1
|
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
|
2
|
Midavaine É, Moraes BC, Benitez J, Rodriguez SR, Braz JM, Kochhar NP, Eckalbar WL, Domingos AI, Pintar JE, Basbaum AI, Kashem SW. Regulatory T cell-derived enkephalin gates nociception. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593442. [PMID: 38798460 PMCID: PMC11118376 DOI: 10.1101/2024.05.11.593442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
T cells have emerged as sex-dependent orchestrators of pain chronification but the sexually dimorphic mechanisms by which T cells control pain sensitivity is not resolved. Here, we demonstrate an influence of regulatory T cells (Tregs) on pain processing that is distinct from their canonical functions of immune regulation and tissue repair. Specifically, meningeal Tregs (mTregs) express the endogenous opioid, enkephalin, and mTreg-derived enkephalin exerts an antinociceptive action through a presynaptic opioid receptor signaling mechanism that is dispensable for immunosuppression. We demonstrate that mTregs are both necessary and sufficient to suppress mechanical pain sensitivity in female, but not male, mice, with this modulation reliant on sex hormones. These results uncover a fundamental sex-specific, and immunologically-derived endogenous opioid circuit for nociceptive regulation with critical implications for pain biology.
Collapse
Affiliation(s)
- Élora Midavaine
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Beatriz C. Moraes
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Jorge Benitez
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Sian R. Rodriguez
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Joao M. Braz
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Nathan P. Kochhar
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Walter L. Eckalbar
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Ana I. Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - John E. Pintar
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Allan I. Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Sakeen W. Kashem
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Dermatology, Veterans Affairs Medical Center, San Francisco, California, USA
| |
Collapse
|
3
|
Gupta A, Gomes I, Osman A, Fujita W, Devi LA. Regulation of Cannabinoid and Opioid Receptor Levels by Endogenous and Pharmacological Chaperones. J Pharmacol Exp Ther 2024; 391:279-288. [PMID: 39103231 PMCID: PMC11493451 DOI: 10.1124/jpet.124.002187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
Cannabinoid and opioid receptor activities can be modulated by a variety of post-translational mechanisms including the formation of interacting complexes. This study examines the involvement of endogenous and exogenous chaperones in modulating the abundance and activity of cannabinoid CB1 receptor (CB1R), δ opioid receptor (DOR), and CB1R-DOR interacting complexes. Focusing on endogenous protein chaperones, namely receptor transporter proteins (RTPs), we examined relative mRNA expression in the mouse spinal cord and found RTP4 to be expressed at higher levels compared with other RTPs. Next, we assessed the effect of RTP4 on receptor abundance by manipulating RTP4 expression in cell lines. Overexpression of RTP4 causes an increase and knock-down causes a decrease in the levels of CB1R, DOR, and CB1R-DOR interacting complexes; this is accompanied by parallel changes in signaling. The ability of small molecule lipophilic ligands to function as exogenous chaperones was examined using receptor-selective antagonists. Long-term treatment leads to increases in receptor abundance and activity with no changes in mRNA supporting a role as pharmacological chaperones. Finally, the effect of cannabidiol (CBD), a small molecule ligand and a major active component of cannabis, on receptor abundance and activity in mice was examined. We find that CBD administration leads to increases in receptor abundance and activity in mouse spinal cord. Together, these results highlight a role for chaperones (proteins and small molecules) in modulating levels and activity of CB1R, DOR, and their interacting complexes potentially through mechanisms including receptor maturation and trafficking. SIGNIFICANCE STATEMENT: This study highlights a role for chaperones (endogenous and small membrane-permeable molecules) in modulating levels of cannabinoid CB1 receptor, delta opioid receptor, and their interacting complexes. These chaperones could be developed as therapeutics for pathologies involving these receptors.
Collapse
MESH Headings
- Animals
- Mice
- Molecular Chaperones/metabolism
- Receptor, Cannabinoid, CB1/metabolism
- Mice, Inbred C57BL
- Spinal Cord/metabolism
- Spinal Cord/drug effects
- Humans
- Cannabidiol/pharmacology
- Receptors, Opioid, delta/metabolism
- Male
- Receptors, Opioid/metabolism
- Receptors, Opioid/genetics
- HEK293 Cells
- Receptors, Cannabinoid/metabolism
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
Collapse
Affiliation(s)
- Achla Gupta
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ivone Gomes
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aya Osman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Wakako Fujita
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
4
|
Staedtler ES, Sapio MR, King DM, Maric D, Ghetti A, Mannes AJ, Iadarola MJ. The μ-opioid receptor differentiates two distinct human nociceptive populations relevant to clinical pain. Cell Rep Med 2024; 5:101788. [PMID: 39413733 PMCID: PMC11513826 DOI: 10.1016/j.xcrm.2024.101788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/26/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
The shortfall in new analgesic agents is a major impediment to reducing reliance on opioid medications for control of severe pain. In both animals and man, attenuating nociceptive transmission from primary afferent neurons with a μ-opioid receptor agonist yields highly effective analgesia. Consequently, deeper molecular characterization of human nociceptive afferents expressing OPRM1, the μ-opioid receptor gene, is a key component for advancing analgesic drug discovery and understanding clinical pain control. A co-expression matrix for the μ-opioid receptor and a variety of nociceptive channels as well as δ- and κ-opioid receptors is established by multiplex in situ hybridization. Our results indicate an OPRM1-positive population with strong molecular resemblance to rodent peptidergic C-nociceptors associated with tissue damage pain and an OPRM1-negative population sharing molecular characteristics of murine non-peptidergic C-nociceptors. The empirical identification of two distinct human nociceptive populations that differ profoundly in their presumed responsiveness to opioids provides an actionable translational framework for human pain control.
Collapse
Affiliation(s)
- Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | | | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
5
|
Fagan RR, Lee DF, Geron M, Scherrer G, von Zastrow M, Ehrlich AT. Selective targeting of mu opioid receptors to primary cilia. Cell Rep 2024; 43:114164. [PMID: 38678559 PMCID: PMC11257377 DOI: 10.1016/j.celrep.2024.114164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2024] [Accepted: 04/11/2024] [Indexed: 05/01/2024] Open
Abstract
Opioid receptors are therapeutically important G protein-coupled receptors (GPCRs) with diverse neuromodulatory effects. The functional consequences of opioid receptor activation are known to depend on receptor location in the plasma membrane, but mechanisms mediating selective localization of receptors to any particular membrane domain remain elusive. Here, we demonstrate the targeting of the mu opioid receptor (MOR) to the primary cilium, a discrete microdomain of the somatic plasma membrane, both in vivo and in cultured cells. We further show that ciliary targeting is specific to MORs, requires a 17-residue sequence unique to the MOR cytoplasmic tail, and additionally requires the Tubby-like protein 3 (TULP3) ciliary adaptor protein. Our results reveal the potential for opioid receptors to undergo selective localization to the primary cilium. We propose that ciliary targeting is mediated through an elaboration of the recycling pathway, directed by a specific C-terminal recycling sequence in cis and requiring TULP3 in trans.
Collapse
Affiliation(s)
- Rita R Fagan
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David F Lee
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matan Geron
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; New York Stem Cell Foundation, Chapel Hill, NC 27599, USA
| | - Mark von Zastrow
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Aliza T Ehrlich
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
6
|
Kawasaki A, Matsuzaki Y, Kawada T. Neuroregulatory Effects of Microcone Patch Stimulation on the Auricular Branch of the Vagus Nerve and the Prefrontal Cortex: A Feasibility Study. J Clin Med 2024; 13:2399. [PMID: 38673672 PMCID: PMC11051441 DOI: 10.3390/jcm13082399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Background: The primary purpose of this study was to preliminarily examine the effects of autonomic nervous system activity on the dorsolateral prefrontal cortex. Recent studies have examined approaches to modulating autonomic activity using invasive and non-invasive methods, but the effects of changes in autonomic activity during cognitive tasks on the dorsolateral prefrontal cortex have not been fully investigated. The purpose of this preliminary investigation was to examine changes in autonomic activity and blood oxygen saturation in the dorsolateral prefrontal cortex during reading tasks induced by vagus nerve stimulation using a microcone patch. Methods: A cohort of 40 typically developing adults was enrolled in this study. We carefully examined changes in autonomic nervous system activity and blood oxygen saturation in the dorsolateral prefrontal cortex during a reading task in two conditions: with and without microcone patch stimulation. Results: Significant changes in brain activation in the dorsolateral prefrontal cortext due to microcone patch stimulation were confirmed. In addition, hierarchical multiple regression analysis revealed specific changes in reading task-related blood oxygen saturation in the dorsolateral prefrontal region during microcone patch stimulation. Conclusions: It should be recognized that this study is a preliminary investigation and does not have immediate clinical applications. However, our results suggest that changes in autonomic nervous system activity induced by external vagal stimulation may affect activity in specific reading-related regions of the dorsolateral prefrontal cortex. Further research and evaluation are needed to fully understand the implications and potential applications of these findings.
Collapse
Affiliation(s)
- Akihiro Kawasaki
- College of Social Csciences, Ritsumeikan University, Kyoto 603-8577, Japan
- Graduate School of Education, Tohoku University, Sendai 980-8576, Japan;
| | - Yutaka Matsuzaki
- Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai 980-8575, Japan;
| | - Taku Kawada
- Graduate School of Education, Tohoku University, Sendai 980-8576, Japan;
- Sendai Shirayuri Gakuen Elementary School, Sendai 981-3205, Japan
| |
Collapse
|
7
|
Negm A, Stobbe K, Ben Fradj S, Sanchez C, Landra-Willm A, Richter M, Fleuriot L, Debayle D, Deval E, Lingueglia E, Rovere C, Noel J. Acid-sensing ion channel 3 mediates pain hypersensitivity associated with high-fat diet consumption in mice. Pain 2024; 165:470-486. [PMID: 37733484 DOI: 10.1097/j.pain.0000000000003030] [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: 09/19/2022] [Accepted: 07/07/2023] [Indexed: 09/23/2023]
Abstract
ABSTRACT Lipid-rich diet is the major cause of obesity, affecting 13% of the worldwide adult population. Obesity is a major risk factor for metabolic syndrome that includes hyperlipidemia and diabetes mellitus. The early phases of metabolic syndrome are often associated with hyperexcitability of peripheral small diameter sensory fibers and painful diabetic neuropathy. Here, we investigated the effect of high-fat diet-induced obesity on the activity of dorsal root ganglion (DRG) sensory neurons and pain perception. We deciphered the underlying cellular mechanisms involving the acid-sensing ion channel 3 (ASIC3). We show that mice made obese through consuming high-fat diet developed the metabolic syndrome and prediabetes that was associated with heat pain hypersensitivity, whereas mechanical sensitivity was not affected. Concurrently, the slow conducting C fibers in the skin of obese mice showed increased activity on heating, whereas their mechanosensitivity was not altered. Although ASIC3 knockout mice fed with high-fat diet became obese, and showed signs of metabolic syndrome and prediabetes, genetic deletion, and in vivo pharmacological inhibition of ASIC3, protected mice from obesity-induced thermal hypersensitivity. We then deciphered the mechanisms involved in the heat hypersensitivity of mice and found that serum from high-fat diet-fed mice was enriched in lysophosphatidylcholine (LPC16:0, LPC18:0, and LPC18:1). These enriched lipid species directly increased the activity of DRG neurons through activating the lipid sensitive ASIC3 channel. Our results identify ASIC3 channel in DRG neurons and circulating lipid species as a mechanism contributing to the hyperexcitability of nociceptive neurons that can cause pain associated with lipid-rich diet consumption and obesity.
Collapse
Affiliation(s)
- Ahmed Negm
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Katharina Stobbe
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Selma Ben Fradj
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Clara Sanchez
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Arnaud Landra-Willm
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Margaux Richter
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | | | | | - Emmanuel Deval
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Eric Lingueglia
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Carole Rovere
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Jacques Noel
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| |
Collapse
|
8
|
Uniyal A, Tiwari V, Tsukamoto T, Dong X, Guan Y, Raja SN. Targeting sensory neuron GPCRs for peripheral neuropathic pain. Trends Pharmacol Sci 2023; 44:1009-1027. [PMID: 37977131 PMCID: PMC10657387 DOI: 10.1016/j.tips.2023.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023]
Abstract
Despite the high prevalence of peripheral neuropathic pain (NP) conditions and significant progress in understanding its underlying mechanisms, the management of peripheral NP remains inadequate. Existing pharmacotherapies for NP act primarily on the central nervous system (CNS) and are often associated with CNS-related adverse effects, limiting their clinical effectiveness. Mounting preclinical evidence indicates that reducing the heightened activity in primary sensory neurons by targeting G-protein-coupled receptors (GPCRs), without activating these receptors in the CNS, relieves pain without central adverse effects. In this review, we focus on recent advancements in GPCR-mediated peripheral pain relief and discuss strategies to advance the development of more effective and safer therapies for peripheral NP by shifting from traditional CNS modulatory approaches toward selective targeting of GPCRs on primary sensory neurons.
Collapse
Affiliation(s)
- Ankit Uniyal
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U), Varanasi, India
| | - Takashi Tsukamoto
- Department of Neurology and Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xinzhong Dong
- Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, USA
| | - Yun Guan
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Neurological Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Srinivasa N Raja
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Neurology and Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Inyang KE, Sim J, Clark KB, Matan G, Monahan K, Evans C, Beng P, Ma JV, Heijnen CJ, Dantzer R, Scherrer G, Kavelaars A, Bernard M, Aldhamen Y, Folger JK, Laumet G. Tonic Meningeal Interleukin-10 Upregulates Delta Opioid Receptor to Prevent Relapse to Pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544200. [PMID: 37333074 PMCID: PMC10274865 DOI: 10.1101/2023.06.08.544200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Chronic pain often alternates between transient remission and relapse of severe pain. While most research on chronic pain has focused on mechanisms maintaining pain, there is a critical unmet need to understand what prevents pain from re-emerging in those who recover from acute pain. We found that interleukin (IL)-10, a pain resolving cytokine, is persistently produced by resident macrophages in the spinal meninges during remission from pain. IL-10 upregulated expression and analgesic activity of δ-opioid receptor (δOR) in the dorsal root ganglion. Genetic or pharmacological inhibition of IL-10 signaling or δOR triggered relapse to pain in both sexes. These data challenge the widespread assumption that remission of pain is simply a return to the naïve state before pain was induced. Instead, our findings strongly suggest a novel concept that: remission is a state of lasting pain vulnerability that results from a long-lasting neuroimmune interactions in the nociceptive system.
Collapse
|
10
|
Wang R, Wu X, Tian Z, Hu T, Cai C, Wu G, Jiang GB, Liu B. Sustained release of hydrogen sulfide from anisotropic ferrofluid hydrogel for the repair of spinal cord injury. Bioact Mater 2023; 23:118-128. [DOI: 10.1016/j.bioactmat.2022.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/26/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
|
11
|
Coutens B, Ingram SL. Key differences in regulation of opioid receptors localized to presynaptic terminals compared to somas: Relevance for novel therapeutics. Neuropharmacology 2023; 226:109408. [PMID: 36584882 PMCID: PMC9898207 DOI: 10.1016/j.neuropharm.2022.109408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/05/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
Opioid receptors are G protein-coupled receptors (GPCRs) that regulate activity within peripheral, subcortical and cortical circuits involved in pain, reward, and aversion processing. Opioid receptors are expressed in both presynaptic terminals where they inhibit neurotransmitter release and postsynaptic locations where they act to hyperpolarize neurons and reduce activity. Agonist activation of postsynaptic receptors at the plasma membrane signal via ion channels or cytoplasmic second messengers. Agonist binding initiates regulatory processes that include phosphorylation by G protein receptor kinases (GRKs) and recruitment of beta-arrestins that desensitize and internalize the receptors. Opioid receptors also couple to effectors from endosomes activating intracellular enzymes and kinases. In contrast to postsynaptic opioid receptors, receptors localized to presynaptic terminals are resistant to desensitization such that there is no loss of signaling in the continuous presence of opioids over the same time scale. Thus, the balance of opioid signaling in circuits expressing pre- and postsynaptic opioid receptors is shifted toward inhibition of presynaptic neurotransmitter release during continuous opioid exposure. The functional implication of this shift is not often acknowledged in behavioral studies. This review covers what is currently understood about regulation of opioid/nociceptin receptors, with an emphasis on opioid receptor signaling in pain and reward circuits. Importantly, the review covers regulation of presynaptic receptors and the critical gaps in understanding this area, as well as the opportunities to further understand opioid signaling in brain circuits. This article is part of the Special Issue on "Opioid-induced changes in addiction and pain circuits".
Collapse
Affiliation(s)
- Basile Coutens
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Susan L Ingram
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
12
|
Smith KM, Nguyen E, Ross SE. The Delta-Opioid Receptor Bidirectionally Modulates Itch. THE JOURNAL OF PAIN 2023; 24:264-272. [PMID: 36464136 PMCID: PMC10866011 DOI: 10.1016/j.jpain.2022.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 12/05/2022]
Abstract
Opioid signaling has been shown to be critically important in the neuromodulation of sensory circuits in the superficial spinal cord. Agonists of the mu-opioid receptor (MOR) elicit itch, whereas agonists of the kappa-opioid receptor (KOR) have been shown to inhibit itch. Despite the clear roles of MOR and KOR for the modulation itch, whether the delta-opioid receptor (DOR) is involved in the regulation of itch remained unknown. Here, we show that intrathecal administration of DOR agonists suppresses chemical itch and that intrathecal application of DOR antagonists is sufficient to evoke itch. We identify that spinal enkephalin neurons co-express neuropeptide Y (NPY), a peptide previously implicated in the inhibition of itch. In the spinal cord, DOR overlapped with both the NPY receptor (NPY1R) and KOR, suggesting that DOR neurons represent a site for convergent itch information in the dorsal horn. Lastly, we found that neurons co-expressing DOR and KOR showed significant Fos induction following pruritogen-evoked itch. These results uncover a role for DOR in the modulation of itch in the superficial dorsal horn. PERSPECTIVE: This article reveals the role of the delta-opioid receptor in itch. Intrathecal administration of delta agonists suppresses itch whereas the administration of delta antagonists is sufficient to induce itch. These studies highlight the importance of delta-opioid signaling for the modulation of itch behaviors, which may represent new targets for the management of itch disorders.
Collapse
Affiliation(s)
- Kelly M Smith
- University of Pittsburgh School of Medicine, Department of Neurobiology,Pittsburgh, Pennsylvania; University of Pittsburgh, Pittsburgh Center for Pain Research, Pittsburgh, Pennsylvania
| | - Eileen Nguyen
- University of Pittsburgh School of Medicine, Department of Neurobiology,Pittsburgh, Pennsylvania; University of Pittsburgh, Pittsburgh Center for Pain Research, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine, Medical Scientist Training Program, Pittsburgh, Pennsylvania
| | - Sarah E Ross
- University of Pittsburgh School of Medicine, Department of Neurobiology,Pittsburgh, Pennsylvania; University of Pittsburgh, Pittsburgh Center for Pain Research, Pittsburgh, Pennsylvania.
| |
Collapse
|
13
|
Zhang J, Junigan JM, Trinh R, Kavelaars A, Heijnen CJ, Grace PM. HDAC6 Inhibition Reverses Cisplatin-Induced Mechanical Hypersensitivity via Tonic Delta Opioid Receptor Signaling. J Neurosci 2022; 42:7862-7874. [PMID: 36096670 PMCID: PMC9617617 DOI: 10.1523/jneurosci.1182-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/20/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022] Open
Abstract
Peripheral neuropathic pain induced by the chemotherapeutic cisplatin can persist for months to years after treatment. Histone deacetylase 6 (HDAC6) inhibitors have therapeutic potential for cisplatin-induced neuropathic pain since they persistently reverse mechanical hypersensitivity and spontaneous pain in rodent models. Here, we investigated the mechanisms underlying reversal of mechanical hypersensitivity in male and female mice by a 2 week treatment with an HDAC6 inhibitor, administered 3 d after the last dose of cisplatin. Mechanical hypersensitivity in animals of both sexes treated with the HDAC6 inhibitor was temporarily reinstated by a single injection of the neutral opioid receptor antagonist 6β-naltrexol or the peripherally restricted opioid receptor antagonist naloxone methiodide. These results suggest that tonic peripheral opioid ligand-receptor signaling mediates reversal of cisplatin-induced mechanical hypersensitivity after treatment with an HDAC6 inhibitor. Pointing to a specific role for δ opioid receptors (DORs), Oprd1 expression was decreased in DRG neurons following cisplatin administration, but normalized after treatment with an HDAC6 inhibitor. Mechanical hypersensitivity was temporarily reinstated in both sexes by a single injection of the DOR antagonist naltrindole. Consistently, HDAC6 inhibition failed to reverse cisplatin-induced hypersensitivity when DORs were genetically deleted from advillin+ neurons. Mechanical hypersensitivity was also temporarily reinstated in both sexes by a single injection of a neutralizing antibody against the DOR ligand met-enkephalin. In conclusion, we reveal that treatment with an HDAC6 inhibitor induces tonic enkephalin-DOR signaling in peripheral sensory neurons to suppress mechanical hypersensitivity.SIGNIFICANCE STATEMENT Over one-fourth of cancer survivors suffer from intractable painful chemotherapy-induced peripheral neuropathy (CIPN), which can last for months to years after treatment ends. HDAC6 inhibition is a novel strategy to reverse CIPN without negatively interfering with tumor growth, but the mechanisms responsible for persistent reversal are not well understood. We built on evidence that the endogenous opioid system contributes to the spontaneous, apparent resolution of pain caused by nerve damage or inflammation, referred to as latent sensitization. We show that blocking the δ opioid receptor or its ligand enkephalin unmasks CIPN in mice treated with an HDAC6 inhibitor (latent sensitization). Our work provides insight into the mechanisms by which treatment with an HDAC6 inhibitor apparently reverses CIPN.
Collapse
Affiliation(s)
- Jixiang Zhang
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Jazzmine M Junigan
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Ronnie Trinh
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Peter M Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
14
|
Vivianne Tawfik, M.D., Ph.D., a Recipient of the 2022 James E. Cottrell, M.D., Presidential Scholar Award. Anesthesiology 2022. [DOI: 10.1097/aln.0000000000004360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
15
|
Jin D, Chen H, Huang Y, Chen SR, Pan HL. δ-Opioid receptors in primary sensory neurons tonically restrain nociceptive input in chronic pain but do not enhance morphine analgesic tolerance. Neuropharmacology 2022; 217:109202. [PMID: 35917874 DOI: 10.1016/j.neuropharm.2022.109202] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/10/2022] [Accepted: 07/18/2022] [Indexed: 11/26/2022]
Abstract
δ-Opioid receptors (DORs, encoded by the Oprd1 gene) are expressed throughout the peripheral and central nervous system, and DOR stimulation reduces nociception. Previous studies suggest that DORs promote the development of analgesic tolerance of μ-opioid receptor (MOR) agonists. It is uncertain whether DORs expressed in primary sensory neurons are involved in regulating chronic pain and MOR agonist-induced tolerance. In this study, we generated Oprd1 conditional knockout (Oprd1-cKO) mice by crossing Advillin-Cre mice with Oprd1-floxed mice. DOR expression in the dorsal root ganglion was diminished in Oprd1-cKO mice. Systemic or intrathecal injection of the DOR agonist SNC-80 produced analgesia in wild-type (WT), but not Oprd1-cKO, mice. In contrast, intracerebroventricular injection of SNC-80 produced a similar analgesic effect in WT and Oprd1-cKO mice. However, morphine-induced analgesia, hyperalgesia, or analgesic tolerance did not differ between WT and Oprd1-cKO mice. Compared with WT mice, Oprd1-cKO mice showed increased mechanical and heat hypersensitivity after nerve injury or tissue inflammation. Furthermore, blocking DORs with naltrindole increased nociceptive sensitivity induced by nerve injury or tissue inflammation in WT, but not Oprd1-cKO, mice. In addition, naltrindole potentiated glutamatergic input from primary afferents to spinal dorsal horn neurons increased by nerve injury or CFA in WT mice; this effect was absent in Oprd1-cKO mice. Our findings indicate that DORs in primary sensory neurons are critically involved in the analgesic effect of DOR agonists but not morphine-induced analgesic tolerance. Presynaptic DORs at primary afferent central terminals constitutively inhibit inflammatory and neuropathic pain by restraining glutamatergic input to spinal dorsal horn neurons.
Collapse
Affiliation(s)
- Daozhong Jin
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hong Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yuying Huang
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Pre-Synaptic GABAA in NaV1.8+ Primary Afferents Is Required for the Development of Punctate but Not Dynamic Mechanical Allodynia following CFA Inflammation. Cells 2022; 11:cells11152390. [PMID: 35954234 PMCID: PMC9368720 DOI: 10.3390/cells11152390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/27/2023] Open
Abstract
Hypersensitivity to mechanical stimuli is a cardinal symptom of neuropathic and inflammatory pain. A reduction in spinal inhibition is generally considered a causal factor in the development of mechanical hypersensitivity after injury. However, the extent to which presynaptic inhibition contributes to altered spinal inhibition is less well established. Here, we used conditional deletion of GABAA in NaV1.8-positive sensory neurons (Scn10aCre;Gabrb3fl/fl) to manipulate selectively presynaptic GABAergic inhibition. Behavioral testing showed that the development of inflammatory punctate allodynia was mitigated in mice lacking pre-synaptic GABAA. Dorsal horn cellular circuits were visualized in single slices using stimulus-tractable dual-labelling of c-fos mRNA for punctate and the cognate c-Fos protein for dynamic mechanical stimulation. This revealed a substantial reduction in the number of cells activated by punctate stimulation in mice lacking presynaptic GABAA and an approximate 50% overlap of the punctate with the dynamic circuit, the relative percentage of which did not change following inflammation. The reduction in dorsal horn cells activated by punctate stimuli was equally prevalent in parvalbumin- and calretinin-positive cells and across all laminae I–V, indicating a generalized reduction in spinal input. In peripheral DRG neurons, inflammation following complete Freund’s adjuvant (CFA) led to an increase in axonal excitability responses to GABA, suggesting that presynaptic GABA effects in NaV1.8+ afferents switch from inhibition to excitation after CFA. In the days after inflammation, presynaptic GABAA in NaV1.8+ nociceptors constitutes an “open gate” pathway allowing mechanoreceptors responding to punctate mechanical stimulation access to nociceptive dorsal horn circuits.
Collapse
|
17
|
Bertels Z, Dripps IJ, Shah P, Moye LS, Tipton AF, Siegersma K, Pradhan AA. Delta opioid receptors in Nav1.8 expressing peripheral neurons partially regulate the effect of delta agonist in models of migraine and opioid-induced hyperalgesia. NEUROBIOLOGY OF PAIN 2022; 12:100099. [PMID: 35859654 PMCID: PMC9289726 DOI: 10.1016/j.ynpai.2022.100099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/07/2022]
Abstract
DOR in Nav1.8 cells do not regulate anti-migraine effects of DOR agonist. DOR in Nav1.8 cells is critical for effect of DOR agonist in peripheral OIH. DOR in Nav1.8 cells is not necessary for effect of DOR agonist in cephalic OIH.
Migraine is one of the most common pain disorders and causes disability in millions of people every year. Delta opioid receptors (DOR) have been identified as a novel therapeutic target for migraine and other headache disorders. DORs are present in both peripheral and central regions and it is unclear which receptor populations regulate migraine-associated effects. The aim of this study was to determine if DOR expressed in peripheral nociceptors regulates headache associated endpoints and the effect of delta agonists within these mouse models. We used a conditional knockout, in which DOR was selectively deleted from Nav1.8 expressing cells. Nav1.8-DOR mice and loxP control littermates were tested in models of chronic migraine-associated allodynia, opioid-induced hyperalgesia, migraine-associated negative affect, and aura. Nav1.8-DOR and loxP mice had comparable effect sizes in all of these models. The anti-allodynic effect of the DOR agonist, SNC80, was slightly diminished in the nitroglycerin model of migraine. Intriguingly, in the OIH model the peripheral effects of SNC80 were completely lost in Nav1.8-DOR mice while the cephalic effects remained intact. Regardless of genotype, SNC80 continued to inhibit conditioned place aversion associated with nitroglycerin and decreased cortical spreading depression events associated with migraine aura. These results suggest that DOR in Nav1.8-expressing nociceptors do not critically regulate the anti-migraine effects of delta agonist; and that brain-penetrant delta agonists would be a more effective drug development strategy.
Collapse
|
18
|
Ma W, Sapio MR, Manalo AP, Maric D, Dougherty MK, Goto T, Mannes AJ, Iadarola MJ. Anatomical Analysis of Transient Potential Vanilloid Receptor 1 (Trpv1+) and Mu-Opioid Receptor (Oprm1+) Co-expression in Rat Dorsal Root Ganglion Neurons. Front Mol Neurosci 2022; 15:926596. [PMID: 35875671 PMCID: PMC9302591 DOI: 10.3389/fnmol.2022.926596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/09/2022] [Indexed: 11/29/2022] Open
Abstract
Primary afferent neurons of the dorsal root ganglia (DRG) transduce peripheral nociceptive signals and transmit them to the spinal cord. These neurons also mediate analgesic control of the nociceptive inputs, particularly through the μ-opioid receptor (encoded by Oprm1). While opioid receptors are found throughout the neuraxis and in the spinal cord tissue itself, intrathecal administration of μ-opioid agonists also acts directly on nociceptive nerve terminals in the dorsal spinal cord resulting in marked analgesia. Additionally, selective chemoaxotomy of cells expressing the TRPV1 channel, a nonselective calcium-permeable ion channel that transduces thermal and inflammatory pain, yields profound pain relief in rats, canines, and humans. However, the relationship between Oprm1 and Trpv1 expressing DRG neurons has not been precisely determined. The present study examines rat DRG neurons using high resolution multiplex fluorescent in situ hybridization to visualize molecular co-expression. Neurons positive for Trpv1 exhibited varying levels of expression for Trpv1 and co-expression of other excitatory and inhibitory ion channels or receptors. A subpopulation of densely labeled Trpv1+ neurons did not co-express Oprm1. In contrast, a population of less densely labeled Trpv1+ neurons did co-express Oprm1. This finding suggests that the medium/low Trpv1 expressing neurons represent a specific set of DRG neurons subserving the opponent processes of both transducing and inhibiting nociceptive inputs. Additionally, the medium/low Trpv1 expressing neurons co-expressed other markers implicated in pathological pain states, such as Trpa1 and Trpm8, which are involved in chemical nociception and cold allodynia, respectively, as well as Scn11a, whose mutations are implicated in familial episodic pain. Conversely, none of the Trpv1+ neurons co-expressed Spp1, which codes for osteopontin, a marker for large diameter proprioceptive neurons, validating that nociception and proprioception are governed by separate neuronal populations. Our findings support the hypothesis that the population of Trpv1 and Oprm1 coexpressing neurons may explain the remarkable efficacy of opioid drugs administered at the level of the DRG-spinal synapse, and that this subpopulation of Trpv1+ neurons is responsible for registering tissue damage.
Collapse
Affiliation(s)
- Wenting Ma
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Matthew R. Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Allison P. Manalo
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD, United States
| | - Mary Kate Dougherty
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Taichi Goto
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
- Symptoms Biology Unit, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Andrew J. Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Michael J. Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Michael J. Iadarola
| |
Collapse
|
19
|
Cahill CM, Holdridge SV, Liu S, Xue L, Magnussen C, Ong E, Grenier P, Sutherland A, Olmstead MC. Delta opioid receptor activation modulates affective pain and modality-specific pain hypersensitivity associated with chronic neuropathic pain. J Neurosci Res 2022; 100:129-148. [PMID: 32623788 PMCID: PMC8218601 DOI: 10.1002/jnr.24680] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/28/2020] [Accepted: 06/04/2020] [Indexed: 01/03/2023]
Abstract
Delta opioid receptor (DOR) agonists alleviate nociceptive behaviors in various chronic pain models, including neuropathic pain, while having minimal effect on sensory thresholds in the absence of injury. The mechanisms underlying nerve injury-induced enhancement of DOR function are unclear. We used a peripheral nerve injury (PNI) model of neuropathic pain to assess changes in the function and localization of DORs in mice and rats. Intrathecal administration of DOR agonists reversed mechanical allodynia and thermal hyperalgesia. The dose-dependent thermal antinociceptive effects of DOR agonists were shifted to the left in PNI rats. Administration of DOR agonists produced a conditioned place preference in PNI, but not in sham, animals, whereas the DOR antagonist naltrindole produced a place aversion in PNI, but not in sham, mice, suggesting the engagement of endogenous DOR activity in suppressing pain associated with the injury. GTPγS autoradiography revealed an increase in DOR function in the dorsal spinal cord, ipsilateral to PNI. Immunogold electron microscopy and in vivo fluorescent agonist assays were used to assess changes in the ultrastructural localization of DORs in the spinal dorsal horn. In shams, DORs were primarily localized within intracellular compartments. PNI significantly increased the cell surface expression of DORs within lamina IV-V dendritic profiles. Using neonatal capsaicin treatment, we identified that DOR agonist-induced thermal antinociception was mediated via receptors expressed on primary afferent sensory neurons but did not alter mechanical thresholds. These data reveal that the regulation of DORs following PNI and suggest the importance of endogenous activation of DORs in regulating chronic pain states.
Collapse
Affiliation(s)
- Catherine M. Cahill
- Dept of Psychiatry & Biobehavioral Sciences, Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA, 90095
| | - Sarah V. Holdridge
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Steve Liu
- Dept of Psychiatry & Biobehavioral Sciences, Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA, 90095,Department of Psychology and Centre for Neuroscience Studies, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Lihua Xue
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Claire Magnussen
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Edmund Ong
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Patrick Grenier
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Anne Sutherland
- Dept of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| | - Mary C. Olmstead
- Department of Psychology and Centre for Neuroscience Studies, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
| |
Collapse
|
20
|
Martin LF, Moutal A, Cheng K, Washington SM, Calligaro H, Goel V, Kranz T, Largent-Milnes TM, Khanna R, Patwardhan A, Ibrahim MM. Green Light Antinociceptive and Reversal of Thermal and Mechanical Hypersensitivity Effects Rely on Endogenous Opioid System Stimulation. THE JOURNAL OF PAIN 2021; 22:1646-1656. [PMID: 34157406 PMCID: PMC8664962 DOI: 10.1016/j.jpain.2021.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/12/2021] [Accepted: 05/23/2021] [Indexed: 12/17/2022]
Abstract
Benefits of phototherapy were characterized in multiple diseases including depression, circadian rhythm disruptions, and neurodegeneration. Studies on migraine and fibromyalgia patients revealed that green light-emitting diodes (GLED) exposure provides a pragmatic and safe therapy to manage chronic pain. In rodents, GLED reversed hypersensitivity related to neuropathic pain. However, little is known about the underlying mechanisms of GLED efficacy. Here, we sought to understand how green light modulates the endogenous opioid system. We first characterized how exposure to GLED stimulates release of β-endorphin and proenkephalin in the central nervous system of male rats. Moreover, by individually editing each of the receptors, we found that µ- and δ-opioid receptors are required for green light's antinociceptive effect in naïve rats and a model of HIV-induced peripheral neuropathy. We investigated how GLED could increase pain thresholds, and explored its potential in reversing hypersensitivity in a model of HIV-related neuropathy. Through behavioral and gene editing approaches, we identified that green light provides antinociception via modulation of the endogenous opioid system in the spinal cord. This work identifies a previously unknown mechanism by which GLED can improve pain management. Clinical translation of these results will advance the development of an innovative therapy devoid of adverse effects. PERSPECTIVE: Development of new pain management therapies, especially for HIV patients, is crucial as long-term opioid prescription is not recommended due to adverse side effects. Green light addresses this necessity. Characterizing the underlying mechanisms of this potentially groundbreaking and safe antinociceptive therapy will advance its clinical translation.
Collapse
Affiliation(s)
- Laurent F Martin
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona; Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Kevin Cheng
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Stephanie M Washington
- Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Hugo Calligaro
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, San Diego, California
| | - Vasudha Goel
- Department of Anesthesiology, The University of Minnesota Medical School, Minneapolis, Minnesota
| | - Tracy Kranz
- Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Tally M Largent-Milnes
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona; Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, Arizona; Neuroscience Graduate Interdisciplinary Program, College of Medicine, The University of Arizona, Tucson, Arizona; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona; Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, Arizona
| | - Amol Patwardhan
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona; Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, Arizona; Department of Neurosurgery, College of Medicine, The University of Arizona, Tucson, Arizona; Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, Arizona
| | - Mohab M Ibrahim
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona; Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, Arizona; Department of Neurosurgery, College of Medicine, The University of Arizona, Tucson, Arizona; Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, Arizona.
| |
Collapse
|
21
|
Komiya E, Tominaga M, Hatano R, Kamikubo Y, Toyama S, Sakairi H, Honda K, Itoh T, Kamata Y, Tsurumachi M, Kishi R, Ohnuma K, Sakurai T, Morimoto C, Takamori K. Peripheral endomorphins drive mechanical alloknesis under the enzymatic control of CD26/DPPIV. J Allergy Clin Immunol 2021; 149:1085-1096. [PMID: 34411589 DOI: 10.1016/j.jaci.2021.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 06/29/2021] [Accepted: 08/02/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Mechanical alloknesis (or innocuous mechanical stimuli-evoked itch) often occurs in dry skin-based disorders such as atopic dermatitis and psoriasis. However, the molecular and cellular mechanisms underlying mechanical alloknesis remain unclear. We recently reported the involvement of CD26 in the regulation of psoriatic itch. This molecule exhibits dipeptidyl peptidase IV (DPPIV) enzyme activity and exerts its biologic effects by processing various substances, including neuropeptides. OBJECTIVE The aim of the present study was to investigate the peripheral mechanisms of mechanical alloknesis by using CD26/DPPIV knockout (CD26KO) mice. METHODS We applied innocuous mechanical stimuli to CD26KO or wild-type mice. The total number of scratching responses was counted as the alloknesis score. Immunohistochemical and behavioral pharmacologic analyses were then performed to examine the physiologic activities of CD26/DPPIV or endomorphins (EMs), endogenous agonists of μ-opioid receptors. RESULTS Mechanical alloknesis was more frequent in CD26KO mice than in wild-type mice. The alloknesis score in CD26KO mice was significantly reduced by the intradermal administration of recombinant DPPIV or naloxone methiodide, a peripheral μ-opioid receptor antagonist, but not by that of mutant DPPIV without enzyme activity. EMs (EM-1 and EM-2), selective ligands for μ-opioid receptors, are substrates for DPPIV. Immunohistochemically, EMs were located in keratinocytes, fibroblasts, and peripheral sensory nerves. Behavioral analyses revealed that EMs preferentially provoked mechanical alloknesis over chemical itch. DPPIV-digested forms of EMs did not induce mechanical alloknesis. CONCLUSION The present results suggest that EMs induce mechanical alloknesis at the periphery under the enzymatic control of CD26/DPPIV.
Collapse
Affiliation(s)
- Eriko Komiya
- Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan
| | - Mitsutoshi Tominaga
- Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan; Anti-Aging Skin Research Laboratory, Graduate School of Medicine, Juntendo University, Chiba, Japan
| | - Ryo Hatano
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yuji Kamikubo
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, Japan
| | - Sumika Toyama
- Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan
| | - Hakushun Sakairi
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kotaro Honda
- Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan
| | - Takumi Itoh
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo, Japan; Atopy (Allergy) Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yayoi Kamata
- Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan; Anti-Aging Skin Research Laboratory, Graduate School of Medicine, Juntendo University, Chiba, Japan
| | - Munehiro Tsurumachi
- Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan; Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Ryoma Kishi
- Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan; Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Kei Ohnuma
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takashi Sakurai
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, Japan
| | - Chikao Morimoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kenji Takamori
- Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, Chiba, Japan; Anti-Aging Skin Research Laboratory, Graduate School of Medicine, Juntendo University, Chiba, Japan; Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan.
| |
Collapse
|
22
|
Lukoyanov N, Watanabe H, Carvalho LS, Kononenko O, Sarkisyan D, Zhang M, Andersen MS, Lukoyanova EA, Galatenko V, Tonevitsky A, Bazov I, Iakovleva T, Schouenborg J, Bakalkin G. Left-right side-specific endocrine signaling complements neural pathways to mediate acute asymmetric effects of brain injury. eLife 2021; 10:e65247. [PMID: 34372969 PMCID: PMC8354641 DOI: 10.7554/elife.65247] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 07/07/2021] [Indexed: 12/14/2022] Open
Abstract
Brain injuries can interrupt descending neural pathways that convey motor commands from the cortex to spinal motoneurons. Here, we demonstrate that a unilateral injury of the hindlimb sensorimotor cortex of rats with completely transected thoracic spinal cord produces hindlimb postural asymmetry with contralateral flexion and asymmetric hindlimb withdrawal reflexes within 3 hr, as well as asymmetry in gene expression patterns in the lumbar spinal cord. The injury-induced postural effects were abolished by hypophysectomy and were mimicked by transfusion of serum from animals with brain injury. Administration of the pituitary neurohormones β-endorphin or Arg-vasopressin-induced side-specific hindlimb responses in naive animals, while antagonists of the opioid and vasopressin receptors blocked hindlimb postural asymmetry in rats with brain injury. Thus, in addition to the well-established involvement of motor pathways descending from the brain to spinal circuits, the side-specific humoral signaling may also add to postural and reflex asymmetries seen after brain injury.
Collapse
Affiliation(s)
- Nikolay Lukoyanov
- Departamento de Biomedicina da Faculdade de Medicina da Universidade do Porto, Instituto de Investigação e Inovação em Saúde, Instituto de Biologia Molecular e CelularPortoPortugal
| | - Hiroyuki Watanabe
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Liliana S Carvalho
- Departamento de Biomedicina da Faculdade de Medicina da Universidade do Porto, Instituto de Investigação e Inovação em Saúde, Instituto de Biologia Molecular e CelularPortoPortugal
| | - Olga Kononenko
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Daniil Sarkisyan
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Mengliang Zhang
- Neuronano Research Center, Department of Experimental Medical Science, Lund UniversityLundSweden
- Department of Molecular Medicine, University of Southern DenmarkOdenseDenmark
| | | | - Elena A Lukoyanova
- Departamento de Biomedicina da Faculdade de Medicina da Universidade do Porto, Instituto de Investigação e Inovação em Saúde, Instituto de Biologia Molecular e CelularPortoPortugal
| | - Vladimir Galatenko
- Faculty of Mechanics and Mathematics, Lomonosov Moscow State UniversityMoscowRussian Federation
| | - Alex Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of EconomicsMoscowRussian Federation
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry RASMoscowRussian Federation
| | - Igor Bazov
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Tatiana Iakovleva
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Jens Schouenborg
- Neuronano Research Center, Department of Experimental Medical Science, Lund UniversityLundSweden
| | - Georgy Bakalkin
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| |
Collapse
|
23
|
Pollard KJ, Bowser DA, Anderson WA, Meselhe M, Moore MJ. Morphine-sensitive synaptic transmission emerges in embryonic rat microphysiological model of lower afferent nociceptive signaling. SCIENCE ADVANCES 2021; 7:7/35/eabj2899. [PMID: 34452921 PMCID: PMC8397270 DOI: 10.1126/sciadv.abj2899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/08/2021] [Indexed: 05/12/2023]
Abstract
Debilitating chronic pain resulting from genetic predisposition, injury, or acquired neuropathy is becoming increasingly pervasive. Opioid analgesics remain the gold standard for intractable pain, but overprescription of increasingly powerful and addictive opioids has contributed to the current prescription drug abuse epidemic. There is a pressing need to screen experimental compounds more efficiently for analgesic potential that remains unmet by conventional research models. The spinal cord dorsal horn is a common target for analgesic intervention, where peripheral nociceptive signals are relayed to the central nervous system through synaptic transmission. Here, we demonstrate that coculturing peripheral and dorsal spinal cord nerve cells in a novel bioengineered microphysiological system facilitates self-directed emergence of native nerve tissue macrostructure and concerted synaptic function. The mechanistically distinct analgesics-morphine, lidocaine, and clonidine-differentially and predictably modulate this microphysiological synaptic transmission. Screening drug candidates for similar microphysiological profiles will efficiently identify therapeutics with analgesic potential.
Collapse
Affiliation(s)
- Kevin J Pollard
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
| | - Devon A Bowser
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
- Bioinnovation Program, Tulane University, New Orleans, LA 70118, USA
| | - Wesley A Anderson
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
- AxoSim Inc., New Orleans, LA 70112, USA
| | - Mostafa Meselhe
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Michael J Moore
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA.
- AxoSim Inc., New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
24
|
Moye LS, Siegersma K, Dripps I, Witkowski W, Mangutov E, Wang D, Scherrer G, Pradhan AA. Delta opioid receptor regulation of calcitonin gene-related peptide dynamics in the trigeminal complex. Pain 2021; 162:2297-2308. [PMID: 33605657 PMCID: PMC8730473 DOI: 10.1097/j.pain.0000000000002235] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022]
Abstract
ABSTRACT Migraine is highly prevalent and is the sixth leading cause worldwide for years lost to disability. Therapeutic options specifically targeting migraine are limited, and delta opioid receptor (DOP) agonists were recently identified as a promising pharmacotherapy. The mechanisms by which DOPs regulate migraine are currently unclear. Calcitonin gene-related peptide (CGRP) has been identified as an endogenous migraine trigger and plays a critical role in migraine initiation and susceptibility. The aim of this study was to determine the behavioral effects of DOP agonists on the development of chronic migraine-associated pain and to investigate DOP coexpression with CGRP and CGRP receptor (CGRPR) in the trigeminal system. Chronic migraine-associated pain was induced in mice through repeated intermittent injection of the known human migraine trigger, nitroglycerin. Chronic nitroglycerin resulted in severe chronic cephalic allodynia which was prevented with cotreatment of the DOP-selective agonist, SNC80. In addition, a corresponding increase in CGRP expression in the trigeminal ganglia and trigeminal nucleus caudalis was observed after chronic nitroglycerin, an augmentation that was blocked by SNC80. Moreover, DOP was also upregulated in these head pain-processing regions following the chronic migraine model. Immunohistochemical analysis of the trigeminal ganglia revealed coexpression of DOP with CGRP as well as with a primary component of the CGRPR, RAMP1. In the trigeminal nucleus caudalis, DOP was not coexpressed with CGRP but was highly coexpressed with RAMP1 and calcitonin receptor-like receptor. These results suggest that DOP agonists inhibit migraine-associated pain by attenuating CGRP release and blocking pronociceptive signaling of the CGRPR.
Collapse
Affiliation(s)
- Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago
| | | | - Isaac Dripps
- Department of Psychiatry, University of Illinois at Chicago
| | | | | | - Dong Wang
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Neurosurgery, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- New York Stem Cell Foundation – Robertson Investigator
| | | |
Collapse
|
25
|
Toll L, Cippitelli A, Ozawa A. The NOP Receptor System in Neurological and Psychiatric Disorders: Discrepancies, Peculiarities and Clinical Progress in Developing Targeted Therapies. CNS Drugs 2021; 35:591-607. [PMID: 34057709 PMCID: PMC8279133 DOI: 10.1007/s40263-021-00821-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/28/2021] [Indexed: 02/01/2023]
Abstract
The nociceptin opioid peptide (NOP) receptor and its endogenous ligand nociceptin/orphanin FQ (N/OFQ) are the fourth members of the opioid receptor and opioid peptide families. Although they have considerable sequence homology to the other family members, they are not considered opioid per se because they do not have pharmacological profiles similar to the other family members. The number of NOP receptors in the brain is higher than the other family members, and NOP receptors can be found throughout the brain. Because of the widespread distribution of NOP receptors, N/OFQ and other peptide and small molecule agonists and antagonists have extensive CNS activities. Originally thought to be anti-opioid, NOP receptor agonists block some opioid activities, potentiate others, and modulate other activities not affected by traditional opiates. Because the effect of receptor activation can be dependent upon site of administration, state of the animal, and other variables, the study of NOP receptors has been fraught with contradictions and inconsistencies. In this article, the actions and controversies pertaining to NOP receptor activation and inhibition are discussed with respect to CNS disorders including pain (acute, chronic, and migraine), drug abuse, anxiety and depression. In addition, progress towards clinical use of NOP receptor-directed compounds is discussed.
Collapse
Affiliation(s)
- Lawrence Toll
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, USA.
| | - Andrea Cippitelli
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, USA
| | - Akihiko Ozawa
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, USA
| |
Collapse
|
26
|
Functional expression of glycine receptors in DRG neurons of mice. Eur J Pharmacol 2021; 899:174034. [PMID: 33727056 DOI: 10.1016/j.ejphar.2021.174034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/25/2021] [Accepted: 03/11/2021] [Indexed: 11/20/2022]
Abstract
Glycine receptor is one of the chloride-permeable ion channels composed of combinations of four α subunits and one β subunit. In adult spinal cord, the glycine receptor α1 subunit is crucial for the generation of inhibitory neurotransmission. The reduced glycinergic inhibition is regarded as one of the key spinal mechanisms underlying pathological pain symptoms. However, the expression and function of glycine receptors in the peripheral system are largely unknown as yet. Here we found that glycine receptor α1 subunit was prevalent in the dorsal root ganglia (DRG) neurons as well as in the sciatic nerves of adult mice. Intraganglionar or intraplantar injection of glycine receptor antagonist strychnine caused the hypersensitivity to mechanical, thermal and cold stimuli, suggesting the functional importance of peripheral glycine receptors in the control of nociceptive signal transmission. Our data showed that peripheral inflammation induced by formalin decreased the expression of glycine receptor α1 subunit on the plasma membrane of DRG neurons, which was attributed to the activation of protein kinase C signaling. Intraplantar application of glycine receptor agonist glycine or positive modulator divalent zinc ion alleviated the first-phase painful behaviors induced by formalin. These data suggested that peripheral glycine receptor might serve as an effective target for pain therapy.
Collapse
|
27
|
Chapman KB, Yousef TA, Vissers KC, van Helmond N, D Stanton-Hicks M. Very Low Frequencies Maintain Pain Relief From Dorsal Root Ganglion Stimulation: An Evaluation of Dorsal Root Ganglion Neurostimulation Frequency Tapering. Neuromodulation 2020; 24:746-752. [PMID: 33227827 DOI: 10.1111/ner.13322] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/09/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Dorsal root ganglion neurostimulation (DRG-S) is effective in treating various refractory chronic pain syndromes. In preclinical studies, DRG-S at very low frequencies (<5 Hz) reduces excitatory output in the superficial dorsal horn. Clinically, we have also observed the effectiveness of DRG-S at low frequencies. We conducted a case series to describe the effect of very low-frequency DRG-S stimulation on clinical outcomes. MATERIALS AND METHODS DRG-S for refractory low back pain was initiated at parameters consistent with published values. Thereafter, the stimulation frequency of DRG-S was reduced in a stepwise fashion to the lowest frequency that maintained pain relief. Pain intensity, disability, and general health status data were collected at baseline, prior to initiation of tapering, and at four weeks after each patient's lowest effective stimulation frequency was reached. RESULTS After device activation (N = 20), DRG-S frequency was tapered from 16 to 4 Hz over a 4- to 17-week period, reducing charge-per-second by nearly two-thirds. Even so, pain relief was maintained at more than 75%, with consistent findings in the other measures. CONCLUSION DRG-S may have utility in treating chronic pain at lower stimulation frequencies than previously recognized. We have previously theorized that the mechanism of action may involve preferential recruitment of low-threshold mechanoreceptor fibers via the endogenous opioid system. Of clinical relevance, lower frequency stimulation maintains DRG-S efficacy regarding improvements in pain, disability, and quality of life. It can extend battery life and may potentially lead to the development of smaller implantable pulse generators.
Collapse
Affiliation(s)
- Kenneth B Chapman
- The Spine & Pain Institute of New York, New York City, NY, USA.,Department of Anesthesiology, New York University Langone Medical Center, New York City, NY, USA.,Department of Anesthesiology, Zucker School of Medicine at Hofstra Northwell, Northwell Health, Manhasset, NY, USA
| | - Tariq A Yousef
- The Spine & Pain Institute of New York, New York City, NY, USA
| | - Kris C Vissers
- Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University, Nijmegen, The Netherlands
| | - Noud van Helmond
- The Spine & Pain Institute of New York, New York City, NY, USA.,Department of Anesthesiology, Cooper Medical School of Rowan University, Cooper University Hospital, Camden, NJ, USA
| | | |
Collapse
|
28
|
Peripheral Mechanobiology of Touch-Studies on Vertebrate Cutaneous Sensory Corpuscles. Int J Mol Sci 2020; 21:ijms21176221. [PMID: 32867400 PMCID: PMC7504094 DOI: 10.3390/ijms21176221] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
The vertebrate skin contains sensory corpuscles that are receptors for different qualities of mechanosensitivity like light brush, touch, pressure, stretch or vibration. These specialized sensory organs are linked anatomically and functionally to mechanosensory neurons, which function as low-threshold mechanoreceptors connected to peripheral skin through Aβ nerve fibers. Furthermore, low-threshold mechanoreceptors associated with Aδ and C nerve fibers have been identified in hairy skin. The process of mechanotransduction requires the conversion of a mechanical stimulus into electrical signals (action potentials) through the activation of mechanosensible ion channels present both in the axon and the periaxonal cells of sensory corpuscles (i.e., Schwann-, endoneurial- and perineurial-related cells). Most of those putative ion channels belong to the degenerin/epithelial sodium channel (especially the family of acid-sensing ion channels), the transient receptor potential channel superfamilies, and the Piezo family. This review updates the current data about the occurrence and distribution of putative mechanosensitive ion channels in cutaneous mechanoreceptors including primary sensory neurons and sensory corpuscles.
Collapse
|
29
|
Uhelski ML, Bruce D, Speltz R, Wilcox GL, Simone DA. Topical Application of Loperamide/Oxymorphindole, Mu and Delta Opioid Receptor Agonists, Reduces Sensitization of C-fiber Nociceptors that Possess Na V1.8. Neuroscience 2020; 446:102-112. [PMID: 32858141 DOI: 10.1016/j.neuroscience.2020.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 06/27/2020] [Accepted: 08/17/2020] [Indexed: 01/09/2023]
Abstract
It was recently shown that local injection, systemic administration or topical application of the peripherally-restricted mu-opioid receptor (MOR) agonist loperamide (Lo) and the delta-opioid receptor (DOR) agonist oxymorphindole (OMI) synergized to produce highly potent anti-hyperalgesia that was dependent on both MOR and DOR located in the periphery. We assessed peripheral mechanisms by which this Lo/OMI combination produces analgesia in mice expressing the light-sensitive protein channelrhodopsin2 (ChR2) in neurons that express NaV1.8 voltage-gated sodium channels. These mice (NaV1.8-ChR2+) enabled us to selectively target and record electrophysiological activity from these neurons (the majority of which are nociceptive) using blue light stimulation of the hind paw. We assessed the effect of Lo/OMI on nociceptor activity in both naïve mice and mice treated with complete Freund's adjuvant (CFA) to induce chronic inflammation of the hind paw. Teased fiber recording of tibial nerve fibers innervating the plantar hind paw revealed that the Lo/OMI combination reduced responses to light stimulation in naïve mice and attenuated spontaneous activity (SA) as well as responses to light and mechanical stimuli in CFA-treated mice. These results show that Lo/OMI reduces activity of C-fiber nociceptors that express NaV1.8 and corroborate recent behavioral studies demonstrating the potent analgesic effects of this drug combination. Because of its peripheral site of action, Lo/OMI might produce effective analgesia without the side effects associated with activation of opioid receptors in the central nervous system.
Collapse
Affiliation(s)
- Megan L Uhelski
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel Bruce
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rebecca Speltz
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - George L Wilcox
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Dermatology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Donald A Simone
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
30
|
Bertels Z, Pradhan AAA. Emerging Treatment Targets for Migraine and Other Headaches. Headache 2020; 59 Suppl 2:50-65. [PMID: 31291018 DOI: 10.1111/head.13585] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
Migraine is a complex disorder that is characterized by an assortment of neurological and systemic effects. While headache is the most prominent feature of migraine, a host of symptoms affecting many physiological functions are also observed before, during, and after an attack. Furthermore, migraineurs are heterogeneous and have a wide range of responses to migraine therapies. The recent approval of calcitonin gene-related-peptide based therapies has opened up the treatment of migraine and generated a renewed interest in migraine research and discovery. Ongoing advances in migraine research have identified a number of other promising therapeutic targets for this disorder. In this review, we highlight emergent treatments within the following biological systems: pituitary adenylate cyclase activating peptdie, 2 non-mu opioid receptors that have low abuse liability - the delta and kappa opioid receptors, orexin, and nitric oxide-based therapies. Multiple mechanisms have been identified in the induction and maintenance of migraine symptoms; and this divergent set of targets have highly distinct biological effects. Increasing the mechanistic diversity of the migraine tool box will lead to more treatment options and better patient care.
Collapse
Affiliation(s)
- Zachariah Bertels
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | | |
Collapse
|
31
|
Lee YR, Trung TQ, Hwang BU, Lee NE. A flexible artificial intrinsic-synaptic tactile sensory organ. Nat Commun 2020; 11:2753. [PMID: 32488078 PMCID: PMC7265430 DOI: 10.1038/s41467-020-16606-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 05/11/2020] [Indexed: 01/04/2023] Open
Abstract
Imbuing bio-inspired sensory devices with intelligent functions of human sensory organs has been limited by challenges in emulating the preprocessing abilities of sensory organs such as reception, filtering, adaptation, and sensory memory at the device level itself. Merkel cells, which is a part of tactile sensory organs, form synapse-like connections with afferent neuron terminals referred to as Merkel cell-neurite complexes. Here, inspired by structure and intelligent functions of Merkel cell-neurite complexes, we report a flexible, artificial, intrinsic-synaptic tactile sensory organ that mimics synapse-like connections using an organic synaptic transistor with ferroelectric nanocomposite gate dielectric of barium titanate nanoparticles and poly(vinylidene fluoride-trifluoroethylene). Modulation of the post-synaptic current of the device induced by ferroelectric dipole switching due to triboelectric-capacitive coupling under finger touch allowed reception and slow adaptation. Modulation of synaptic weight by varying the nanocomposite composition of gate dielectric layer enabled tuning of filtering and sensory memory functions.
Collapse
Affiliation(s)
- Yu Rim Lee
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyunggi-do, 16419, Korea
| | - Tran Quang Trung
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyunggi-do, 16419, Korea
| | - Byeong-Ung Hwang
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyunggi-do, 16419, Korea
| | - Nae-Eung Lee
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyunggi-do, 16419, Korea.
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyunggi-do, 16419, Korea.
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyunggi-do, 16419, Korea.
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyunggi-do, 16419, Korea.
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyunggi-do, 16419, Korea.
| |
Collapse
|
32
|
Chapman KB, Groenen PS, Vissers KC, van Helmond N, Stanton-Hicks MD. The Pathways and Processes Underlying Spinal Transmission of Low Back Pain: Observations From Dorsal Root Ganglion Stimulation Treatment. Neuromodulation 2020; 24:610-621. [PMID: 32329155 DOI: 10.1111/ner.13150] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/02/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Dorsal root ganglion stimulation (DRG-S) is a novel approach to treat chronic pain. Lead placement at L2 has been reported to be an effective treatment for axial low back pain (LBP) primarily of discogenic etiology. We have recently shown, in a diverse cohort including cases of multilevel instrumentation following extensive prior back surgeries, that DRG-S lead placement at T12 is another promising target. Local effects at the T12 DRG, alone, are insufficient to explain these results. MATERIALS AND METHODS We performed a literature review to explore the mechanisms of LBP relief with T12 DRG-S. FINDINGS Branches of individual spinal nerve roots innervate facet joints and posterior spinal structures, while the discs and anterior vertebrae are carried via L2, and converge in the dorsal horn (DH) of the spinal cord at T8-T9. The T12 nerve root contains cutaneous afferents from the low back and enters the DH of the spinal cord at T10. Low back Aδ and C-fibers then ascend via Lissauer's tract (LT) to T8-T9, converging with other low back afferents. DRG-S at T12, then, results in inhibition of the converged low back fibers via endorphin-mediated and GABAergic frequency-dependent mechanisms. Therefore, T12 lead placement may be the optimal location for DRG-S to treat LBP.
Collapse
Affiliation(s)
- Kenneth B Chapman
- The Spine & Pain Institute of New York, New York City, NY, USA.,Department of Anesthesiology, New York University Langone Medical Center, New York City, NY, USA.,Northwell Health Systems, New York City, NY, USA
| | - Pauline S Groenen
- The Spine & Pain Institute of New York, New York City, NY, USA.,College of Medicine, Radboud University, Nijmegen, the Netherlands
| | - Kris C Vissers
- Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University, Nijmegen, the Netherlands
| | - Noud van Helmond
- The Spine & Pain Institute of New York, New York City, NY, USA.,Department of Anesthesiology, Cooper Medical School of Rowan University, Cooper University Hospital, Camden, NJ, USA
| | | |
Collapse
|
33
|
Berthiaume S, Abdallah K, Blais V, Gendron L. Alleviating pain with delta opioid receptor agonists: evidence from experimental models. J Neural Transm (Vienna) 2020; 127:661-672. [PMID: 32189076 DOI: 10.1007/s00702-020-02172-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
The use of opioids for the relief of pain and headache disorders has been studied for years. Nowadays, particularly because of its ability to produce analgesia in various pain models, delta opioid receptor (DOPr) emerges as a promising target for the development of new pain therapies. Indeed, their potential to avoid the unwanted effects commonly observed with clinically used opioids acting at the mu opioid receptor (MOPr) suggests that DOPr agonists could be a therapeutic option. In this review, we discuss the use of opioids in the management of pain in addition to describing the evidence of the analgesic potency of DOPr agonists in animal models.
Collapse
Affiliation(s)
- Sophie Berthiaume
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
| | - Khaled Abdallah
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
| | - Véronique Blais
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
| | - Louis Gendron
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada.
| |
Collapse
|
34
|
Bai Y, Li MY, Ma JB, Li JN, Teng XY, Chen YB, Yin JB, Huang J, Chen J, Zhang T, Qiu XT, Chen T, Li H, Wu SX, Peng YN, Li X, Kou ZZ, Li YQ. Enkephalinergic Circuit Involved in Nociceptive Modulation in the Spinal Dorsal Horn. Neuroscience 2020; 429:78-91. [PMID: 31917345 DOI: 10.1016/j.neuroscience.2019.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 11/24/2022]
Abstract
Enkephalin (ENK) has been implicated in pain modulation within the spinal dorsal horn (SDH). Revealing the mechanisms underlying ENK analgesia entails the anatomical and functional knowledge of spinal ENK-ergic circuits. Herein, we combined morphological and electrophysiological studies to unravel local ENK-ergic circuitry within the SDH. First, the distribution pattern of spinal ENK-ergic neurons was observed in adult preproenkephalin (PPE)-GFP knock-in mice. Next, the retrograde tracer tetramethylrhodamine (TMR) or horseradish peroxidase (HRP) was injected into the parabrachial nucleus (PBN) in PPE-GFP mice. Immunofluorescent staining showed I-isolectin B4 (IB4) labeled non-peptidergic afferents were in close apposition to TMR-labeled PBN-projecting neurons within lamina I as well as PPE-immunoreactivity (-ir) neurons within lamina II. Some TMR-labeled neurons were simultaneously in close association with both IB4 and PPE-ir terminals. Synaptic connections of these components were further confirmed by electron microscopy. Finally, TMR was injected into the PBN in adult C57BL/6 mice. Whole-cell patch recordings showed that δ-opioid receptor (DOR) agonist, [D-Pen2,5]-enkephalin (DPDPE, 1 µM), significantly reduced the frequency of miniature excitatory postsynaptic current (mEPSC) and decreased the activity of TMR-labeled neurons. In conclusion, spinal ENKergic neurons receive direct excitatory inputs from primary afferents, which might be directly recruited to release ENK under the condition of noxious stimuli; ENK could inhibit the glutamatergic transmission towards projecting neurons via presynaptic and postsynaptic DORs. These morphological and functional evidence may explain the mechanisms underlying the analgesic effects exerted by ENK within the SDH.
Collapse
Affiliation(s)
- Yang Bai
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Meng-Ying Li
- Department of Endocrinology and Metabolism, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiang-Bo Ma
- Department of Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan, China
| | - Jia-Ni Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Xiao-Yu Teng
- Department of Anatomy, Guangxi Medical University, Nanning, China
| | - Ying-Biao Chen
- Department of Anatomy, Fujian Health College, Fuzhou, China
| | - Jun-Bin Yin
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Jing Huang
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Jing Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Ting Zhang
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Xin-Tong Qiu
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Tao Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Sheng-Xi Wu
- Department of Neurobiology, The Fourth Military Medical University, Xi'an, China
| | - Ya-Nan Peng
- Joint Laboratory of Neuroscience at Hainan Medical University and The Fourth Military Medical University, Hainan Medical University, Haikou, China
| | - Xiang Li
- Department of Orthopaedics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| | - Zhen-Zhen Kou
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China; Joint Laboratory of Neuroscience at Hainan Medical University and The Fourth Military Medical University, Hainan Medical University, Haikou, China.
| |
Collapse
|
35
|
Martínez-Navarro M, Cabañero D, Wawrzczak-Bargiela A, Robe A, Gavériaux-Ruff C, Kieffer BL, Przewlocki R, Baños JE, Maldonado R. Mu and delta opioid receptors play opposite nociceptive and behavioural roles on nerve-injured mice. Br J Pharmacol 2020; 177:1187-1205. [PMID: 31655493 DOI: 10.1111/bph.14911] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/01/2019] [Accepted: 10/12/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE Mu and delta opioid receptors(MOP, DOP) contribution to the manifestations of pathological pain is not understood. We used genetic approaches to investigate the opioid mechanisms modulating neuropathic pain and its comorbid manifestations. EXPERIMENTAL APPROACH We generated conditional knockout mice with MOP or DOP deletion in sensoryNav1.8-positive neurons (Nav1.8), in GABAergic forebrain neurons (DLX5/6) orconstitutively (CMV). Mutant mice and wild-type littermates were subjected topartial sciatic nerve ligation (PSNL) or sham surgery and their nociception wascompared. Anxiety-, depressivelike behaviour and cognitive performance were also measured. Opioid receptor mRNA expression, microgliosis and astrocytosis were assessed in the dorsalroot ganglia (DRG) and/or the spinal cord (SC). KEY RESULTS Constitutive CMV-MOP knockouts after PSNL displayed reduced mechanical allodynia and enhanced heat hyperalgesia. This phenotype was accompanied by increased DOP expression in DRG and SC, and reduced microgliosis and astrocytosis in deep dorsal horn laminae. Conditional MOP knockouts and control mice developed similar hypersensitivity after PSNL, except for anenhanced heat hyperalgesia by DLX5/6-MOP male mice. Neuropathic pain-induced anxiety was aggravated in CMV-MOP and DLX5/6-MOP knockouts. Nerve-injured CMV-DOP mice showed increased mechanical allodynia, whereas Nav1.8-DOP and DLX5/8-DOP mice had partial nociceptive enhancement. CMV-DOP and DLX5/6-DOP mutants showed increased depressive-like behaviour after PSNL. CONCLUSIONS AND IMPLICATIONS MOP activity after nerve injury increased anxiety-like responses involving forebrain GABAergic neurons and enhanced mechanical pain sensitivity along with repression of DOP expression and spinal cord gliosis. In contrast, DOP shows a protective function limiting nociceptive and affective manifestations of neuropathic pain.
Collapse
Affiliation(s)
- Miriam Martínez-Navarro
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Agnieszka Wawrzczak-Bargiela
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Anne Robe
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, Strasbourg, France.,IGBMC, Université de Strasbourg, Illkirch, France.,Laboratory UMR7104, Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Unit U 1258, Institut National de la Santé et de la Recherche Médicale, U 1258, Illkirch, France
| | - Claire Gavériaux-Ruff
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, Strasbourg, France.,IGBMC, Université de Strasbourg, Illkirch, France.,Laboratory UMR7104, Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Unit U 1258, Institut National de la Santé et de la Recherche Médicale, U 1258, Illkirch, France
| | - Brigitte L Kieffer
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, Strasbourg, France.,IGBMC, Université de Strasbourg, Illkirch, France.,Laboratory UMR7104, Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Unit U 1258, Institut National de la Santé et de la Recherche Médicale, U 1258, Illkirch, France.,Faculty of Medicine, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Josep E Baños
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Laboratory of Neuropharmacology, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
36
|
Sierra S, Gupta A, Gomes I, Fowkes M, Ram A, Bobeck EN, Devi LA. Targeting Cannabinoid 1 and Delta Opioid Receptor Heteromers Alleviates Chemotherapy-Induced Neuropathic Pain. ACS Pharmacol Transl Sci 2019; 2:219-229. [PMID: 31565698 PMCID: PMC6764458 DOI: 10.1021/acsptsci.9b00008] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Indexed: 01/10/2023]
Abstract
Cannabinoid 1 (CB1R) and delta opioid receptors (DOR) associate to form heteromers that exhibit distinct pharmacological properties. Not much is known about CB1R-DOR heteromer location or signaling along the pain circuit in either animal models or patients with chemotherapy-induced peripheral neuropathy (CIPN). Here, we use paclitaxel to induce CIPN in mice and confirm the development of mechanical allodynia. Under these conditions, we find significant increases in CB1R-DOR heteromers in the dorsal spinal cord of mice with CIPN as well as in postmortem spinal cords from human subjects with CIPN compared to controls. Next, we investigated receptor signaling in spinal cords of mice with CIPN and found that treatment with a combination of low signaling doses of CB1R and DOR ligands leads to significant enhancement in G-protein activity that could be selectively blocked by the CB1R-DOR antibody. Consistent with this, administration of subthreshold doses of a combination of ligands (CB1R agonist, Hu-210, and DOR agonist, SNC80) leads to significant attenuation of allodynia in mice with CIPN that is not seen with the administration of individual ligands, and this could be blocked by the CB1R-DOR antibody. Together, these results imply that CB1R-DOR heteromers upregulated during CIPN-associated mechanical allodynia could serve as a potential target for treatment of neuropathic pain including CIPN.
Collapse
Affiliation(s)
- Salvador Sierra
- Department
of Pharmacological Sciences and Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Achla Gupta
- Department
of Pharmacological Sciences and Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Ivone Gomes
- Department
of Pharmacological Sciences and Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Mary Fowkes
- Department
of Pharmacological Sciences and Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Akila Ram
- Department
of Biology, Utah State University, Logan, Utah 84322, United States
| | - Erin N. Bobeck
- Department
of Biology, Utah State University, Logan, Utah 84322, United States
| | - Lakshmi A. Devi
- Department
of Pharmacological Sciences and Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
37
|
Custodio-Patsey L, Donahue RR, Fu W, Lambert J, Smith BN, Taylor BK. Sex differences in kappa opioid receptor inhibition of latent postoperative pain sensitization in dorsal horn. Neuropharmacology 2019; 163:107726. [PMID: 31351975 DOI: 10.1016/j.neuropharm.2019.107726] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/11/2019] [Accepted: 07/25/2019] [Indexed: 12/31/2022]
Abstract
Tissue injury produces a delicate balance between latent pain sensitization (LS) and compensatory endogenous opioid receptor analgesia that continues for months, even after re-establishment of normal pain thresholds. To evaluate the contribution of mu (MOR), delta (DOR), and/or kappa (KOR) opioid receptors to the silencing of chronic postoperative pain, we performed plantar incision at the hindpaw, waited 21 days for the resolution of hyperalgesia, and then intrathecally injected subtype-selective ligands. We found that the MOR-selective inhibitor CTOP (1-1000 ng) dose-dependently reinstated mechanical hyperalgesia. Two DOR-selective inhibitors naltrindole (1-10 μg) and TIPP[Ψ] (1-20 μg) reinstated mechanical hyperalgesia, but only at the highest dose that also produced itching, licking, and tail biting. Both the prototypical KOR-selective inhibitors nor-BNI (0.1-10 μg) and the newer KOR inhibitor with more canonical pharmocodynamic effects, LY2456302 (0.1-10 μg), reinstated mechanical hyperalgesia. Furthermore, LY2456302 (10 μg) increased the expression of phosphorylated signal-regulated kinase (pERK), a marker of central sensitization, in dorsal horn neurons but not glia. Sex studies revealed that LY2456302 (0.3 μg) reinstated hyperalgesia and pERK expression to a greater degree in female as compared to male mice. Our results suggest that spinal MOR and KOR, but not DOR, maintain LS within a state of remission to reduce the intensity and duration of postoperative pain, and that endogenous KOR but not MOR analgesia is greater in female mice.
Collapse
Affiliation(s)
- Lilian Custodio-Patsey
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA
| | - Renée R Donahue
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA
| | - Weisi Fu
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA
| | - Joshua Lambert
- Department of Statistics, College of Arts and Sciences, University of Kentucky, 302 Multidisciplinary Science Building, Lexington, KY, 40536-0082, USA
| | - Bret N Smith
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA; Department of Neuroscience, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA
| | - Bradley K Taylor
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Pittsburgh Project to End Opioid Misuse, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
38
|
Zheng Y, Liu P, Bai L, Trimmer JS, Bean BP, Ginty DD. Deep Sequencing of Somatosensory Neurons Reveals Molecular Determinants of Intrinsic Physiological Properties. Neuron 2019; 103:598-616.e7. [PMID: 31248728 DOI: 10.1016/j.neuron.2019.05.039] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 04/16/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023]
Abstract
Dorsal root ganglion (DRG) sensory neuron subtypes defined by their in vivo properties display distinct intrinsic electrical properties. We used bulk RNA sequencing of genetically labeled neurons and electrophysiological analyses to define ion channel contributions to the intrinsic electrical properties of DRG neuron subtypes. The transcriptome profiles of eight DRG neuron subtypes revealed differentially expressed and functionally relevant genes, including voltage-gated ion channels. Guided by these data, electrophysiological analyses using pharmacological and genetic manipulations as well as computational modeling of DRG neuron subtypes were undertaken to assess the functions of select voltage-gated potassium channels (Kv1, Kv2, Kv3, and Kv4) in shaping action potential (AP) waveforms and firing patterns. Our findings show that the transcriptome profiles have predictive value for defining ion channel contributions to sensory neuron subtype-specific intrinsic physiological properties. The distinct ensembles of voltage-gated ion channels predicted to underlie the unique intrinsic physiological properties of eight DRG neuron subtypes are presented.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA; Neuroscience Training Program, Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Pin Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ling Bai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA; Neuroscience Training Program, Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, USA; Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Birdsong WT, Jongbloets BC, Engeln KA, Wang D, Scherrer G, Mao T. Synapse-specific opioid modulation of thalamo-cortico-striatal circuits. eLife 2019; 8:45146. [PMID: 31099753 PMCID: PMC6541437 DOI: 10.7554/elife.45146] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/15/2019] [Indexed: 11/19/2022] Open
Abstract
The medial thalamus (MThal), anterior cingulate cortex (ACC) and striatum play important roles in affective-motivational pain processing and reward learning. Opioids affect both pain and reward through uncharacterized modulation of this circuitry. This study examined opioid actions on glutamate transmission between these brain regions in mouse. Mu-opioid receptor (MOR) agonists potently inhibited MThal inputs without affecting ACC inputs to individual striatal medium spiny neurons (MSNs). MOR activation also inhibited MThal inputs to the pyramidal neurons in the ACC. In contrast, delta-opioid receptor (DOR) agonists disinhibited ACC pyramidal neuron responses to MThal inputs by suppressing local feed-forward GABA signaling from parvalbumin-positive interneurons. As a result, DOR activation in the ACC facilitated poly-synaptic (thalamo-cortico-striatal) excitation of MSNs by MThal inputs. These results suggest that opioid effects on pain and reward may be shaped by the relative selectivity of opioid drugs to the specific circuit components.
Collapse
Affiliation(s)
- William T Birdsong
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Bart C Jongbloets
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Kim A Engeln
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Dong Wang
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University, Stanford, United States.,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Stanford, United States.,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Stanford, United States
| | - Grégory Scherrer
- Department of Anesthesiology Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University, Stanford, United States.,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Stanford, United States.,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Stanford, United States.,New York Stem Cell Foundation - Robertson Investigator, Stanford University, Palo Alto, United States
| | - Tianyi Mao
- Vollum Institute, Oregon Health & Science University, Portland, United States
| |
Collapse
|
40
|
Irmak D, Fatima A, Gutiérrez-Garcia R, Rinschen MM, Wagle P, Altmüller J, Arrigoni L, Hummel B, Klein C, Frese CK, Sawarkar R, Rada-Iglesias A, Vilchez D. Mechanism suppressing H3K9 trimethylation in pluripotent stem cells and its demise by polyQ-expanded huntingtin mutations. Hum Mol Genet 2019; 27:4117-4134. [PMID: 30452683 DOI: 10.1093/hmg/ddy304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Pluripotent stem cells are invaluable resources to study development and disease, holding a great promise for regenerative medicine. Here we use human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) from patients with Huntington's disease (HD-iPSCs) to shed light into the normal function of huntingtin (HTT) and its demise in disease. We find that HTT binds ATF7IP, a regulator of the histone H3 methyltransferase SETDB1. HTT inhibits the interaction of the ATF7IP-SETDB1 complex with other heterochromatin regulators and transcriptional repressors, maintaining low levels of H3K9 trimethylation (H3K9me3) in hESCs. Loss of HTT promotes global increased H3K9me3 levels and enrichment of H3K9me3 marks at distinct genes, including transcriptional regulators of neuronal differentiation. Although these genes are normally expressed at low amounts in hESCs, HTT knockdown (KD) reduces their induction during neural differentiation. Notably, mutant expanded polyglutamine repeats in HTT diminish its interaction with ATF7IP-SETDB1 complex and trigger H3K9me3 in HD-iPSCs. Conversely, KD of ATF7IP in HD-iPSCs reduces H3K9me3 alterations and ameliorates gene expression changes in their neural counterparts. Taken together, our results indicate ATF7IP as a potential target to correct aberrant H3K9me3 levels induced by mutant HTT.
Collapse
Affiliation(s)
- Dilber Irmak
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany
| | - Azra Fatima
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany
| | - Ricardo Gutiérrez-Garcia
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany
| | - Markus M Rinschen
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany
| | - Prerana Wagle
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany
| | - Janine Altmüller
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Strasse 21, Cologne, Germany.,Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Laura Arrigoni
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Barbara Hummel
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Corinna Klein
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany
| | - Christian K Frese
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany
| | - Ritwick Sawarkar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Alvaro Rada-Iglesias
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Strasse 21, Cologne, Germany
| | - David Vilchez
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne, Germany
| |
Collapse
|
41
|
Moye LS, Tipton AF, Dripps I, Sheets Z, Crombie A, Violin JD, Pradhan AA. Delta opioid receptor agonists are effective for multiple types of headache disorders. Neuropharmacology 2019; 148:77-86. [PMID: 30553828 PMCID: PMC6467218 DOI: 10.1016/j.neuropharm.2018.12.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/19/2018] [Accepted: 12/12/2018] [Indexed: 12/20/2022]
Abstract
Headaches are highly disabling and are among the most common neurological disorders worldwide. Despite the high prevalence of headache, therapeutic options are limited. We recently identified the delta opioid receptor (DOR) as an emerging therapeutic target for migraine. In this study, we examined the effectiveness of a hallmark DOR agonist, SNC80, in disease models reflecting diverse headache disorders including: chronic migraine, post-traumatic headache (PTH), medication overuse headache by triptans (MOH), and opioid-induced hyperalgesia (OIH). To model chronic migraine C57BL/6J mice received chronic intermittent treatment with the known human migraine trigger, nitroglycerin. PTH was modeled by combining the closed head weight drop model with the nitroglycerin model of chronic migraine. For MOH and OIH, mice were chronically treated with sumatriptan or morphine, respectively. The development of periorbital and peripheral allodynia was observed in all four models; and SNC80 significantly inhibited allodynia in all cases. In addition, we also determined if chronic daily treatment with SNC80 would induce MOH/OIH, and we observed limited hyperalgesia relative to sumatriptan or morphine. Together, our results indicate that DOR agonists could be effective in multiple headache disorders, despite their distinct etiology, thus presenting a novel therapeutic target for headache.
Collapse
Affiliation(s)
- Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago, USA
| | - Alycia F Tipton
- Department of Psychiatry, University of Illinois at Chicago, USA
| | - Isaac Dripps
- Department of Psychiatry, University of Illinois at Chicago, USA
| | - Zoie Sheets
- Department of Psychiatry, University of Illinois at Chicago, USA
| | | | | | - Amynah A Pradhan
- Department of Psychiatry, University of Illinois at Chicago, USA.
| |
Collapse
|
42
|
Gendron L, Nagi K, Zeghal M, Giguère PM, Pineyro G. Molecular aspects of delta opioid receptors. OPIOID HORMONES 2019; 111:49-90. [DOI: 10.1016/bs.vh.2019.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
43
|
Abstract
Since the discovery of the NOP receptor and N/OFQ as the endogenous ligand, evidence has appeared demonstrating the involvement of this receptor system in pain. This was not surprising for members of the opioid receptor and peptide families, particularly since both the receptor and N/OFQ are highly expressed in brain regions involved in pain, spinal cord, and dorsal root ganglia. What has been surprising is the complicated picture that has emerged from 25 years of research. The original finding that N/OFQ decreased tail flick and hotplate latency, when administered i.c.v., led to the hypothesis that NOP receptor antagonists could have analgesic activity without abuse liability. However, as data accumulated, it became clear that not only the potency but the activity per se was different when N/OFQ or small molecule NOP agonists were administered in the brain versus the spinal cord and it also depended upon the pain assay used. When administered systemically, NOP receptor agonists are generally ineffective in attenuating heat pain but are antinociceptive in an acute inflammatory pain model. Most antagonists administered systemically have no antinociceptive activity of their own, even though selective peptide NOP antagonists have potent antinociceptive activity when administered i.c.v. Chronic pain models provide different results as well, as small molecule NOP receptor agonists have potent anti-allodynic and anti-hyperalgesic activity after systemic administration. A considerable number of electrophysiological and anatomical experiments, in particular with NOP-eGFP mice, have been conducted in an attempt to explain the complicated profile resulting from NOP receptor modulation, to examine receptor plasticity, and to elucidate mechanisms by which selective NOP agonists, bifunctional NOP/mu agonists, or NOP receptor antagonists modulate acute and chronic pain.
Collapse
Affiliation(s)
- Lawrence Toll
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA.
| | - Akihiko Ozawa
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Andrea Cippitelli
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
44
|
Snyder LM, Chiang MC, Loeza-Alcocer E, Omori Y, Hachisuka J, Sheahan TD, Gale JR, Adelman PC, Sypek EI, Fulton SA, Friedman RL, Wright MC, Duque MG, Lee YS, Hu Z, Huang H, Cai X, Meerschaert KA, Nagarajan V, Hirai T, Scherrer G, Kaplan DH, Porreca F, Davis BM, Gold MS, Koerber HR, Ross SE. Kappa Opioid Receptor Distribution and Function in Primary Afferents. Neuron 2018; 99:1274-1288.e6. [PMID: 30236284 PMCID: PMC6300132 DOI: 10.1016/j.neuron.2018.08.044] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/06/2018] [Accepted: 08/21/2018] [Indexed: 02/02/2023]
Abstract
Primary afferents are known to be inhibited by kappa opioid receptor (KOR) signaling. However, the specific types of somatosensory neurons that express KOR remain unclear. Here, using a newly developed KOR-cre knockin allele, viral tracing, single-cell RT-PCR, and ex vivo recordings, we show that KOR is expressed in several populations of primary afferents: a subset of peptidergic sensory neurons, as well as low-threshold mechanoreceptors that form lanceolate or circumferential endings around hair follicles. We find that KOR acts centrally to inhibit excitatory neurotransmission from KOR-cre afferents in laminae I and III, and this effect is likely due to KOR-mediated inhibition of Ca2+ influx, which we observed in sensory neurons from both mouse and human. In the periphery, KOR signaling inhibits neurogenic inflammation and nociceptor sensitization by inflammatory mediators. Finally, peripherally restricted KOR agonists selectively reduce pain and itch behaviors, as well as mechanical hypersensitivity associated with a surgical incision. These experiments provide a rationale for the use of peripherally restricted KOR agonists for therapeutic treatment.
Collapse
Affiliation(s)
- Lindsey M Snyder
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael C Chiang
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emanuel Loeza-Alcocer
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yu Omori
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Junichi Hachisuka
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tayler D Sheahan
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jenna R Gale
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Peter C Adelman
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Elizabeth I Sypek
- Department of Anesthesiology, Perioperative, and Pain Medicine, Department of Molecular and Cellular Physiology, and Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Stephanie A Fulton
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Robert L Friedman
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Margaret C Wright
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Melissa Giraldo Duque
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yeon Sun Lee
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Zeyu Hu
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Huizhen Huang
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Tsinghua University School of Medicine Beijing, Beijing 100084, China
| | - Xiaoyun Cai
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kimberly A Meerschaert
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Vidhya Nagarajan
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Toshiro Hirai
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Gregory Scherrer
- Department of Anesthesiology, Perioperative, and Pain Medicine, Department of Molecular and Cellular Physiology, and Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; New York Stem Cell Foundation-Robertson Investigator, Stanford University, Palo Alto, CA 94304, USA
| | - Daniel H Kaplan
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Frank Porreca
- Department of Pharmacology, University of Arizona, Tucson, AZ 85719, USA
| | - Brian M Davis
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael S Gold
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - H Richard Koerber
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Sarah E Ross
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
45
|
Presynaptic Inhibition of Primary Nociceptive Signals to Dorsal Horn Lamina I Neurons by Dopamine. J Neurosci 2018; 38:8809-8821. [PMID: 30143577 DOI: 10.1523/jneurosci.0323-18.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/18/2018] [Accepted: 08/09/2018] [Indexed: 01/10/2023] Open
Abstract
The dorsal horn of the spinal cord represents the first relay station in the pain pathway where primary nociceptive inputs are modulated by local circuits and by descending signals before being relayed to supraspinal nuclei. To determine whether dopamine can modulate primary nociceptive Aδ- and C-fiber signals, the effects of dopamine were tested on the excitatory postsynaptic currents (EPSCs) recorded from large lamina I neurons and from retrograde-labeled spinoparabrachial lamina I neurons upon stimulation of the L4/L5 dorsal root in horizontal spinal cord slices in vitro Dopamine inhibited the EPSCs in a dose-dependent manner, with substantial inhibition (33%) at 1 μm and maximum inhibition (∼70%) at 10-20 μm Dopamine reduced the frequency of miniature EPSCs recorded from large lamina I neurons, increased the paired pulse depression ratio of paired EPSCs, and induced similar inhibition of EPSCs after dialysis of large lamina I neurons with GDP-β-S, consistent with actions at presynaptic sites. Pharmacological experiments suggested that the inhibitory effects of dopamine were largely mediated by D4 receptors (53%). Similar inhibition (66%) by dopamine was observed on EPSCs recorded from ipsilateral large lamina I neurons 6 d after injection of complete Freund's adjuvant in the hindpaw, suggesting that dopamine downregulates primary nociceptive inputs to lamina I neurons during chronic inflammatory pain. We propose that presynaptic inhibition of primary nociceptive inputs to lamina I projection neurons is a mechanism whereby dopamine can inhibit incoming noxious stimuli to the dorsal horn of the spinal cord.SIGNIFICANCE STATEMENT Lamina I projection neurons represent the main output for the pain signals from the dorsal horn of the spinal cord to brainstem and thalamic nuclei. We found that dopamine inhibits the nociceptive Aδ- and C-fiber synaptic inputs to lamina I projection neurons via presynaptic actions. Similar inhibitory effects of dopamine on the EPSCs were observed in rats subjected to complete Freund's adjuvant to induce peripheral inflammation, suggesting that dopamine inhibits the synaptic inputs to lamina I neurons in the setting of injury. A better understanding of how primary nociceptive inputs to the dorsal horn of the spinal cord are modulated by descending monoaminergic signals may help in the development of new pharmacological strategies to selectively downregulate the output from lamina I projection neurons.
Collapse
|
46
|
Ceredig RA, Pierre F, Doridot S, Alduntzin U, Salvat E, Yalcin I, Gaveriaux-Ruff C, Barrot M, Massotte D. Peripheral delta opioid receptors mediate duloxetine antiallodynic effect in a mouse model of neuropathic pain. Eur J Neurosci 2018; 48:2231-2246. [PMID: 30059180 DOI: 10.1111/ejn.14093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/30/2018] [Accepted: 07/23/2018] [Indexed: 02/06/2023]
Abstract
Peripheral delta opioid (DOP) receptors are essential for the antiallodynic effect of the tricyclic antidepressant nortriptyline. However, the population of DOP-expressing cells affected in neuropathic conditions or underlying the antiallodynic activity of antidepressants remains unknown. Using a mouse line in which DOP receptors were selectively ablated in cells expressing Nav1.8 sodium channels (DOP cKO), we established that these DOP peripheral receptors were mandatory for duloxetine to alleviate mechanical allodynia in a neuropathic pain model based on sciatic nerve cuffing. We then examined the impact of nerve cuffing and duloxetine treatment on DOP-positive populations using a knock-in mouse line expressing a fluorescent version of the DOP receptor fused with the enhanced green fluorescent protein (DOPeGFP). Eight weeks postsurgery, we observed a reduced proportion of DOPeGFP-positive small peptidergic sensory neurons (calcitonin gene-related peptide (CGRP) positive) in dorsal root ganglia and a lower density of DOPeGFP-positive free nerve endings in the skin. These changes were not present in nerve-injured mice chronically treated with oral duloxetine. In addition, increased DOPeGFP translocation to the plasma membrane was observed in neuropathic conditions but not in duloxetine-treated neuropathic mice, which may represent an additional level of control of the neuronal activity by DOP receptors. Our results therefore established a parallel between changes in the expression profile of peripheral DOP receptors and mechanical allodynia induced by sciatic nerve cuffing.
Collapse
Affiliation(s)
- Rhian Alice Ceredig
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Florian Pierre
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Stéphane Doridot
- Centre National de la Recherche Scientifique, Chronobiotron, Strasbourg, France
| | - Unai Alduntzin
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Eric Salvat
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France.,Centre d'Evaluation et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Claire Gaveriaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, Illkirch, France
| | - Michel Barrot
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Dominique Massotte
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
47
|
Ozawa A, Brunori G, Cippitelli A, Toll N, Schoch J, Kieffer BL, Toll L. Analysis of the distribution of spinal NOP receptors in a chronic pain model using NOP-eGFP knock-in mice. Br J Pharmacol 2018; 175:2662-2675. [PMID: 29582417 PMCID: PMC6003644 DOI: 10.1111/bph.14225] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/16/2018] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The nociceptin/orphanin FQ opioid peptide (NOP) receptor system plays a significant role in the regulation of pain. This system functions differently in the spinal cord and brain. The mechanism by which the NOP receptor agonists regulate pain transmission in these regions is not clearly understood. Here, we investigate the peripheral and spinal NOP receptor distribution and antinociceptive effects of intrathecal nociceptin/orphanin FQ (N/OFQ) in chronic neuropathic pain. EXPERIMENTAL APPROACH We used immunohistochemistry to determine changes in NOP receptor distribution triggered by spinal nerve ligation (SNL) using NOP-eGFP knock-in mice. Antinociceptive effects of intrathecal N/OFQ on SNL-mediated allodynia and heat/cold hyperalgesia were assessed in wild-type mice. KEY RESULTS NOP-eGFP immunoreactivity was decreased by SNL in the spinal laminae I and II outer, regions that mediate noxious heat stimuli. In contrast, immunoreactivity of NOP-eGFP was unchanged in the ventral border of lamina II inner, which is an important region for the development of allodynia. NOP-eGFP expression was also decreased in a large number of primary afferents in the L4 dorsal root ganglion (DRG) of SNL mice. However, SNL mice showed increased sensitivity, compared to sham animals to the effects of i.t administered N/OFQ with respect to mechanical as well as thermal stimuli. CONCLUSIONS AND IMPLICATIONS Our findings suggest that the spinal NOP receptor system attenuates injury-induced hyperalgesia by direct inhibition of the projection neurons in the spinal cord that send nociceptive signals to the brain and not by inhibiting presynaptic terminals of DRG neurons in the superficial lamina.
Collapse
Affiliation(s)
- Akihiko Ozawa
- Torrey Pines Institute for Molecular StudiesPort St. LucieFL34987USA
| | - Gloria Brunori
- Torrey Pines Institute for Molecular StudiesPort St. LucieFL34987USA
- Department of Biomedical Science, Charles E. Schmidt College of MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Andrea Cippitelli
- Torrey Pines Institute for Molecular StudiesPort St. LucieFL34987USA
| | - Nicholas Toll
- Torrey Pines Institute for Molecular StudiesPort St. LucieFL34987USA
| | - Jennifer Schoch
- Torrey Pines Institute for Molecular StudiesPort St. LucieFL34987USA
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, Faculty of MedicineMcGill UniversityMontrealQCH4H 1R3Canada
| | - Lawrence Toll
- Torrey Pines Institute for Molecular StudiesPort St. LucieFL34987USA
- Department of Biomedical Science, Charles E. Schmidt College of MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
| |
Collapse
|
48
|
Montilla-García Á, Perazzoli G, Tejada MÁ, González-Cano R, Sánchez-Fernández C, Cobos EJ, Baeyens JM. Modality-specific peripheral antinociceptive effects of μ-opioid agonists on heat and mechanical stimuli: Contribution of sigma-1 receptors. Neuropharmacology 2018; 135:328-342. [PMID: 29580951 DOI: 10.1016/j.neuropharm.2018.03.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/05/2018] [Accepted: 03/20/2018] [Indexed: 11/16/2022]
Abstract
Morphine induces peripherally μ-opioid-mediated antinociception to heat but not to mechanical stimulation. Peripheral sigma-1 receptors tonically inhibit μ-opioid antinociception to mechanical stimuli, but it is unknown whether they modulate μ-opioid heat antinociception. We hypothesized that sigma-1 receptors might play a role in the modality-specific peripheral antinociceptive effects of morphine and other clinically relevant μ-opioid agonists. Mechanical nociception was assessed in mice with the paw pressure test (450 g), and heat nociception with the unilateral hot plate (55 °C) test. Local peripheral (intraplantar) administration of morphine, buprenorphine or oxycodone did not induce antinociception to mechanical stimulation but had dose-dependent antinociceptive effects on heat stimuli. Local sigma-1 antagonism unmasked peripheral antinociception by μ-opioid agonists to mechanical stimuli, but did not modify their effects on heat stimulation. TRPV1+ and IB4+ cells are segregated populations of small neurons in the dorsal root ganglia (DRG) and the density of sigma-1 receptors was higher in IB4+ cells than in the rest of small nociceptive neurons. The in vivo ablation of TRPV1-expressing neurons with resiniferatoxin did not alter IB4+ neurons in the DRG, mechanical nociception, or the effects of sigma-1 antagonism on local morphine antinociception in this type of stimulus. However, it impaired the responses to heat stimuli and the effect of local morphine on heat nociception. In conclusion, peripheral opioid antinociception to mechanical stimuli is limited by sigma-1 tonic inhibitory actions, whereas peripheral opioid antinociception to heat stimuli (produced in TRPV1-expressing neurons) is not. Therefore, sigma-1 receptors contribute to the modality-specific peripheral effects of opioid analgesics.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/pathology
- Hot Temperature
- Hyperalgesia/drug therapy
- Hyperalgesia/metabolism
- Hyperalgesia/pathology
- Mice, Knockout
- Nociceptors/drug effects
- Nociceptors/metabolism
- Nociceptors/pathology
- Random Allocation
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/agonists
- Receptors, sigma/antagonists & inhibitors
- Receptors, sigma/genetics
- Receptors, sigma/metabolism
- TRPV Cation Channels/metabolism
- Touch
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Gloria Perazzoli
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Miguel Á Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Rafael González-Cano
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Cristina Sánchez-Fernández
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain; Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain; Teófilo Hernando Institute for Drug Discovery, 28029 Madrid, Spain.
| | - José M Baeyens
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Parque Tecnológico de Ciencias de la Salud, 18100 Armilla, Granada, Spain; Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain.
| |
Collapse
|
49
|
Abstract
Opioids are the most commonly used and effective analgesic treatments for severe pain, but they have recently come under scrutiny owing to epidemic levels of abuse and overdose. These compounds act on the endogenous opioid system, which comprises four G protein-coupled receptors (mu, delta, kappa, and nociceptin) and four major peptide families (β-endorphin, enkephalins, dynorphins, and nociceptin/orphanin FQ). In this review, we first describe the functional organization and pharmacology of the endogenous opioid system. We then summarize current knowledge on the signaling mechanisms by which opioids regulate neuronal function and neurotransmission. Finally, we discuss the loci of opioid analgesic action along peripheral and central pain pathways, emphasizing the pain-relieving properties of opioids against the affective dimension of the pain experience.
Collapse
Affiliation(s)
- Gregory Corder
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, California 94304, USA; .,Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, California 94304, USA.,Department of Neurosurgery, Stanford University, Palo Alto, California 94304, USA.,Stanford Neurosciences Institute, Palo Alto, California 94304, USA
| | - Daniel C Castro
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63130, USA;
| | - Michael R Bruchas
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63130, USA; .,Division of Basic Research, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, California 94304, USA; .,Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, California 94304, USA.,Department of Neurosurgery, Stanford University, Palo Alto, California 94304, USA.,Stanford Neurosciences Institute, Palo Alto, California 94304, USA.,New York Stem Cell Foundation - Robertson Investigator, Stanford University, Palo Alto, California 94304, USA
| |
Collapse
|
50
|
Guerrero-Alba R, Valdez-Morales EE, Jiménez-Vargas NN, Bron R, Poole D, Reed D, Castro J, Campaniello M, Hughes PA, Brierley SM, Bunnett N, Lomax AE, Vanner S. Co-expression of μ and δ opioid receptors by mouse colonic nociceptors. Br J Pharmacol 2018; 175:2622-2634. [PMID: 29579315 DOI: 10.1111/bph.14222] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE To better understand opioid signalling in visceral nociceptors, we examined the expression and selective activation of μ and δ opioid receptors by dorsal root ganglia (DRG) neurons innervating the mouse colon. EXPERIMENTAL APPROACH DRG neurons projecting to the colon were identified by retrograde tracing. δ receptor-GFP reporter mice, in situ hybridization, single-cell RT-PCR and μ receptor-specific antibodies were used to characterize expression of μ and δ receptors. Voltage-gated Ca2+ currents and neuronal excitability were recorded in small diameter nociceptive neurons (capacitance <30 pF) by patch clamp and ex vivo single-unit afferent recordings were obtained from the colon. KEY RESULTS In situ hybridization of oprm1 expression in Fast Blue-labelled DRG neurons was observed in 61% of neurons. μ and δ receptors were expressed by 36-46% of colon DRG neurons, and co-expressed by ~25% of neurons. μ and δ receptor agonists inhibited Ca2+ currents in DRG, effects blocked by opioid antagonists. One or both agonists inhibited action potential firing by colonic afferent endings. Incubation of neurons with supernatants from inflamed colon segments inhibited Ca2+ currents and neuronal excitability. Antagonists of μ, but not δ receptors, inhibited the effects of these supernatant on Ca2+ currents, whereas both antagonists inhibited their actions on neuronal excitability. CONCLUSIONS AND IMPLICATIONS A significant number of small diameter colonic nociceptors co-express μ and δ receptors and are inhibited by agonists and endogenous opioids in inflamed tissues. Thus, opioids that act at μ or δ receptors, or their heterodimers may be effective in treating visceral pain.
Collapse
Affiliation(s)
- Raquel Guerrero-Alba
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | | | | | - Romke Bron
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Daniel Poole
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - David Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - Joel Castro
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Melissa Campaniello
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Patrick A Hughes
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Nigel Bunnett
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC, Australia.,Departments of Surgery and Pharmacology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - Stephen Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, ON, Canada
| |
Collapse
|