suggest that activation of homeostatic osteoclasts alone is not sufficient to induce skeletal pain in mice. Significance statement The role of osteoclasts in peripheral sensitization of sensory neurones is not fully understood. This study reports on the direct link between oestrogen‐independent osteoclast activation and skeletal pain. Administration of exogenous receptor activator of nuclear factor kappa‐B ligand (RANKL) increases bone resorption, but does not produce pro‐nociceptive changes in behavioural pain thresholds. Our data demonstrates that physiologic osteoclasts are not essential for skeletal pain behaviours.
Collapse
Affiliation(s)
- Larissa de Clauser
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK.,Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Sonia Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Shafaq Sikandar
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, UK.,William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Mary University of London, London, UK
| |
Collapse
|
46
|
Bagdas D, Paris JJ, Carper M, Wodarski R, Rice AS, Knapp PE, Hauser KF, Damaj MI. Conditional expression of HIV-1 tat in the mouse alters the onset and progression of tonic, inflammatory and neuropathic hypersensitivity in a sex-dependent manner. Eur J Pain 2020; 24:1609-1623. [PMID: 32533878 PMCID: PMC7856573 DOI: 10.1002/ejp.1618] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 05/21/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND At least one-third of HIV-1-afflicted individuals experience peripheral neuropathy. Although the underlying mechanisms are not known, they may involve neurotoxic HIV-1 proteins. METHODS We assessed the influence of the neurotoxic HIV-1 regulatory protein, Tat, on inflammatory and neuropathic nociceptive behaviours using transgenic male and female mice that conditionally expressed (or did not express) HIV-1 Tat1-86 in fibrillary acidic protein-expressing glia in the central and peripheral nervous systems. RESULTS Tat induction significantly attenuated the time spent paw-licking following formalin injection (2.5%, i.pl.) in both male and female mice. However, significant sex differences were observed in the onset and magnitude of inflammation and sensory sensitivity following complete Freund's adjuvant (CFA) injection (10%, i.pl.) after Tat activation. Unlike female mice, male mice showed a significant attenuation of paw swelling and an absence of mechanical/thermal hypersensitivity in response to CFA after Tat induction. Male Tat(+) mice also showed accelerated recovery from chronic constrictive nerve injury (CCI)-induced neuropathic mechanical and thermal hypersensitivity compared to female Tat(+) mice. Morphine (3.2 mg/kg) fully reversed CCI-induced mechanical hypersensitivity in female Tat(-) mice, but not in Tat(+) females. CONCLUSIONS The ability of Tat to decrease oedema, paw swelling, and limit allodynia suggests a sequel of events in which Tat-induced functional deficits precede the onset of mechanical hypersensitivity. Moreover, HIV-1 Tat attenuated responses to inflammatory and neuropathic insults in a sex-dependent manner. HIV-1 Tat appears to directly contribute to HIV sensory neuropathy and reveals sex differences in HIV responsiveness and/or the underlying peripheral neuroinflammatory and nociceptive mechanisms.
Collapse
Affiliation(s)
- Deniz Bagdas
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- The Center for the Study for Tobacco Products, Virginia Commonwealth University, Richmond, VA 23284-2018, USA
| | - Jason J. Paris
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA
- Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA
| | - Moriah Carper
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Rachel Wodarski
- Pain Research Group, Department of Surgery & Cancer, Imperial College, London, SW10 9NH, UK
| | - Andrew S.C. Rice
- Pain Research Group, Department of Surgery & Cancer, Imperial College, London, SW10 9NH, UK
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Department of Anatomy & Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Institute for Drug and Alcohol Studies, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Department of Anatomy & Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Institute for Drug and Alcohol Studies, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- The Center for the Study for Tobacco Products, Virginia Commonwealth University, Richmond, VA 23284-2018, USA
- Translational Research Initiative for Pain and Neuropathy at VCU, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| |
Collapse
|
47
|
Israel MR, Dash TS, Bothe SN, Robinson SD, Deuis JR, Craik DJ, Lampert A, Vetter I, Durek T. Characterization of Synthetic Tf2 as a Na V1.3 Selective Pharmacological Probe. Biomedicines 2020; 8:biomedicines8060155. [PMID: 32545167 PMCID: PMC7345637 DOI: 10.3390/biomedicines8060155] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022] Open
Abstract
NaV1.3 is a subtype of the voltage-gated sodium channel family. It has been implicated in the pathogenesis of neuropathic pain, although the contribution of this channel to neuronal excitability is not well understood. Tf2, a β-scorpion toxin previously identified from the venom of Tityus fasciolatus, has been reported to selectively activate NaV1.3. Here, we describe the activity of synthetic Tf2 and assess its suitability as a pharmacological probe for NaV1.3. As described for the native toxin, synthetic Tf2 (1 µM) caused early channel opening, decreased the peak current, and shifted the voltage dependence of NaV1.3 activation in the hyperpolarizing direction by −11.3 mV, with no activity at NaV1.1, NaV1.2, and NaV1.4-NaV1.8. Additional activity was found at NaV1.9, tested using the hNav1.9_C4 chimera, where Tf2 (1 µM) shifted the voltage dependence of activation by −6.3 mV. In an attempt to convert Tf2 into an NaV1.3 inhibitor, we synthetized the analogue Tf2[S14R], a mutation previously described to remove the excitatory activity of related β-scorpion toxins. Indeed, Tf2[S14R](10 µM) had reduced excitatory activity at NaV1.3, although it still caused a small −5.8 mV shift in the voltage dependence of activation. Intraplantar injection of Tf2 (1 µM) in mice caused spontaneous flinching and swelling, which was not reduced by the NaV1.1/1.3 inhibitor ICA-121431 nor in NaV1.9-/- mice, suggesting off-target activity. In addition, despite a loss of excitatory activity, intraplantar injection of Tf2[S14R](10 µM) still caused swelling, providing strong evidence that Tf2 has additional off-target activity at one or more non-neuronal targets. Therefore, due to activity at NaV1.9 and other yet to be identified target(s), the use of Tf2 as a selective pharmacological probe may be limited.
Collapse
Affiliation(s)
- Mathilde R. Israel
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.R.I.); (T.S.D.); (S.D.R.); (J.R.D.); (D.J.C.)
| | - Thomas S. Dash
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.R.I.); (T.S.D.); (S.D.R.); (J.R.D.); (D.J.C.)
| | - Stefanie N. Bothe
- Institute of Physiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (S.N.B.); (A.L.)
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, 52074 Aachen, Germany
| | - Samuel D. Robinson
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.R.I.); (T.S.D.); (S.D.R.); (J.R.D.); (D.J.C.)
| | - Jennifer R. Deuis
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.R.I.); (T.S.D.); (S.D.R.); (J.R.D.); (D.J.C.)
| | - David J. Craik
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.R.I.); (T.S.D.); (S.D.R.); (J.R.D.); (D.J.C.)
| | - Angelika Lampert
- Institute of Physiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany; (S.N.B.); (A.L.)
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, 52074 Aachen, Germany
- Research Training Group 2415 ME3T, RWTH Aachen University, 52074 Aachen, Germany
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.R.I.); (T.S.D.); (S.D.R.); (J.R.D.); (D.J.C.)
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
- Correspondence: (I.V.); (T.D.); Tel.: +61-7-3346-2660 (I.V.); +61-7-3346-2021 (T.D.)
| | - Thomas Durek
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.R.I.); (T.S.D.); (S.D.R.); (J.R.D.); (D.J.C.)
- Correspondence: (I.V.); (T.D.); Tel.: +61-7-3346-2660 (I.V.); +61-7-3346-2021 (T.D.)
| |
Collapse
|
48
|
Grubinska B, Chen L, Alsaloum M, Rampal N, Matson DJ, Yang C, Taborn K, Zhang M, Youngblood B, Liu D, Galbreath E, Allred S, Lepherd M, Ferrando R, Kornecook TJ, Lehto SG, Waxman SG, Moyer BD, Dib-Hajj S, Gingras J. Rat Na V1.7 loss-of-function genetic model: Deficient nociceptive and neuropathic pain behavior with retained olfactory function and intra-epidermal nerve fibers. Mol Pain 2020; 15:1744806919881846. [PMID: 31550995 PMCID: PMC6831982 DOI: 10.1177/1744806919881846] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recapitulating human disease pathophysiology using genetic animal models is a
powerful approach to enable mechanistic understanding of genotype–phenotype
relationships for drug development. NaV1.7 is a sodium channel
expressed in the peripheral nervous system with strong human genetic validation
as a pain target. Efforts to identify novel analgesics that are nonaddictive
resulted in industry exploration of a class of sulfonamide compounds that bind
to the fourth voltage-sensor domain of NaV1.7. Due to sequence
differences in this region, sulfonamide blockers generally are potent on human
but not rat NaV1.7 channels. To test sulfonamide-based chemical
matter in rat models of pain, we generated a humanized NaV1.7 rat
expressing a chimeric NaV1.7 protein containing the
sulfonamide-binding site of the human gene sequence as a replacement for the
equivalent rat sequence. Unexpectedly, upon transcription, the human insert was
spliced out, resulting in a premature stop codon. Using a validated antibody,
NaV1.7 protein was confirmed to be lost in the brainstem, dorsal
root ganglia, sciatic nerve, and gastrointestinal tissue but not in nasal
turbinates or olfactory bulb in rats homozygous for the knock-in allele
(HOM-KI). HOM-KI rats exhibited normal intraepidermal nerve fiber density with
reduced tetrodotoxin-sensitive current density and action potential firing in
small diameter dorsal root ganglia neurons. HOM-KI rats did not exhibit
nociceptive pain responses in hot plate or capsaicin-induced flinching assays
and did not exhibit neuropathic pain responses following spinal nerve ligation.
Consistent with expression of chimeric NaV1.7 in olfactory tissue,
HOM-KI rats retained olfactory function. This new genetic model highlights the
necessity of NaV1.7 for pain behavior in rats and indicates that
sufficient inhibition of NaV1.7 in humans may reduce pain in
neuropathic conditions. Due to preserved olfactory function, this rat model
represents an alternative to global NaV1.7 knockout mice that require
time-intensive hand feeding during early postnatal development.
Collapse
Affiliation(s)
- B Grubinska
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Voyager Therapeutics, Cambridge, MA, USA
| | - L Chen
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - M Alsaloum
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - N Rampal
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - D J Matson
- Neuroscience Department, Amgen Research, Cambridge, MA, USA
| | - C Yang
- Neuroscience Department, Amgen Research, Cambridge, MA, USA
| | - K Taborn
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Wave Life Sciences, Ltd, Cambridge, MA, USA
| | - M Zhang
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - B Youngblood
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - D Liu
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - E Galbreath
- Comparative Biology and Safety Sciences, Amgen Research, Cambridge, MA, USA.,Takeda Pharmaceutical Company Ltd, Cambridge, MA, USA
| | - S Allred
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,Seattle Genetics, Bothell, WA, USA
| | - M Lepherd
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,Genentech, Inc. South San Francisco, CA, USA
| | - R Ferrando
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,AbbVie Stemcentrx, Inc., South San Francisco, CA, USA
| | - T J Kornecook
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA.,Biogen Inc., Cambridge, MA, USA
| | - S G Lehto
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - S G Waxman
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - B D Moyer
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - S Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - J Gingras
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Homology Medicine Inc., Bedford, MA, USA
| |
Collapse
|
49
|
Inprasit C, Lin YW. TRPV1 Responses in the Cerebellum Lobules V, VIa and VII Using Electroacupuncture Treatment for Inflammatory Hyperalgesia in Murine Model. Int J Mol Sci 2020; 21:ijms21093312. [PMID: 32392831 PMCID: PMC7247539 DOI: 10.3390/ijms21093312] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/01/2020] [Accepted: 05/06/2020] [Indexed: 01/01/2023] Open
Abstract
Inflammatory pain sensation is an important symptom which protects the body against additional tissue damage and promotes healing. Discovering long-term and effective treatments for pain remains crucial in providing efficient healthcare. Electroacupuncture (EA) is a successful therapy used for pain relief. We aimed to investigate effects and mechanisms of Complete Freund’s Adjuvant (CFA)-inducing inflammatory pain in the cerebellum, and the inhibition of this inflammatory hyperalgesia using EA at Zusanli acupoint (ST36). The results display a significant increase in mechanical and thermal sensitivities in the CFA and CFA + SHAM groups, which was significantly reduced in the CFA+EA and CFA + KO groups. This evidence was substantiated in the protein levels observed using immunoblotting, and presented with significant escalations after CFA inducing inflammatory hyperalgesia in CFA and CFA + SHAM groups. Then, they were significantly attenuated by EA in the CFA + EA group. Furthermore, the CFA + transient receptor vanilloid member 1 (TRPV1)−/− group indicated similar significant decreases of protein expression. Additionally, a concomitant overexpression in lobule VIa was also observed in immunofluorescence. These consequences suggest that CFA-induced inflammatory pain provokes modifications in cerebellum lobules V, VIa and VII, which can subsequently be regulated by EA treatment at the ST36 through its action on TRPV1 and related molecular pathways.
Collapse
Affiliation(s)
- Chanya Inprasit
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan;
| | - Yi-Wen Lin
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan;
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Correspondence:
| |
Collapse
|
50
|
Rosenberger DC, Blechschmidt V, Timmerman H, Wolff A, Treede RD. Challenges of neuropathic pain: focus on diabetic neuropathy. J Neural Transm (Vienna) 2020; 127:589-624. [PMID: 32036431 PMCID: PMC7148276 DOI: 10.1007/s00702-020-02145-7] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/19/2020] [Indexed: 02/07/2023]
Abstract
Neuropathic pain is a frequent condition caused by a lesion or disease of the central or peripheral somatosensory nervous system. A frequent cause of peripheral neuropathic pain is diabetic neuropathy. Its complex pathophysiology is not yet fully elucidated, which contributes to underassessment and undertreatment. A mechanism-based treatment of painful diabetic neuropathy is challenging but phenotype-based stratification might be a way to develop individualized therapeutic concepts. Our goal is to review current knowledge of the pathophysiology of peripheral neuropathic pain, particularly painful diabetic neuropathy. We discuss state-of-the-art clinical assessment, validity of diagnostic and screening tools, and recommendations for the management of diabetic neuropathic pain including approaches towards personalized pain management. We also propose a research agenda for translational research including patient stratification for clinical trials and improved preclinical models in relation to current knowledge of underlying mechanisms.
Collapse
Affiliation(s)
- Daniela C Rosenberger
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Vivian Blechschmidt
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Hans Timmerman
- Department of Anesthesiology, Pain Center, University Medical Center of Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - André Wolff
- Department of Anesthesiology, Pain Center, University Medical Center of Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Rolf-Detlef Treede
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|