1
|
Tassou A, Richebe P, Rivat C. Mechanisms of chronic postsurgical pain. Reg Anesth Pain Med 2025; 50:77-85. [PMID: 39909543 DOI: 10.1136/rapm-2024-105964] [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: 08/14/2024] [Accepted: 10/25/2024] [Indexed: 02/07/2025]
Abstract
Chronic pain after surgery, also known as chronic postsurgical pain (CPSP), is recognized as a significant public health issue with serious medical and economic consequences. Current research on CPSP underscores the significant roles of both peripheral and central sensitization in pain development and maintenance. Peripheral sensitization occurs at the site of injury, through the hyperexcitability of nerve fibers due to surgical damage and the release of inflammatory mediators. This leads to increased expression of pronociceptive ion channels and receptors, such as transient receptor potential and acid-sensing ion channels (ASIC), enhancing pain signal transmission. Central sensitization involves long-term changes in the central nervous system, particularly in the spinal cord. In this context, sensitized spinal neurons become more responsive to pain signals, driven by continuous nociceptive input from the periphery, which results in an enhanced pain response characterized by hyperalgesia and/or allodynia. Key players in this process include N-methyl-D-aspartate receptor and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, along with proinflammatory cytokines and chemokines released by activated glia. These glial cells release substances that further increase neuronal excitability, maintaining the sensitized state and contributing to persistent pain. The activation of antinociceptive systems is required for the resolution of pain after surgery, and default in these systems may also be considered as an important component of CPSP. In this review, we will examine the clinical factors underlying CPSP in patients and the mechanisms previously established in preclinical models of CPSP that may explain how acute postoperative pain may transform into chronic pain in patients.
Collapse
Affiliation(s)
- Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Philippe Richebe
- Department of Anesthesiology and Pain Medicine, Polyclinique Bordeaux Nord Aquitaine (PBNA), Bordeaux, France
- Anesthesiology and Pain Medicine, Maisonneuve Rosemont Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Cyril Rivat
- University of Montpellier, Montpellier, France
- Institut des Neurosciences de Montpellier INSERM U1298, Montpellier, France
| |
Collapse
|
2
|
Inyang KE, Sim J, Clark KB, Geron M, Monahan K, Evans C, O'Connell P, Laumet S, Peng B, Ma J, Heijnen CJ, Dantzer R, Scherrer G, Kavelaars A, Bernard M, Aldhamen YA, Folger JK, Bavencoffe A, Laumet G. Upregulation of delta opioid receptor by meningeal interleukin-10 prevents relapsing pain. Brain Behav Immun 2025; 123:399-410. [PMID: 39349285 PMCID: PMC11624093 DOI: 10.1016/j.bbi.2024.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024] Open
Abstract
Chronic pain often includes periods of transient amelioration and even remission that alternate with severe relapsing pain. While most research on chronic pain has focused on pain development and maintenance, there is a critical unmet need to better understand the mechanisms that underlie pain remission and relapse. We found that interleukin (IL)-10, a pain resolving cytokine, is produced by resident macrophages in the spinal meninges during remission from pain and signaled to IL-10 receptor-expressing sensory neurons. Using unbiased RNA-sequencing, we identified that IL-10 upregulated expression and antinociceptive activity of δ-opioid receptor (δOR) in the dorsal root ganglion. Genetic or pharmacological inhibition of either IL-10 signaling or δOR triggered relapsing pain. Overall, our findings, from electrophysiology, genetic manipulation, flow cytometry, pharmacology, and behavioral approaches, indicate that remission of pain is not simply a return to the naïve state. Instead, remission is an adapted homeostatic state associated with lasting pain vulnerability resulting from persisting neuroimmune interactions within the nociceptive system. Broadly, this sheds light on the elusive mechanisms underlying recurrence a common aspect across various chronic pain conditions.
Collapse
Affiliation(s)
| | - Jaewon Sim
- Department of Physiology, Michigan State University, East Lansing, MI, USA; Cell and Molecular Biology Graduate Program, Michigan State University, East Lansing, MI, USA
| | - Kimberly B Clark
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matan Geron
- Department of Cell Biology and Physiology, Department of Pharmacology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Karli Monahan
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Christine Evans
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Patrick O'Connell
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Sophie Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Bo Peng
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiacheng Ma
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cobi J Heijnen
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, Department of Pharmacology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA; New York Stem Cell Foundation - Robertson Investigator, University of North Carolina, Chapel Hill, NC, USA
| | - Annemieke Kavelaars
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew Bernard
- Flow Cytometry Core, Michigan State University, East Lansing, MI, USA
| | - Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Joseph K Folger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA; Cell and Molecular Biology Graduate Program, Michigan State University, East Lansing, MI, USA; Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
3
|
Kitanaka N, Arai K, Takehara K, Hall FS, Tomita K, Igarashi K, Sato T, Uhl GR, Kitanaka J. Opioid receptor antagonists reduce motivated wheel-running behavior in mice. Behav Pharmacol 2024; 35:114-121. [PMID: 38451023 DOI: 10.1097/fbp.0000000000000769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
We hypothesized that opioid receptor antagonists would inhibit motivated behavior produced by a natural reward. To evaluate motivated responses to a natural reward, mice were given access to running wheels for 71.5 h in a multi-configuration testing apparatus. In addition to a running wheel activity, locomotor activity (outside of the wheel), food and water intake, and access to a food container were measured in the apparatus. Mice were also tested separately for novel-object exploration to investigate whether naloxone affects behavior unrelated to natural reward. In untreated mice wheel running increased from day 1 to day 3. The selective µ-opioid receptor antagonist β-funaltrexamine (β-FNA) (5 mg/kg) slightly decreased wheel running, but did not affect the increase in wheel running from day 1 to day 3. The non-selective opioid receptor antagonist naloxone produced a greater reduction in wheel running than β-FNA and eliminated the increase in wheel running that occurred over time in the other groups. Analysis of food access, locomotor behavior, and behavior in the novel-object test suggested that the reduction in wheel running was selective for this highly reinforcing behavior. These results indicate that opioid receptor antagonism reduces responses to the natural rewarding effects of wheel running and that these effects involve multiple opioid receptors since the non-selective opioid receptor antagonist had greater effects than the selective µ-opioid receptor antagonist. It is possible that at the doses employed, other receptor systems than opioid receptors might be involved, at least in part, in the effect of naloxone and β-FNA.
Collapse
Affiliation(s)
- Nobue Kitanaka
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Kanayo Arai
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Kaoko Takehara
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, Ohio, USA
| | - Kazuo Tomita
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kento Igarashi
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tomoaki Sato
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - George R Uhl
- Neurology, VA Maryland Healthcare System
- Departments of Neurology
- Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Junichi Kitanaka
- Neurology, VA Maryland Healthcare System
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Kobe, Japan
| |
Collapse
|
4
|
Fuller AM, Bharde S, Sikandar S. The mechanisms and management of persistent postsurgical pain. FRONTIERS IN PAIN RESEARCH 2023; 4:1154597. [PMID: 37484030 PMCID: PMC10357043 DOI: 10.3389/fpain.2023.1154597] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
An estimated 10%-50% of patients undergoing a surgical intervention will develop persistent postsurgical pain (PPP) lasting more than 3 months despite adequate acute pain management and the availability of minimally invasive procedures. The link between early and late pain outcomes for surgical procedures remains unclear-some patients improve while others develop persistent pain. The elective nature of a surgical procedure offers a unique opportunity for prophylactic or early intervention to prevent the development of PPP and improve our understanding of its associated risk factors, such as pre-operative anxiety and the duration of severe acute postoperative pain. Current perioperative pain management strategies often include opioids, but long-term consumption can lead to tolerance, addiction, opioid-induced hyperalgesia, and death. Pre-clinical models provide the opportunity to dissect mechanisms underpinning the transition from acute to chronic, or persistent, postsurgical pain. This review highlights putative mechanisms of PPP, including sensitisation of peripheral sensory neurons, neuroplasticity in the central nervous system and nociceptive signalling along the neuro-immune axis.
Collapse
|
5
|
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
|
6
|
Jin Y, Mao Y, Chen D, Tai Y, Hu R, Yang CL, Zhou J, Chen L, Liu X, Gu E, Jia C, Zhang Z, Tao W. Thalamocortical circuits drive remifentanil-induced postoperative hyperalgesia. J Clin Invest 2022; 132:158742. [PMID: 36519547 PMCID: PMC9754001 DOI: 10.1172/jci158742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 10/18/2022] [Indexed: 12/15/2022] Open
Abstract
Remifentanil-induced hyperalgesia (RIH) is a severe but common postoperative clinical problem with elusive underlying neural mechanisms. Here, we discovered that glutamatergic neurons in the thalamic ventral posterolateral nucleus (VPLGlu) exhibited significantly elevated burst firing accompanied by upregulation of Cav3.1 T-type calcium channel expression and function in RIH model mice. In addition, we identified a glutamatergic neuronal thalamocortical circuit in the VPL projecting to hindlimb primary somatosensory cortex glutamatergic neurons (S1HLGlu) that mediated RIH. In vivo calcium imaging and multi-tetrode recordings revealed heightened S1HLGlu neuronal activity during RIH. Moreover, preoperative suppression of Cav3.1-dependent burst firing in VPLGlu neurons or chemogenetic inhibition of VPLGlu neuronal terminals in the S1HL abolished the increased S1HLGlu neuronal excitability while alleviating RIH. Our findings suggest that remifentanil induces postoperative hyperalgesia by upregulating T-type calcium channel-dependent burst firing in VPLGlu neurons to activate S1HLGlu neurons, thus revealing an ion channel-mediated neural circuit basis for RIH that can guide analgesic development.
Collapse
Affiliation(s)
- Yan Jin
- Stroke Center and Department of Neurology and,Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Yu Mao
- Stroke Center and Department of Neurology and,Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Danyang Chen
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Yingju Tai
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Rui Hu
- Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chen-Ling Yang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jing Zhou
- Department of head, neck, and breast Surgery, Western district of the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Lijian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Erwei Gu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chunhui Jia
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Zhi Zhang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, China
| | - Wenjuan Tao
- Stroke Center and Department of Neurology and,Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
David MS, Jones J, Lauriello A, Nnake I, Plazas Montana M, Lasko K, Buri-Nagua C, Olagbaju Y, Williams E, Sears M, Salzberg B, Lanzkron SM, Carroll CP. Converting adults with sickle cell disease from full agonist opioids to buprenorphine: A reliable method with safety and early evidence of reduced acute care utilization. Am J Hematol 2022; 97:1435-1442. [PMID: 36053825 DOI: 10.1002/ajh.26699] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/21/2022] [Accepted: 08/13/2022] [Indexed: 01/28/2023]
Abstract
Buprenorphine, a novel opioid with complex pharmacology, is effective for treating pain and is qualitatively safer than high-dose full agonist opioid therapy; but transitioning to buprenorphine can be technically complex and carries some risk of precipitated withdrawal. We report our clinic's experience converting 36 patients with sickle cell disease (SCD) from full agonist opioids to buprenorphine using a method developed in the past 10 years. Thirty of these patients were induced using a standard outpatient protocol and six were induced during medical admissions. Typically, patients were on high-dose chronic opioid therapy (COT) with inadequate response, and often with very high acute care utilization. Unlike prior case series, the method of induction, dosing, and management of withdrawal are detailed, as are post-induction adverse events. There were seven adverse events in the first 3 days following standard induction, and two of which were judged to be definitely related to the induction but none with any lasting sequelae. At 6 months follow-up, five participants had discontinued buprenorphine (16.67%), and overall acute care visits dropped from a mean of 10.50 (SD 11.35) in the 6 months pre-induction to 2.89 (SD 3.40) in the 6 months post-induction. In an appropriately interdisciplinary care setting, buprenorphine shows promise as a safe alternative to COT with early evidence of benefit for high-utilizing patients with SCD.
Collapse
Affiliation(s)
- Mandy S David
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Jones
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ashley Lauriello
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ijeoma Nnake
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Manuela Plazas Montana
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kyra Lasko
- Department of Emergency Medicine, University of Maryland Medical System, Baltimore, Maryland, USA
| | | | - Yetunde Olagbaju
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth Williams
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matthew Sears
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Sophie M Lanzkron
- Department of Medicine, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C Patrick Carroll
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Chen Q, Heinricher MM. Shifting the Balance: How Top-Down and Bottom-Up Input Modulate Pain via the Rostral Ventromedial Medulla. FRONTIERS IN PAIN RESEARCH 2022; 3:932476. [PMID: 35836737 PMCID: PMC9274196 DOI: 10.3389/fpain.2022.932476] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
The sensory experience of pain depends not only on the transmission of noxious information (nociception), but on the state of the body in a biological, psychological, and social milieu. A brainstem pain-modulating system with its output node in the rostral ventromedial medulla (RVM) can regulate the threshold and gain for nociceptive transmission. This review considers the current understanding of how RVM pain-modulating neurons, namely ON-cells and OFF-cells, are engaged by “top-down” cognitive and emotional factors, as well as by “bottom-up” sensory inputs, to enhance or suppress pain.
Collapse
Affiliation(s)
- Qiliang Chen
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
| | - Mary M. Heinricher
- Department of Neurological Surgery and Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- *Correspondence: Mary M. Heinricher
| |
Collapse
|
9
|
Cooper AH, Hedden NS, Corder G, Lamerand SR, Donahue RR, Morales-Medina JC, Selan L, Prasoon P, Taylor BK. Endogenous µ-opioid receptor activity in the lateral and capsular subdivisions of the right central nucleus of the amygdala prevents chronic postoperative pain. J Neurosci Res 2022; 100:48-65. [PMID: 33957003 PMCID: PMC8571119 DOI: 10.1002/jnr.24846] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/29/2021] [Indexed: 01/03/2023]
Abstract
Tissue injury induces a long-lasting latent sensitization (LS) of spinal nociceptive signaling that is kept in remission by an opposing µ-opioid receptor (MOR) constitutive activity. To test the hypothesis that supraspinal sites become engaged, we induced hindpaw inflammation, waited 3 weeks for mechanical hypersensitivity to resolve, and then injected the opioid receptor inhibitors naltrexone, CTOP or β-funaltrexamine subcutaneously, and/or into the cerebral ventricles. Intracerebroventricular injection of each inhibitor reinstated hypersensitivity and produced somatic signs of withdrawal, indicative of LS and endogenous opioid dependence, respectively. In naïve or sham controls, systemic naloxone (3 mg/kg) produced conditioned place aversion, and systemic naltrexone (3 mg/kg) increased Fos expression in the central nucleus of the amygdala (CeA). In LS animals tested 3 weeks after plantar incision, systemic naltrexone reinstated mechanical hypersensitivity and produced an even greater increase in Fos than in sham controls, particularly in the capsular subdivision of the right CeA. One third of Fos+ profiles co-expressed protein kinase C delta (PKCδ), and 35% of PKCδ neurons co-expressed tdTomato+ in Oprm1Cre ::tdTomato transgenic mice. CeA microinjection of naltrexone (1 µg) reinstated mechanical hypersensitivity only in male mice and did not produce signs of somatic withdrawal. Intra-CeA injection of the MOR-selective inhibitor CTAP (300 ng) reinstated hypersensitivity in both male and female mice. We conclude that MORs in the capsular subdivision of the right CeA prevent the transition from acute to chronic postoperative pain.
Collapse
Affiliation(s)
- Andrew H. Cooper
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and the Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Naomi S. Hedden
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and the Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Gregory Corder
- Department of Psychiatry and Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sydney R. Lamerand
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and the Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neurosciences at the University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Renee R. Donahue
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | | | - Lindsay Selan
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and the Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Pranav Prasoon
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and the Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Bradley K. Taylor
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and the Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
10
|
Gerum M, Simonin F. Behavioral characterization, potential clinical relevance and mechanisms of latent pain sensitization. Pharmacol Ther 2021; 233:108032. [PMID: 34763010 DOI: 10.1016/j.pharmthera.2021.108032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
Chronic pain is a debilitating disorder that can occur as painful episodes that alternates with bouts of remission and occurs despite healing of the primary insult. Those episodes are often triggered by stressful events. In the last decades, a similar situation has been evidenced in a wide variety of rodent models (including inflammatory pain, neuropathy and opioid-induced hyperalgesia) where animals develop a chronic latent hyperalgesia that silently persists after behavioral signs of pain resolution. This state, referred as latent pain sensitization, is due to the compensatory activation of antinociceptive systems, such as the opioid system or NPY and its receptors. A transitory phase of hyperalgesia can then be reinstated by pharmacological or genetic blockade of these antinociceptive systems or by submitting animals to acute stress. Those observations reveal that there is a constant endogenous analgesia responsible for chronic pain inhibition that might paradoxically contribute to maintain this maladaptive state and could then participate to the transition from acute to chronic pain. Thus, demonstration of the existence of this phenomenon in humans and a better understanding of the mechanisms by which latent pain sensitization develops and maintains over long periods of time will be of particular interest to help identifying new therapeutic strategies and targets for chronic pain treatment. The present review aims to recapitulate behavioral expression, potential clinical relevance, cellular mechanisms and intracellular signaling pathways involved so far in latent pain sensitization.
Collapse
Affiliation(s)
- Manon Gerum
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS, Université de Strasbourg, Institut du Médicament de Strasbourg, Illkirch-Graffenstaden, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS, Université de Strasbourg, Institut du Médicament de Strasbourg, Illkirch-Graffenstaden, France.
| |
Collapse
|
11
|
Shaheed G, Manjooran AP, Reddy AJ, Nawathey N, Habib S, Brahmbhatt H. Low-Dose Naltrexone Co-Treatment in the Prevention of Opioid-Induced Hyperalgesia. Cureus 2021; 13:e17667. [PMID: 34646707 DOI: 10.7759/cureus.17667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 11/05/2022] Open
Abstract
Opioid-induced hyperalgesia (OIH) is characterized by a heightened sensitivity to pain that occurs in patients following opioid use. Prescription of opioids is currently the standard form of pain management for both neuropathic and nociceptive pain, due to the relief that patients typically report following their use. Opioids, which aim to provide analgesic effects, can paradoxically cause increasing degrees of pain among the users. The increased nociception can be either due to the underlying pain for which the opioid was initially prescribed, or other unrelated pain. As a result, those who are initially prescribed opioids for chronic pain relief may instead be left with no overall relief, and experience additional algesia. While OIH can be treated through the reduction of opioid use, antagonistic treatment can also be utilized. In an attempt to reduce OIH in patients, low doses of the opioid antagonist naltrexone can be given concurrently. This review will analyze the current role and effectiveness of the use of naltrexone in managing OIH in opioid users as described in clinical and non-clinical studies. Additionally, it seeks to characterize the underlying mechanisms that enable opioid antagonist naltrexone to reduce OIH while still allowing opioids to act as an analgesic. The authors find that OIH is a prevalent condition, and in order to effectively combat it, clinicians and patients can benefit from an extended study on how naltrexone can be utilized as a treatment alongside opioids prescribed for pain management.
Collapse
Affiliation(s)
- Gurneet Shaheed
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, USA
| | | | - Akshay J Reddy
- Opthalmology, California Northstate University College of Medicine, Elk Grove, USA
| | - Neel Nawathey
- Health Sciences, California Northstate University, Rancho Cordova, USA
| | - Samuel Habib
- Health Sciences, Santa Clara University, Santa Clara, USA
| | | |
Collapse
|
12
|
García-López J, Polanco-García M, Montes A. Factors Associated With the Risk of Developing Moderate to Severe Acute Postoperative Pain After Primary Total Knee Arthroplasty: Results From the PAIN OUT Registry. J Arthroplasty 2021; 36:1966-1973. [PMID: 33637382 DOI: 10.1016/j.arth.2021.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/08/2021] [Accepted: 02/01/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Total knee arthroplasty (TKA) is one of the most common procedures in orthopedic surgery and not always matches with patient's expectations of pain relief and function improvement. The aim of this study was to assess risk factors for developing moderate to severe acute postoperative pain (APOP) after TKA using the PAIN OUT questionnaire. METHODS Prospective, multicentre, international cohort study within the PAIN OUT project. Patients' outcomes were measured with 11-point numerical rating scales (0 = null, 10 = worst possible). Patient and analgesic/anesthetic treatment were assessed. Odds ratio for moderate-severe pain was calculated for each variable and if they were statistically significant in the univariate logistic model, variables were fitted into a multivariate logistic regression model. The effect size was assessed by Cohen's d coefficient. RESULTS In total, 968 patients were evaluated. The multivariate model identified chronic preoperative pain (P < .001), general anesthesia (P = .020), and receiving chronic opioids before (P = .020) or after the surgery (P < .001) as factors associated with moderate-severe APOP. No protective factors were observed. CONCLUSIONS Our model identified several risk factors for APOP. From our results, preoperative chronic pain, general anesthesia and the use of opioid analgesics could be predictors for higher APOP. These findings may help establish new strategies for the treatment of pain in TKR. More studies should be carried out to identify acute pain predictors and to develop better strategies of pain management for risk patients.
Collapse
Affiliation(s)
- Jaume García-López
- Department of Anaesthesiology IMIM-Hospital del Mar-UAB, Barcelona, Spain
| | | | - Antonio Montes
- Department of Anaesthesiology IMIM-Hospital del Mar-UAB, Barcelona, Spain
| |
Collapse
|
13
|
Inyang KE, George SR, Laumet G. The µ-δ opioid heteromer masks latent pain sensitization in neuropathic and inflammatory pain in male and female mice. Brain Res 2021; 1756:147298. [PMID: 33516809 DOI: 10.1016/j.brainres.2021.147298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 01/16/2023]
Abstract
The episodic nature of chronic pain can be studied in the rodent model of latent pain sensitization. After remission, central sensitization is opposed by activation of opioid receptors. At the behavioral level, latent pain sensitization is unmasked when pain hypersensitivity is reinstated by opioid receptor (OR) antagonism. Previous studies have focused on inflammatory pain and male rodents. Whether latent pain sensitization occurs in models of chemotherapy-induced neuropathic pain in female and male mice is unknown. The first aim of this study was to investigate whether μ- and δ-OR suppress latent pain sensitization in our model of chemotherapy-induced neuropathic pain in both sexes. Mounting evidence suggests that μ-and δ-ORs form a heteromer and that the heteromer modulates pain sensitivity. Potential implications of the μ-δ OR heteromer in latent pain sensitization have not been fully explored due to a lack of tools to effectively modulate the heteromer. To specifically target the μ-δ OR heteromer, we used a specific interfering peptide blocking the heteromerization. The second aim of this study was to investigate whether disruption of the μ-δOR heteromer, after remission, reinstates pain hypersensitivity. After remission from cisplatin-induced neuropathic pain, antagonism of µ-OR and δOR reinstates pain hypersensitivity in both sexes. After remission from cisplatin-induced neuropathic pain and postoperative pain, disruption of the μ-δOR heteromer reinstates pain hypersensitivity in both sexes. Taken together our findings suggest that the μ-δOR heteromer plays a crucial role in remission in various pain models and may represent a novel therapeutic target to prevent the relapse to pain and the transition to chronic pain.
Collapse
Affiliation(s)
| | - Susan R George
- Department of Medicine and Pharmacology, University of Toronto, Toronto, Ontario, Canada
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
14
|
Chen W, McRoberts JA, Ennes HS, Marvizon JC. cAMP signaling through protein kinase A and Epac2 induces substance P release in the rat spinal cord. Neuropharmacology 2021; 189:108533. [PMID: 33744339 DOI: 10.1016/j.neuropharm.2021.108533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 11/18/2022]
Abstract
Using neurokinin 1 receptor (NK1R) internalization to measure of substance P release in rat spinal cord slices, we found that it was induced by the adenylyl cyclase (AC) activator forskolin, by the protein kinase A (PKA) activators 6-Bnz-cAMP and 8-Br-cAMP, and by the activator of exchange protein activated by cAMP (Epac) 8-pCPT-2-O-Me-cAMP (CPTOMe-cAMP). Conversely, AC and PKA inhibitors decreased substance P release induced by electrical stimulation of the dorsal root. Therefore, the cAMP signaling pathway mediates substance P release in the dorsal horn. The effects of forskolin and 6-Bnz-cAMP were not additive with NMDA-induced substance P release and were decreased by the NMDA receptor blocker MK-801. In cultured dorsal horn neurons, forskolin increased NMDA-induced Ca2+ entry and the phosphorylation of the NR1 and NR2B subunits of the NMDA receptor. Therefore, cAMP-induced substance P release is mediated by the activating phosphorylation by PKA of NMDA receptors. Voltage-gated Ca2+ channels, but not by TRPV1 or TRPA1, also contributed to cAMP-induced substance P release. Activation of PKA was required for the effects of forskolin and the three cAMP analogs. Epac2 contributed to the effects of forskolin and CPTOMe-cAMP, signaling through a Raf - mitogen-activated protein kinase pathway to activate Ca2+ channels. Epac1 inhibitors induced NK1R internalization independently of substance P release. In rats with latent sensitization to pain, the effect of 6-Bnz-cAMP was unchanged, whereas the effect of forskolin was decreased due to the loss of the stimulatory effect of Epac2. Hence, substance P release induced by cAMP decreases during pain hypersensitivity.
Collapse
Affiliation(s)
- Wenling Chen
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - James A McRoberts
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Helena S Ennes
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Juan Carlos Marvizon
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA.
| |
Collapse
|
15
|
Zhang Y, Jeske NA. GRK2 Dictates a Functional Switch of the Peripheral Mu-Opioid Receptor. ACS Chem Neurosci 2020; 11:4376-4386. [PMID: 33174729 DOI: 10.1021/acschemneuro.0c00622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The peripheral mu-opioid receptor (MOR) has been recognized as a potential target to provide safer analgesia with reduced central side effects. Although analgesic incompetence of the peripheral MOR in the absence of inflammation was initially identified more than a decade ago, there has been very limited investigation into the underlying signaling mechanisms. Here we identify that G protein-coupled receptor kinase 2 (GRK2) constitutively interacts with the MOR in peripheral sensory neurons to suppress peripheral MOR activity. Brief exposure to bradykinin (BK) causes uncoupling of GRK2 from the MOR and subsequent restoration of MOR functionality in dorsal root ganglion (DRG) neurons. Interestingly, prolonged BK treatment induces constitutive activation of the MOR through a mechanism that involves protein kinase C (PKC) activation. After silencing Raf kinase inhibitory protein (RKIP) by RNA interference, BK-induced constitutive MOR activation is completely abrogated, which agrees with previous findings that BK activates PKC signaling to initiate GRK2 sequestration by RKIP. Furthermore, we demonstrate that constitutive, peripheral MOR activity requires GRK2 uncoupling and that the FDA-approved SSRI paroxetine promotes this state of uncoupling. Collectively, these results indicate that GRK2 tightly regulates MOR functional states and controls constitutive MOR activity in peripheral sensory neurons, supporting the potential for targeting the kinase to provide safer analgesia.
Collapse
|
16
|
Springborg AD, Jensen EK, Kreilgaard M, Petersen MA, Papathanasiou T, Lund TM, Taylor BK, Werner MU. High-dose naloxone: Effects by late administration on pain and hyperalgesia following a human heat injury model. A randomized, double-blind, placebo-controlled, crossover trial with an enriched enrollment design. PLoS One 2020; 15:e0242169. [PMID: 33180816 PMCID: PMC7660513 DOI: 10.1371/journal.pone.0242169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/27/2020] [Indexed: 11/18/2022] Open
Abstract
Severe chronic postsurgical pain has a prevalence of 4–10% in the surgical population. The underlying nociceptive mechanisms have not been well characterized. Following the late resolution phase of an inflammatory injury, high-dose μ-opioid-receptor inverse agonists reinstate hypersensitivity to nociceptive stimuli. This unmasking of latent pain sensitization has been a consistent finding in rodents while only observed in a limited number of human volunteers. Latent sensitization could be a potential triggering venue in chronic postsurgical pain. The objective of the present trial was in detail to examine the association between injury-induced secondary hyperalgesia and naloxone-induced unmasking of latent sensitization. Healthy volunteers (n = 80) received a cutaneous heat injury (47°C, 420 s, 12.5 cm2). Baseline secondary hyperalgesia areas were assessed 1 h post-injury. Utilizing an enriched enrollment design, subjects with a magnitude of secondary hyperalgesia areas in the upper quartile (‘high-sensitizers’ [n = 20]) and the lower quartile (‘low-sensitizers’ [n = 20]) were selected for further study. In four consecutive experimental sessions (Sessions 1 to 4), the subjects at two sessions (Sessions 1 and 3) received a cutaneous heat injury followed 168 h later (Sessions 2 and 4) by a three-step target-controlled intravenous infusion of naloxone (3.25 mg/kg), or normal saline. Assessments of secondary hyperalgesia areas were made immediately before and stepwise during the infusions. Simple univariate statistics revealed no significant differences in secondary hyperalgesia areas between naloxone and placebo treatments (P = 0.215), or between ‘high-sensitizers’ and ‘low-sensitizers’ (P = 0.757). In a mixed-effects model, secondary hyperalgesia areas were significantly larger following naloxone as compared to placebo for ‘high-sensitizers’ (P < 0.001), but not ‘low-sensitizers’ (P = 0.651). Although we could not unequivocally demonstrate naloxone-induced reinstatement of heat injury-induced hyperalgesia, further studies in clinical postsurgical pain models are warranted.
Collapse
Affiliation(s)
| | | | - Mads Kreilgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Aagaard Petersen
- The Research Unit, Department of Palliative Medicine, Copenhagen University Hospitals, Copenhagen, Denmark
| | - Theodoros Papathanasiou
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Meldgaard Lund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bradley Kenneth Taylor
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Pittsburgh Project to End Opioid Misuse, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mads Utke Werner
- Neuroscience Center, Copenhagen University Hospitals, Copenhagen, Denmark
- Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
17
|
Chen W, Marvizón JC. A Src family kinase maintains latent sensitization in rats, a model of inflammatory and neuropathic pain. Brain Res 2020; 1746:146999. [PMID: 32579948 PMCID: PMC10866137 DOI: 10.1016/j.brainres.2020.146999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/29/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Latent sensitization is a long-term model of chronic pain in which hyperalgesia is continuously suppressed by opioid receptors, as demonstrated by the induction of mechanical allodynia by opioid antagonists. Different intracellular signals may mediate the initiation, maintenance and expression of latent sensitization. Our criterion for the involvement of a signal in the maintenance of latent sensitization is that inhibitors should permanently eliminate the allodynia produced by an opioid antagonist. We hypothesized that Src family kinases (SFKs) maintain latent sensitization and tested this hypothesis by inducing latent sensitization in rats with complete Freund's adjuvant (CFA) or spared nerve injury. After measures of mechanical allodynia returned to baseline, vehicle or the SFK inhibitor PP2 were injected intrathecally. The opioid antagonist naltrexone injected intrathecally 15 min later produced allodynia in control rats but not in rats injected with PP2. Vehicle or PP2 were injected daily for two more days and naltrexone was injected five days later. Again, naltrexone induced allodynia in the control rats but not in the rats injected with PP2. Results were similar when latent sensitization was induced with CFA or spared nerve injury. We concluded that an SFK, likely Fyn, maintains latent sensitization induced by inflammation or nerve injury.
Collapse
Affiliation(s)
- Wenling Chen
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Juan Carlos Marvizón
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
18
|
Chen W, Marvizon JC. Neurokinin 1 receptor activation in the rat spinal cord maintains latent sensitization, a model of inflammatory and neuropathic chronic pain. Neuropharmacology 2020; 177:108253. [PMID: 32736088 PMCID: PMC10863619 DOI: 10.1016/j.neuropharm.2020.108253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022]
Abstract
Latent sensitization is a model of chronic pain in which a persistent state of pain hypersensitivity is suppressed by opioid receptors, as evidenced by the ability of opioid antagonists to induce a period of mechanical allodynia. Our objective was to determine if substance P and its neurokinin 1 receptor (NK1R) mediate the maintenance of latent sensitization. Latent sensitization was induced by injecting rats in the hindpaw with complete Freund's adjuvant (CFA), or by tibial spared nerve injury (SNI). When responses to von Frey filaments returned to baseline (day 28), the rats were injected intrathecally with saline or the NK1R antagonist RP67580, followed 15 min later by intrathecal naltrexone. In both pain models, the saline-injected rats developed allodynia for 2 h after naltrexone, but not the RP67580-injected rats. Saline or RP67580 were injected daily for two more days. Five days later (day 35), naltrexone was injected intrathecally. Again, the saline-injected rats, but not the RP67580-injected rats, developed allodynia in response to naltrexone. To determine if there is sustained activation of NK1Rs during latent sensitization, NK1R internalization was measured in lamina I neurons in rats injected in the paw with saline or CFA, and then injected intrathecally with saline or naltrexone on day 28. The rats injected with CFA had a small amount of NK1R internalization that was significantly higher than in the saline-injected rats. Naltrexone increased NK1R internalization in the CFA-injected rats but nor in the saline-injected rats. Therefore, sustained activation of NK1Rs maintains pain hypersensitivity during latent sensitization.
Collapse
Affiliation(s)
- Wenling Chen
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Juan Carlos Marvizon
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA.
| |
Collapse
|
19
|
Sham surgeries for central and peripheral neural injuries persistently enhance pain-avoidance behavior as revealed by an operant conflict test. Pain 2020; 160:2440-2455. [PMID: 31323014 DOI: 10.1097/j.pain.0000000000001642] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Studies using rodent models of neuropathic pain use sham surgery control procedures that cause deep tissue damage. Sham surgeries would thus be expected to induce potentially long-lasting postsurgical pain, but little evidence for such pain has been reported. Operant tests of voluntary behavior can reveal negative motivational and cognitive aspects of pain that may provide sensitive tools for detecting pain-related alterations. In a previously described operant mechanical conflict test involving lengthy familiarization and training, rodents freely choose to either escape from a brightly lit chamber by crossing sharp probes or refuse to cross. Here, we describe a brief (2-day) mechanical conflict protocol that exploits rats' innate exploratory response to a novel environment to detect persistently enhanced pain-avoidance behavior after sham surgeries for 2 neural injury models: thoracic spinal cord injury and chronic constriction injury of the sciatic nerve. Pitting the combined motivations to avoid the bright light and to explore the novel device against pain from crossing noxious probes disclosed a conflicting, hyperalgesia-related reluctance to repeatedly cross the probes after injury. Rats receiving standard sham surgeries demonstrated enhanced pain-like avoidance behavior compared with naive controls, and this behavior was similar to that of corresponding chronic constriction injury or spinal cord injury rats weeks or months after injury. In the case of sham surgery for spinal cord injury, video analysis of voluntary exploratory behavior directed at the probes revealed enhanced forepaw withdrawal responses. These findings have important implications for preclinical investigations into behavioral alterations and physiological mechanisms associated with postsurgical and neuropathic pain.
Collapse
|
20
|
Assessing the Effects of Parthenolide on Inflammation, Bone Loss, and Glial Cells within a Collagen Antibody-Induced Arthritis Mouse Model. Mediators Inflamm 2020; 2020:6245798. [PMID: 32189995 PMCID: PMC7073477 DOI: 10.1155/2020/6245798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/16/2019] [Accepted: 01/27/2020] [Indexed: 12/21/2022] Open
Abstract
Rheumatoid arthritis is characterised by a chronic inflammatory response resulting in destruction of the joint and significant pain. Although a range of treatments are available to control disease activity in RA, bone destruction and joint pain exist despite suppression of inflammation. This study is aimed at assessing the effects of parthenolide (PAR) on paw inflammation, bone destruction, and pain-like behaviour in a mild collagen antibody-induced arthritis (CAIA) mouse model. CAIA was induced in BALB/c mice and treated daily with 1 mg/kg or 4 mg/kg PAR. Clinical paw inflammation was scored daily, and mechanical hypersensitivity was assessed on alternate days. At end point, bone volume and swelling in the paws were assessed using micro-CT. Paw tissue sections were assessed for inflammation and pre-/osteoclast-like cells. The lumbar spinal cord and the periaqueductal grey (PAG) and rostral ventromedulla (RVM) regions of the brain were stained for glial fibrillary acidic protein (GFAP) and ionised calcium-binding adaptor molecule 1 (IBA1) to assess for glial reactivity. Paw scores increased in CAIA mice from days 5-10 and were reduced with 1 mg/kg and 4 mg/kg PAR on days 8-10. Osteoclast-like cells on the bone surface of the radiocarpal joint and within the soft tissue of the hind paw were significantly lower following PAR treatment (p < 0.005). GFAP- and IBA1-positive cells in the PAG and RVM were significantly lower following treatment with 1 mg/kg (p < 0.0001 and p = 0.0004, respectively) and 4 mg/kg PAR (p < 0.0001 and p = 0.001, respectively). In the lumbar spinal cord, IBA1-positive cells were significantly lower in CAIA mice treated with 4 mg/kg PAR (p = 0.001). The findings indicate a suppressive effect of both low- and moderate-dose PAR on paw inflammation, osteoclast presence, and glial cell reactivity in a mild CAIA mouse model.
Collapse
|
21
|
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
|
22
|
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
|
23
|
Feehan AK, Zadina JE. Morphine immunomodulation prolongs inflammatory and postoperative pain while the novel analgesic ZH853 accelerates recovery and protects against latent sensitization. J Neuroinflammation 2019; 16:100. [PMID: 31109346 PMCID: PMC6528320 DOI: 10.1186/s12974-019-1480-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/15/2019] [Indexed: 01/12/2023] Open
Abstract
Background Numerous studies have identified the proinflammatory, pronociceptive effects of morphine which ultimately exacerbate pain. Our novel endomorphin analog ZH853 does not produce proinflammatory effects on its own and gives potent, long-lasting analgesia. This study investigates whether ZH853’s lack of interaction with the neuroimmune system reduces the risk of prolonged pain. Methods Adult male Sprague-Dawley rats were subjected to one of two treatment paradigms. Either (1) chronic pain followed by chronic treatment with morphine, ZH853 or vehicle, or (2) chronic drug administered prior to pain induction. Complete Freund’s adjuvant (CFA) was injected or paw incision surgery was performed on the left hind plantar foot pad. Drugs were administered through Alzet osmotic minipumps at a rate of 1 μl/h for 5 days at appropriate doses based on prior experiments. Animals were tested for mechanical allodynia and thermal hyperalgesia using von Frey filaments and the Hargreaves apparatus, respectively. Additionally, several gait parameters were measured using the CatWalk XT. When all animals had recovered from pain, 1 mg/kg of naltrexone was administered to test for development of latent sensitization (LS). A second set of animals was used to investigate dorsal horn inflammation following CFA and drug treatment. ANOVAs were used to assess differences between drug treatment groups. Results As expected, morphine increased and prolonged pain in all experiments compared to vehicle treatment. However, ZH853 treatment reduced the overall time spent in pain and the severity of pain scores compared to morphine. ZH853 not only reduced inflammation versus morphine treatment but also, in some instances, acted as an anti-inflammatory drug compared to vehicle treatment. Finally, ZH853 prevented the development of LS while vehicle- and morphine-treated animals showed robust relapse to pain. Conclusions ZH853 has a favorable side effect profile versus morphine and provides superior analgesia in a number of pain states. We now know that chronic use of this compound reduces time spent in a chronic pain state, the opposite of common opioids like morphine, and reduces the risk of LS, making ZH853 an excellent candidate for clinical development in humans for inflammatory and postoperative pain.
Collapse
Affiliation(s)
- Amy K Feehan
- Brain Institute, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - James E Zadina
- SE LA Veterans Health Care System, New Orleans, LA, 70119, USA. .,Dept. of Medicine, Neuroscience Lab #8516, 1430 Tulane Avenue, New Orleans, LA, 70112, USA.
| |
Collapse
|
24
|
Chen W, Taché Y, Marvizón JC. Corticotropin-Releasing Factor in the Brain and Blocking Spinal Descending Signals Induce Hyperalgesia in the Latent Sensitization Model of Chronic Pain. Neuroscience 2019; 381:149-158. [PMID: 29776484 DOI: 10.1016/j.neuroscience.2018.03.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 02/21/2018] [Accepted: 03/16/2018] [Indexed: 12/25/2022]
Abstract
Latent sensitization is a model of chronic pain in which an injury triggers a period of hyperalgesia followed by an apparent recovery, but in which pain sensitization persists but is suppressed by opioid and adrenergic receptors. One important characteristic of latent sensitization is that hyperalgesia can be triggered by acute stress. To determine whether the effect of stress is mimicked by the activation of corticotropin-releasing factor (CRF) signaling in the brain, rats with latent sensitization induced by injecting complete Freund's adjuvant (CFA, 50 μl) in one hind paw were given an intracerebroventricular (i.c.v.) injection of CRF. The i.c.v. injection of CRF (0.6 μg, 10 μl), but not saline, induced bilateral mechanical hyperalgesia in rats with latent sensitization. In contrast, CRF i.c.v. did not induce hyperalgesia in rats without latent sensitization (injected with saline in the hind paw). To determine whether descending pain inhibition mediates the suppression of hyperalgesia in latent sensitization, rats with CFA-induced latent sensitization received an intrathecal injection of lidocaine (10%, 1 μl) at the cervical-thoracic spinal cord to produce a spinal block. Lidocaine-injected rats, but not rats injected intrathecally with saline, developed bilateral mechanical hyperalgesia. Intrathecal lidocaine did not induce hyperalgesia in rats without latent sensitization (injected with saline in the hind paw). These results show that i.c.v. CRF mimicked the hyperalgesic response triggered by stress during latent sensitization, possibly by blocking inhibitory spinal descending signals that suppress hyperalgesia.
Collapse
Affiliation(s)
- Wenling Chen
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States.
| | - Yvette Taché
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States.
| | - Juan Carlos Marvizón
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
25
|
Severino A, Chen W, Hakimian JK, Kieffer BL, Gaveriaux-Ruff C, Walwyn W, Marvizón JCG. Mu-opioid receptors in nociceptive afferents produce a sustained suppression of hyperalgesia in chronic pain. Pain 2019; 159:1607-1620. [PMID: 29677019 DOI: 10.1097/j.pain.0000000000001247] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The latent sensitization model of chronic pain reveals that recovery from some types of long-term hyperalgesia is an altered state in which nociceptive sensitization persists but is suppressed by the ongoing activity of analgesic receptors such as μ-opioid receptors (MORs). To determine whether these MORs are the ones present in nociceptive afferents, we bred mice expressing Cre-recombinase under the Nav1.8 channel promoter (Nav1.8cre) with MOR-floxed mice (flMOR). These Nav1.8cre/flMOR mice had reduced MOR expression in primary afferents, as revealed by quantitative PCR, in situ hybridization, and immunofluorescence colocalization with the neuropeptide calcitonin gene-related peptide. We then studied the recovery from chronic pain of these mice and their flMOR littermates. When Nav1.8cre/flMOR mice were injected in the paw with complete Freund adjuvant they developed mechanical hyperalgesia that persisted for more than 2 months, whereas the responses of flMOR mice returned to baseline after 3 weeks. We then used the inverse agonist naltrexone to assess ongoing MOR activity. Naltrexone produced a robust reinstatement of hyperalgesia in control flMOR mice, but produced no effect in the Nav1.8/flMOR males and a weak reinstatement of hyperalgesia in Nav1.8/flMOR females. Naltrexone also reinstated swelling of the hind paw in flMOR mice and female Nav1.8cre/flMOR mice, but not male Nav1.8cre/flMOR mice. The MOR agonist DAMGO inhibited substance P release in flMOR mice but not Nav1.8cre/flMOR mice, demonstrating a loss of MOR function at the central terminals of primary afferents. We conclude that MORs in nociceptive afferents mediate an ongoing suppression of hyperalgesia to produce remission from chronic pain.
Collapse
Affiliation(s)
- Amie Severino
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Wenling Chen
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Joshua K Hakimian
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Brigitte L Kieffer
- Department of Psychiatry, Douglas Mental Health Institute, McGill University, Montreal, QC, Canada
| | - Claire Gaveriaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Wendy Walwyn
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Juan Carlos G Marvizón
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
26
|
Simultaneous quantification of high-dose naloxone and naloxone-3-β-d-glucuronide in human plasma by UHPLC-MS/MS. Bioanalysis 2019; 11:165-173. [PMID: 30661370 DOI: 10.4155/bio-2018-0134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: High-dose administration of the μ-opioid receptor inverse agonist naloxone (NX), has previously been demonstrated to reinstate nocifensive behavior in the late phase of inflammatory injuries. However, no current analytical methods can provide pharmacokinetic insight into the pharmacodynamic response of high-dose administration of NX. Materials & methods: Based on protein precipitation using 50 μl human plasma, NX and naloxone-β-d-glucuronide (NXG) was analysed by UHPLC-MS/MS with 6 min cycle time. Results: A method for quantification of high-dose administered NX and NXG was developed and validated with intra- and interday precision and accuracy within ≤8.5% relative standard deviation (RSD) and -1.2-5.5% relative error (RE) for NX and ≤9.6% RSD and 0.6-6.5% RE for NXG. The method show excellent internal standard corrected matrix effects. Conclusion: A rapid UHPLC-MS/MS method was developed for quantification of NX and NXG in human plasma within 10-4000 ng/ml.
Collapse
|
27
|
High-dose naloxone, an experimental tool uncovering latent sensitisation: pharmacokinetics in humans. Br J Anaesth 2019; 123:e204-e214. [PMID: 30915992 DOI: 10.1016/j.bja.2018.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Naloxone, an opioid receptor antagonist, is used as a pharmacological tool to detect tonic endogenous activation of opioid receptors in experimental pain models. We describe a pharmacokinetic model linking naloxone pharmacokinetics to its main metabolite after high-dose naloxone infusion. METHODS Eight healthy volunteers received a three-stage stepwise high-dose i.v. naloxone infusion (total dose 3.25 mg kg-1). Naloxone and naloxone-3-glucuronide (N3G) plasma concentrations were sampled from infusion onset to 334 min after infusion discontinuation. Pharmacokinetic analysis was performed using non-linear mixed effect models (NONMEM). The predictive performances of Dowling's and Yassen's models were evaluated, and target-controlled infusion simulations were performed. RESULTS Three- and two-compartment disposition models with linear elimination kinetics described the naloxone and N3G concentration time-courses, respectively. Two covariate models were developed: simple (weight proportional) and complex (with the shallow peripheral volume of distribution linearly increasing with body weight). The median prediction error (MDPE) and wobble for Dowling's model were -32.5% and 33.4%, respectively. For Yassen's model, the MDPE and wobble were 1.2% and 19.9%, respectively. CONCLUSIONS A parent-metabolite pharmacokinetic model was developed for naloxone and N3G after high-dose naloxone infusion. No saturable pharmacokinetics were observed. Whereas Dowling's model was inaccurate and over-predicted naloxone concentrations, Yassen's model accurately predicted naloxone pharmacokinetics. The newly developed covariate models may be used for high-dose TCI-naloxone for experimental and clinical practice. CLINICAL TRIALS REGISTRATION NCT01992146.
Collapse
|
28
|
Taylor BK, Sinha GP, Donahue RR, Grachen CM, Morón JA, Doolen S. Opioid receptors inhibit the spinal AMPA receptor Ca 2+ permeability that mediates latent pain sensitization. Exp Neurol 2019; 314:58-66. [PMID: 30660616 DOI: 10.1016/j.expneurol.2019.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/24/2018] [Accepted: 01/05/2019] [Indexed: 01/02/2023]
Abstract
Acute inflammation induces sensitization of nociceptive neurons and triggers the accumulation of calcium permeable (CP) α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) in the dorsal horn of the spinal cord. This coincides with behavioral signs of acute inflammatory pain, but whether CP-AMPARs contribute to chronic pain remains unclear. To evaluate this question, we first constructed current-voltage (IV) curves of C-fiber stimulus-evoked, AMPAR-mediated EPSCs in lamina II to test for inward rectification, a key characteristic of CP-AMPARs. We found that the intraplantar injection of complete Freund's adjuvant (CFA) induced an inward rectification at 3 d that persisted to 21 d after injury. Furthermore, the CP- AMPAR antagonist IEM-1460 (50 μM) inhibited AMPAR-evoked Ca2+ transients 21d after injury but had no effect in uninflamed mice. We then used a model of long-lasting vulnerability for chronic pain that is determined by the balance between latent central sensitization (LCS) and mu opioid receptor constitutive activity (MORCA). When administered 21 d after the intraplantar injection of CFA, intrathecal administration of the MORCA inverse agonist naltrexone (NTX, 1 μg, i.t.) reinstated mechanical hypersensitivity, and superfusion of spinal cord slices with NTX (10 μM) increased the peak amplitude of AMPAR-evoked Ca2+ transients in lamina II neurons. The CP-AMPAR antagonist naspm (0-10 nmol, i.t.) inhibited these NTX-induced increases in mechanical hypersensitivity. NTX had no effect in uninflamed mice. Subsequent western blot analysis of the postsynaptic density membrane fraction from lumbar dorsal horn revealed that CFA increased GluA1 expression at 2 d and GluA4 expression at both 2 and 21 d post-injury, indicating that not just the GluA1 subunit, but also the GluA4 subunit, contributes to the expression of CP-AMPARs and synaptic strength during hyperalgesia. GluA2 expression increased at 21 d, an unexpected result that requires further study. We conclude that after tissue injury, dorsal horn AMPARs retain a Ca2+ permeability that underlies LCS. Because of their effectiveness in reducing naltrexone-induced reinstatement of hyperalgesia and potentiation of AMPAR-evoked Ca2+ signals, CP-AMPAR inhibitors are a promising class of agents for the treatment of chronic inflammatory pain.
Collapse
Affiliation(s)
- Bradley K Taylor
- Department of Anesthesiology, Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, 200 Lothrop St. Pittsburgh, PA 15213, USA; Department of Physiology, University of Kentucky School of Medicine, 800 Rose, St. Lexington, KY 40536-0298, USA.
| | - Ghanshyam P Sinha
- Department of Anesthesiology, Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, 200 Lothrop St. Pittsburgh, PA 15213, USA; Department of Physiology, University of Kentucky School of Medicine, 800 Rose, St. Lexington, KY 40536-0298, USA.
| | - Renee R Donahue
- Department of Physiology, University of Kentucky School of Medicine, 800 Rose, St. Lexington, KY 40536-0298, USA.
| | - Carolyn M Grachen
- Department of Anesthesiology, Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, 200 Lothrop St. Pittsburgh, PA 15213, USA; Department of Physiology, University of Kentucky School of Medicine, 800 Rose, St. Lexington, KY 40536-0298, USA.
| | - Jose A Morón
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, 600 South Euclid, St Louis, MO 63110, USA.
| | - Suzanne Doolen
- Department of Anesthesiology, Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, 200 Lothrop St. Pittsburgh, PA 15213, USA; Department of Physiology, University of Kentucky School of Medicine, 800 Rose, St. Lexington, KY 40536-0298, USA.
| |
Collapse
|
29
|
Abstract
Abstract
The development of chronic pain is considered a major complication after surgery. Basic science research in animal models helps us understand the transition from acute to chronic pain by identifying the numerous molecular and cellular changes that occur in the peripheral and central nervous systems. It is now well recognized that inflammation and nerve injury lead to long-term synaptic plasticity that amplifies and also maintains pain signaling, a phenomenon referred to as pain sensitization. In the context of surgery in humans, pain sensitization is both responsible for an increase in postoperative pain via the expression of wound hyperalgesia and considered a critical factor for the development of persistent postsurgical pain. Using specific drugs that block the processes of pain sensitization reduces postoperative pain and prevents the development of persistent postoperative pain. This narrative review of the literature describes clinical investigations evaluating different preventative pharmacologic strategies that are routinely used by anesthesiologists in their daily clinical practices for preventing persistent postoperative pain. Nevertheless, further efforts are needed in both basic and clinical science research to identify preclinical models and novel therapeutics targets. There remains a need for more patient numbers in clinical research, for more reliable data, and for the development of the safest and the most effective strategies to limit the incidence of persistent postoperative pain.
Collapse
|
30
|
Abstract
We previously developed a model of opioid-induced neuroplasticity in the peripheral terminal of the nociceptor that could contribute to opioid-induced hyperalgesia, type II hyperalgesic priming. Repeated administration of mu-opioid receptor (MOR) agonists, such as DAMGO, at the peripheral terminal of the nociceptor, induces long-lasting plasticity expressed, prototypically as opioid-induced hyperalgesia and prolongation of prostaglandin E2-induced hyperalgesia. In this study, we evaluated the mechanisms involved in the maintenance of type II priming. Opioid receptor antagonist, naloxone, induced hyperalgesia in DAMGO-primed paws. When repeatedly injected, naloxone-induced hyperalgesia, and hyperalgesic priming, supporting the suggestion that maintenance of priming involves changes in MOR signaling. However, the knockdown of MOR with oligodeoxynucleotide antisense did not reverse priming. Mitogen-activated protein kinase and focal adhesion kinase, which are involved in the Src signaling pathway, previously implicated in type II priming, also inhibited the expression, but not maintenance of priming. However, when Src and mitogen-activated protein kinase inhibitors were coadministered, type II priming was reversed, in male rats. A second model of priming, latent sensitization, induced by complete Freund's adjuvant was also reversed, in males. In females, the inhibitor combination was only able to inhibit the expression and maintenance of DAMGO-induced priming when knockdown of G-protein-coupled estrogen receptor 30 (GPR30) in the nociceptor was performed. These findings demonstrate that the maintenance of DAMGO-induced type II priming, and latent sensitization is mediated by an interaction between, Src and MAP kinases, which in females is GPR30 dependent.
Collapse
|
31
|
Chen W, Ennes HS, McRoberts JA, Marvizón JC. Mechanisms of μ-opioid receptor inhibition of NMDA receptor-induced substance P release in the rat spinal cord. Neuropharmacology 2017; 128:255-268. [PMID: 29042318 DOI: 10.1016/j.neuropharm.2017.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/21/2017] [Accepted: 10/11/2017] [Indexed: 01/17/2023]
Abstract
The interaction between NMDA receptors and μ-opioid receptors in primary afferent terminals was studied by using NMDA to induce substance P release, measured as neurokinin 1 receptor internalization. In rat spinal cord slices, the μ-opioid receptor agonists morphine, DAMGO and endomorphin-2 inhibited NMDA-induced substance P release, whereas the antagonist CTAP right-shifted the concentration response of DAMGO. In vivo, substance P release induced by intrathecal NMDA after priming with BDNF was inhibited by DAMGO. ω-Conotoxins MVIIC and GVIA inhibited about half of the NMDA-induced substance P release, showing that it was partially mediated by the opening of voltage-gated calcium (Cav) channels. In contrast, DAMGO or ω-conotoxins did not inhibit capsaicin-induced substance P release. In cultured DRG neurons, DAMGO but not ω-conotoxin inhibited NMDA-induced increases in intracellular calcium, indicating that μ-opioid receptors can inhibit NMDA receptor function by mechanisms other than inactivation of Cav channels. Moreover, DAMGO decreased the ω-conotoxin-insensitive component of the substance P release. Potent inhibition by ifenprodil showed that these NMDA receptors have the NR2B subunit. Activators of adenylyl cyclase and protein kinase A (PKA) induced substance P release and this was decreased by the NMDA receptor blocker MK-801 and by DAMGO. Conversely, inhibitors of adenylyl cyclase and PKA, but not of protein kinase C, decreased NMDA-induced substance P release. Hence, these NMDA receptors are positively modulated by the adenylyl cyclase-PKA pathway, which is inhibited by μ-opioid receptors. In conclusion, μ-opioid receptors inhibit NMDA receptor-induced substance P release through Cav channel inactivation and adenylyl cyclase inhibition.
Collapse
Affiliation(s)
- Wenling Chen
- Veteran Affairs Greater Los Angeles Healthcare System, 11310 Wilshire Blvd., Building 115, Los Angeles, CA 90073, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, 900 Veterans Ave., Warren Hall Building, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Helena S Ennes
- Vatche and Tamar Manoukian Division of Digestive Diseases, 900 Veterans Ave., Warren Hall Building, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - James A McRoberts
- Vatche and Tamar Manoukian Division of Digestive Diseases, 900 Veterans Ave., Warren Hall Building, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Juan Carlos Marvizón
- Veteran Affairs Greater Los Angeles Healthcare System, 11310 Wilshire Blvd., Building 115, Los Angeles, CA 90073, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, 900 Veterans Ave., Warren Hall Building, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
32
|
Postoperative Pain Management in Spanish Hospitals: A Cohort Study Using the PAIN-OUT Registry. THE JOURNAL OF PAIN 2017; 18:1237-1252. [DOI: 10.1016/j.jpain.2017.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 05/31/2017] [Indexed: 12/16/2022]
|
33
|
Romero A, García-Carmona JA, Laorden ML, Puig MM. Role of CRF1 receptor in post-incisional plasma extravasation and nociceptive responses in mice. Toxicol Appl Pharmacol 2017; 332:121-128. [DOI: 10.1016/j.taap.2017.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/12/2017] [Accepted: 04/19/2017] [Indexed: 12/30/2022]
|
34
|
Nakamoto K, Aizawa F, Miyagi K, Yamashita T, Mankura M, Koyama Y, Kasuya F, Hirasawa A, Kurihara T, Miyata A, Tokuyama S. Dysfunctional GPR40/FFAR1 signaling exacerbates pain behavior in mice. PLoS One 2017; 12:e0180610. [PMID: 28723961 PMCID: PMC5516985 DOI: 10.1371/journal.pone.0180610] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 06/19/2017] [Indexed: 11/22/2022] Open
Abstract
We previously showed that activation of G protein-coupled receptor 40/free fatty acid receptor 1 (GPR40/FFAR1) signaling modulates descending inhibition of pain. In this study, we investigated the involvement of fatty acid-GPR40/FFAR1 signaling in the transition from acute to chronic pain. We used GPR40/FFAR1-knockout (GPR40KO) mice and wild-type (WT) mice. A plantar incision was performed, and mechanical allodynia and thermal hyperalgesia were evaluated with a von Frey filament test and plantar test, respectively. Immunohistochemistry was used to localize GPR40/FFAR1, and the levels of free fatty acids in the hypothalamus were analyzed with liquid chromatography-tandem mass spectrometry. The repeated administration of GW1100, a GPR40/FFAR1 antagonist, exacerbated the incision-induced mechanical allodynia and significantly increased the levels of phosphorylated extracellular signal-regulated kinase in the spinal cord after low-threshold touch stimulation in the mice compared to vehicle-treated mice. The levels of long-chain free fatty acids, such as docosahexaenoic acid, oleic acid, and palmitate, which are GPR40/FFAR1 agonists, were significantly increased in the hypothalamus two days after the surgery compared to levels in the sham group. Furthermore, the incision-induced mechanical allodynia was exacerbated in the GPR40KO mice compared to the WT mice, while the response in the plantar test was not changed. These findings suggested that dysfunction of the GPR40/FFAR1 signaling pathway altered the endogenous pain control system and that this dysfunction might be associated with the development of chronic pain.
Collapse
Affiliation(s)
- Kazuo Nakamoto
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Japan
| | - Fuka Aizawa
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Japan
| | - Kei Miyagi
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Japan
| | - Takuya Yamashita
- Biochemical Toxicology Laboratory, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Japan
| | - Mitsumasa Mankura
- Faculty of Food Culture, Kurashiki Sakuyo University, Kurashiki, Okayama, Japan
| | - Yutaka Koyama
- Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Fumiyo Kasuya
- Biochemical Toxicology Laboratory, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Japan
| | - Akira Hirasawa
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Shogo Tokuyama
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Japan
- * E-mail:
| |
Collapse
|
35
|
Gu W, Zhang W, Lei Y, Cui Y, Chu S, Gu X, Ma Z. Activation of spinal alpha-7 nicotinic acetylcholine receptor shortens the duration of remifentanil-induced postoperative hyperalgesia by upregulating KCC2 in the spinal dorsal horn in rats. Mol Pain 2017; 13:1744806917704769. [PMID: 28425312 PMCID: PMC6997724 DOI: 10.1177/1744806917704769] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 02/28/2017] [Accepted: 03/15/2017] [Indexed: 12/15/2022] Open
Abstract
Background Accumulating evidence has shown that the signal from spinal brain-derived neurotrophic factor/tyrosine receptor kinase B-K+-Cl- cotransporter-2 plays a critical role in the process of pain hypersensitivity. The activation of alpha-7 nicotinic acetylcholine receptors could have an analgesic effect on remifentanil-induced postoperative hyperalgesia. Nevertheless, whether intrathecal administration of PNU-120596, an alpha-7 nicotinic acetylcholine receptors selective type II positive allosteric modulator, before surgery could affect the duration of remifentanil-induced postoperative hyperalgesia remains unknown, and the effects of alpha-7 nicotinic acetylcholine receptors activation on the brain-derived neurotrophic factor/tyrosine receptor kinase B-K+-Cl- cotransporter-2 signal in the spinal dorsal horn of rats with remifentanil-induced postoperative hyperalgesia is still enigmatic. Results We demonstrated that the brain-derived neurotrophic factor/tyrosine receptor kinase B-K+-Cl- cotransporter-2 signal played a critical role in the development of remifentanil-induced postoperative hyperalgesia. Intrathecal administration of PNU-120596 (8 µg/kg, 15 min before surgery) was associated with earlier signs of recovery from remifentanil-induced postoperative hyperalgesia. Simultaneously, remifentanil-induced postoperative hyperalgesia-induced K+-Cl- cotransporter-2 downregulation was partly reversed and coincided with a decreased expression of brain-derived neurotrophic factor/tyrosine receptor kinase B in the spinal dorsal horn, approximately correlating with the time course of the nociceptive behavior. Moreover, intrathecal administration of the K+-Cl- cotransporter-2 inhibitor VU0240551 significantly reduced the analgesic effect of PNU-120596 on remifentanil-induced postoperative hyperalgesia. Conclusions The activation of alpha-7 nicotinic acetylcholine receptors induced a shorter duration of remifentanil-induced postoperative hyperalgesia by restoring the brain-derived neurotrophic factor/tyrosine receptor kinase B-K+-Cl- cotransporter-2 signal in the spinal dorsal horn of rats, which provides new insight into treatment in clinical postoperative pain management.
Collapse
Affiliation(s)
- Wei Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Yishan Lei
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, China
| | - Yin Cui
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, China
| | - Shuaishuai Chu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, China
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, China
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
36
|
Springborg AD, Jensen EK, Taylor BK, Werner MU. Effects of target-controlled infusion of high-dose naloxone on pain and hyperalgesia in a human thermal injury model: a study protocol: A randomized, double-blind, placebo-controlled, crossover trial with an enriched design. Medicine (Baltimore) 2016; 95:e5336. [PMID: 27861362 PMCID: PMC5120919 DOI: 10.1097/md.0000000000005336] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mu-opioid-receptor antagonists have been extensively studied in experimental research as pharmacological tools uncovering mechanisms of pain modulation by the endogenous opioid system. In rodents, administration of high doses of mu-opioid-receptor antagonists after the resolution of an inflammatory injury has demonstrated reinstatement of nociceptive hypersensitivity indicating unmasking of latent sensitization. In a recent human study, pain hypersensitivity assessed as secondary hyperalgesia area (SHA), was reinstated 7 days after a mild thermal injury, in 4 out of 12 subjects after a naloxone infusion.The aims of the present study are first, to replicate our previous findings in a larger-sized study; second, to examine if high sensitizers (subjects presenting with large SHA after a thermal injury) develop a higher degree of hypersensitivity after naloxone challenge than low sensitizers (subjects presenting with restricted SHA after a thermal injury); and third to examine a dose-response relationship between 3 stable naloxone concentrations controlled by target-controlled infusion, and the unmasking of latent sensitization.Healthy participants (n = 80) underwent a screening day (day 0) with induction of a thermal skin injury (47°C, 420 seconds, 12.5 cm). Assessment of SHA was performed 1 and 2 hours after the injury. Using an enriched design, only participants belonging to the upper quartile of SHA (Q4, high sensitizers; n = 20) and the lower quartile of SHA (Q1, low sensitizers; n = 20) continued the study, comprising 4 consecutive days-days 1 to 4. Thermal skin injuries were repeated on day 1 and day 3, whereas day 2 and day 4 (7 days after day 1 and day 3, respectively) were target-controlled infusion days in which the subjects were randomly allocated to receive either naloxone (3.25 mg/kg, 4 mg/mL) or placebo (normal saline) intravenous. The primary outcome was SHA assessed by weighted-pin instrument (128 mN) 0, 1, 2, and 165 to 169 hours after the thermal injury (day 1-4). The secondary outcomes were pin-prick pain thresholds assessed by weighted-pin instrument (8-512 mN) at primary and secondary hyperalgesia areas (days 1-4).The naloxone-induced unmasking of latent sensitization is an interesting model for exploring the transition from acute to chronic pain. The results from the present study may provide valuable information regarding future research in persistent postsurgical pain states.
Collapse
Affiliation(s)
| | | | - Bradley K. Taylor
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Mads U. Werner
- Neuroscience Center, Copenhagen University Hospitals, Copenhagen, Denmark
| |
Collapse
|
37
|
Sun J, Lin H, Feng X, Dong J, Ansong E, Xu X. A comparison of intrathecal magnesium and ketamine in attenuating remifentanil-induced hyperalgesia in rats. BMC Anesthesiol 2016; 16:74. [PMID: 27599837 PMCID: PMC5013621 DOI: 10.1186/s12871-016-0235-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 08/24/2016] [Indexed: 11/10/2022] Open
Abstract
Background Activation of NMDA receptors play an important role in the development of remifentanil-induced hyperalgesia. We hypothesized that in addition to ketamine, intrathecal MgSO4 could also relieve thermal and mechanical hyperalgesia in rats. Methods Initially, 24 Sprague–Dawley rats were divided into control group, remifentanil group, surgical incision group and remifentanil combined with surgical incision group to create an experimental model. Subsequently, 40 rats were divided into control group, model group, model group plus 100 μg MgSO4, 300 μg MgSO4 and 10 μg ketamine respectively. Paw withdrawal mechanical thresholds and paw withdrawal thermal latency tests were performed at −24 h, 2 h, 6 h, 24 h, 48 h, 72 h and 7 day after the surgical procedure. After behavior assessment on the 7th day, remifentanil was given again to ascertain whether or not NMDA antagonists could suppress the re-exposure of remifentanil-induced hyperalgesia. Results Remifentanil administration plus surgical incision induced significant postoperative hyperalgesia, as indicated by decreased paw withdrawal mechanical thresholds and paw withdrawal thermal latency to mechanical and thermal stimulation. In addition to ketamine, intrathecal MgSO4 (100, 300 μg) dose-dependently reduced remifentanil-induced mechanical and thermal hyperalgesia. Ketamine had less mechanical hyperalgesia in 6 h (p = 0.018), 24 h (p = 0.014) and 48 h (p = 0.011) than 300 μg MgSO4. There was no difference in inhibiting thermal hyperalgesia between the group ketamine and group MgSO4 (300 μg). The rats were given remifentanil again 7 days later after the first exposure of remifentanil. The hyperalgesic effect induced by re-exposure of remifentanil was not reversed in any groups of MgSO4 or ketamine. Conclusions In addition to ketamine, intrathecal administration of MgSO4 dose-dependently reduced remifentanil-induced hyperalgesia in a surgical incision mode. Re-exposure to remifentanil 1 week later again produced hyperalgesia, and this was not altered by the prior intrathecal treatments in any 4 groups treated with MgSO4 or ketamine. Electronic supplementary material The online version of this article (doi:10.1186/s12871-016-0235-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiehao Sun
- Department of Anesthesiology, 1st Affiliated Hospital of Wenzhou Medical University, 1# shangcaicun, Wenzhou, 325000, China
| | - Hai Lin
- Department of Anesthesiology, 1st Affiliated Hospital of Wenzhou Medical University, 1# shangcaicun, Wenzhou, 325000, China.,Department of Anesthesiology and Pain medicine, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaona Feng
- Department of Anesthesiology, 1st Affiliated Hospital of Wenzhou Medical University, 1# shangcaicun, Wenzhou, 325000, China
| | - Jiaojiao Dong
- Department of Anesthesiology, 1st Affiliated Hospital of Wenzhou Medical University, 1# shangcaicun, Wenzhou, 325000, China
| | - Emmanuel Ansong
- Department of Anesthesiology, 1st Affiliated Hospital of Wenzhou Medical University, 1# shangcaicun, Wenzhou, 325000, China
| | - Xuzhong Xu
- Department of Anesthesiology, 1st Affiliated Hospital of Wenzhou Medical University, 1# shangcaicun, Wenzhou, 325000, China.
| |
Collapse
|
38
|
Involvement of CCL3/CCR5 Signaling in Dorsal Root Ganglion in Remifentanil-induced Hyperalgesia in Rats. Clin J Pain 2016; 32:702-10. [DOI: 10.1097/ajp.0000000000000319] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
39
|
Romero-Alejo E, Puig MM, Romero A. Antihyperalgesic effects of dexketoprofen and tramadol in a model of postoperative pain in mice - effects on glial cell activation. ACTA ACUST UNITED AC 2016; 68:1041-50. [PMID: 27291294 DOI: 10.1111/jphp.12584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/14/2016] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To define likely targets (i.e. glia) and protocols (analgesic combinations) to improve postoperative pain outcomes and reduce chronic pain after surgery. Specifically, to assess the antihyperalgesic effects of the dexketoprofen : tramadol (DEX : TRM) combination, exploring the implication of glial activation. METHODS In a mouse model of postincisional pain, we evaluated mechanical nociceptive thresholds (Von Frey) for 21 days postoperatively. We assessed DEX and TRM alone and combined (1 : 1 ratio) on postoperative hyperalgesia (POH, day 1) and delayed latent pain sensitisation (substantiated by a naloxone challenge; PS, day 21). The interactions were analysed using isobolograms, and concomitant changes in spinal glial cell activation were measured. KEY FINDINGS On day 1, DEX completely blocked POH, whereas TRM induced 32% inhibition. TRM, but not DEX, partially (47%) protected against PS, at 21 days. Co-administration of DEX : TRM (1 : 1 ratio) showed additivity for antihyperalgesia. Both drugs and their combination totally inhibited surgery-induced microglia activation on day 1, but had no effect on surgery-induced astrocyte activation (1 day) or re-activation after naloxone (21 days). CONCLUSIONS The DEX : TRM combination could have clinical advantages: a complete prevention of POH after surgery, together with a substantial (48%) inhibition of the development of PS by TRM. Microglia, but not astrocyte activation, could play a relevant role in the development of postoperative pain hypersensitivity.
Collapse
Affiliation(s)
- Elizabeth Romero-Alejo
- Pain Research Unit, Department of Anesthesiology, IMIM-Institut Hospital del Mar d'Investigacions Biomèdiques, Universitat Autònoma de Barcelona School of Medicine, Barcelona, Spain
| | - Margarita M Puig
- Pain Research Unit, Department of Anesthesiology, IMIM-Institut Hospital del Mar d'Investigacions Biomèdiques, Universitat Autònoma de Barcelona School of Medicine, Barcelona, Spain
| | - Asunción Romero
- Pain Research Unit, Department of Anesthesiology, IMIM-Institut Hospital del Mar d'Investigacions Biomèdiques, Universitat Autònoma de Barcelona School of Medicine, Barcelona, Spain
| |
Collapse
|
40
|
Sustained Suppression of Hyperalgesia during Latent Sensitization by μ-, δ-, and κ-opioid receptors and α2A Adrenergic Receptors: Role of Constitutive Activity. J Neurosci 2016; 36:204-21. [PMID: 26740662 DOI: 10.1523/jneurosci.1751-15.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Many chronic pain disorders alternate between bouts of pain and periods of remission. The latent sensitization model reproduces this in rodents by showing that the apparent recovery ("remission") from inflammatory or neuropathic pain can be reversed by opioid antagonists. Therefore, this remission represents an opioid receptor-mediated suppression of a sustained hyperalgesic state. To identify the receptors involved, we induced latent sensitization in mice and rats by injecting complete Freund's adjuvant (CFA) in the hindpaw. In WT mice, responses to mechanical stimulation returned to baseline 3 weeks after CFA. In μ-opioid receptor (MOR) knock-out (KO) mice, responses did not return to baseline but partially recovered from peak hyperalgesia. Antagonists of α2A-adrenergic and δ-opioid receptors reinstated hyperalgesia in WT mice and abolished the partial recovery from hyperalgesia in MOR KO mice. In rats, antagonists of α2A adrenergic and μ-, δ-, and κ-opioid receptors reinstated hyperalgesia during remission from CFA-induced hyperalgesia. Therefore, these four receptors suppress hyperalgesia in latent sensitization. We further demonstrated that suppression of hyperalgesia by MORs was due to their constitutive activity because of the following: (1) CFA-induced hyperalgesia was reinstated by the MOR inverse agonist naltrexone (NTX), but not by its neutral antagonist 6β-naltrexol; (2) pro-enkephalin, pro-opiomelanocortin, and pro-dynorphin KO mice showed recovery from hyperalgesia and reinstatement by NTX; (3) there was no MOR internalization during remission; (4) MORs immunoprecipitated from the spinal cord during remission had increased Ser(375) phosphorylation; and (5) electrophysiology recordings from dorsal root ganglion neurons collected during remission showed constitutive MOR inhibition of calcium channels. SIGNIFICANCE STATEMENT Chronic pain causes extreme suffering to millions of people, but its mechanisms remain to be unraveled. Latent sensitization is a phenomenon studied in rodents that has many key features of chronic pain: it is initiated by a variety of noxious stimuli, has indefinite duration, and pain appears in episodes that can be triggered by stress. Here, we show that, during latent sensitization, there is a sustained state of pain hypersensitivity that is continuously suppressed by the activation of μ-, δ-, and κ-opioid receptors and by adrenergic α2A receptors in the spinal cord. Furthermore, we show that the activation of μ-opioid receptors is not due to the release of endogenous opioids, but rather to its ligand-independent constitutive activity.
Collapse
|
41
|
Sahbaie P, Liang DY, Shi XY, Sun Y, Clark JD. Epigenetic regulation of spinal cord gene expression contributes to enhanced postoperative pain and analgesic tolerance subsequent to continuous opioid exposure. Mol Pain 2016; 12:12/0/1744806916641950. [PMID: 27094549 PMCID: PMC4956243 DOI: 10.1177/1744806916641950] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 02/12/2016] [Indexed: 11/25/2022] Open
Abstract
Background Opioids have become the mainstay for treatment of moderate to severe pain and are commonly used to treat surgical pain. While opioid administration has been shown to cause opioid-induced hyperalgesia and tolerance, interactions between opioid administration and surgery with respect to these problematic adaptations have scarcely been addressed. Accumulating evidence suggests opioids and nociceptive signaling may converge on epigenetic mechanisms in spinal cord to enhance or prolong neuroplastic changes. Epigenetic regulation of Bdnf (brain-derived neurotrophic factor) and Pdyn (prodynorphin) genes may be involved. Results Four days of ascending doses of morphine treatment caused opioid-induced hyperalgesia and reduced opioid analgesic efficacy in mice. Both opioid-induced hyperalgesia and the reduced opioid analgesic efficacy were enhanced in mice that received hindpaw incisions. The expression of Bdnf and Pdyn (qPCR) was increased after morphine treatment and incision. Chromatin immunoprecipitation assays demonstrated that the Pdyn and Bdnf promoters were more strongly associated with acetylated H3K9 after morphine plus incision than in the morphine or incision alone groups. Selective tropomyosin-related kinase B (ANA-12) and κ-opioid receptor (nor-binaltorphimine) antagonists were administered intrathecally, both reduced hyperalgesia one or three days after surgery. Administration of ANA-12 or nor-binaltorphimine attenuated the decreased morphine analgesic efficacy on day 1, but only nor-binaltorphimine was effective on day 3 after incision in opioid-exposed group. Coadministration of histone acetyltransferase inhibitor anacardic acid daily with morphine blocked the development of opioid-induced hyperalgesia and attenuated incision-enhanced hyperalgesia in morphine-treated mice. Anacardic acid had similar effects on analgesic tolerance, showing the involvement of histone acetylation in the interactions detected. Conclusions Spinal epigenetic changes involving Bdnf and Pdyn may contribute to the enhanced postoperative nociceptive sensitization and analgesic tolerance observed after continuous opioid exposure. Treatments blocking the epigenetically mediated up-regulation of these genes or administration of TrkB or κ-opioid receptor antagonists may improve the clinical utility of opioids, particularly after surgery.
Collapse
Affiliation(s)
- Peyman Sahbaie
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA Anaesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - De-Yong Liang
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA Anaesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Xiao-You Shi
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA Anaesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Yuan Sun
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA Anaesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - J David Clark
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA Anaesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
42
|
Pereira MP, Werner MU, Dahl JB, Pereira MP, Utke Werner M, Berg Dahl J. Effect of a high-dose target-controlled naloxone infusion on pain and hyperalgesia in patients following groin hernia repair: study protocol for a randomized controlled trial. Trials 2015; 16:511. [PMID: 26554360 PMCID: PMC4640219 DOI: 10.1186/s13063-015-1021-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 10/20/2015] [Indexed: 12/04/2022] Open
Abstract
Background Central sensitization is modulated by the endogenous opioid system and plays a major role in the development and maintenance of pain. Recent animal studies performed following resolution of inflammatory pain showed reinstatement of tactile hypersensitivity induced by administration of a mu-opioid-antagonist, suggesting latent sensitization is mediated by endogenous opioids. In a recent crossover study in healthy volunteers, following resolution of a first-degree burn, 4 out of 12 volunteers developed large secondary areas of hyperalgesia areas after a naloxone infusion, while no volunteer developed significant secondary hyperalgesia after the placebo infusion. In order to consistently demonstrate latent sensitization in humans, a pain model inducing deep tissue inflammation, as used in animal studies, might be necessary. The aim of the present study is to examine whether a high-dose target-controlled naloxone infusion can reinstate pain and hyperalgesia following recovery from open groin hernia repair and thus consistently demonstrate opioid-mediated latent sensitization in humans. Methods/Design Patients submitted to unilateral, primary, open groin hernia repair will be included in this randomized, placebo-controlled, double-blind, crossover study. The experimental days take place 6–8 weeks after surgery, time-points at which patients are expected to be almost pain- free. Prior to administration of naloxone or placebo, the primary outcome (a summated measure of pain: at rest, during transition from supine to standing position, and evoked by pressure algometry) and the secondary outcomes (secondary hyperalgesia/allodynia, pressure pain thresholds, assessed at the surgical site and at the mirror-site in the contralateral groin, and, opioid withdrawal symptoms) will be assessed. These assessments will be repeated at each step of the target-controlled infusion of placebo or naloxone at estimated median (95 % CI) plasma concentrations of 344 ng/ml (130;567), 1059 ng/ml (400;1752) and 3196 ng/ml (1205;5276). Discussion We aim to demonstrate opioid-mediated latent sensitization in a post-surgical setting, using pain as a clinical relevant variable. Impairment of the protective endogenous opioid system may play an important role in the transition from acute to chronic pain. In order to sufficiently block the endogenous opioid system, a high-dose target-controlled naloxone-infusion is used, in accordance with recent findings in animal studies. Trial registration number EUDRACT: 2015-000793-36 (Registration date: 16 February 2015) Clinicaltrials.gov: NCT01992146 (Registration date: 12 December 2014)
Collapse
Affiliation(s)
- M P Pereira
- Department of Anaesthesiology, Centre of Head and Orthopaedics, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark.
| | - M U Werner
- Multidisciplinary Pain Center, Neuroscience Center, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark.
| | - J B Dahl
- Department of Anaesthesiology, Centre of Head and Orthopaedics, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark.
| | - Manuel Pedro Pereira
- Department of Anaesthesiology, Centre of Head and Orthopaedics, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark. .,Multidisciplinary Pain Center, Neuroscience Center, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark.
| | - Mads Utke Werner
- Multidisciplinary Pain Center, Neuroscience Center, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark.
| | - Joergen Berg Dahl
- Department of Anaesthesiology, Centre of Head and Orthopaedics, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark.
| |
Collapse
|
43
|
Becerra L, Bishop J, Barmettler G, Xie Y, Navratilova E, Porreca F, Borsook D. Triptans disrupt brain networks and promote stress-induced CSD-like responses in cortical and subcortical areas. J Neurophysiol 2015; 115:208-17. [PMID: 26490291 DOI: 10.1152/jn.00632.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/18/2015] [Indexed: 12/24/2022] Open
Abstract
A number of drugs, including triptans, promote migraine chronification in susceptible individuals. In rats, a period of triptan administration over 7 days can produce "latent sensitization" (14 days after discontinuation of drug) demonstrated as enhanced sensitivity to presumed migraine triggers such as environmental stress and lowered threshold for electrically induced cortical spreading depression (CSD). Here we have used fMRI to evaluate the early changes in brain networks at day 7 of sumatriptan administration that may induce latent sensitization as well as the potential response to stress. After continuous infusion of sumatriptan, rats were scanned to measure changes in resting state networks and the response to bright light environmental stress. Rats receiving sumatriptan, but not saline infusion, showed significant differences in default mode, autonomic, basal ganglia, salience, and sensorimotor networks. Bright light stress produced CSD-like responses in sumatriptan-treated but not control rats. Our data show the first brain-related changes in a rat model of medication overuse headache and suggest that this approach could be used to evaluate the multiple brain networks involved that may promote this condition.
Collapse
Affiliation(s)
- L Becerra
- P.A.I.N. Group, Boston Children's Hospital, Waltham, Massachusetts; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and
| | - J Bishop
- P.A.I.N. Group, Boston Children's Hospital, Waltham, Massachusetts
| | - G Barmettler
- P.A.I.N. Group, Boston Children's Hospital, Waltham, Massachusetts
| | - Y Xie
- Department of Pharmacology, University of Arizona, Tucson, Arizona
| | - E Navratilova
- Department of Pharmacology, University of Arizona, Tucson, Arizona
| | - F Porreca
- Department of Pharmacology, University of Arizona, Tucson, Arizona
| | - D Borsook
- P.A.I.N. Group, Boston Children's Hospital, Waltham, Massachusetts; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and
| |
Collapse
|
44
|
Pereira MP, Donahue RR, Dahl JB, Werner M, Taylor BK, Werner MU. Endogenous Opioid-Masked Latent Pain Sensitization: Studies from Mouse to Human. PLoS One 2015; 10:e0134441. [PMID: 26305798 PMCID: PMC4549112 DOI: 10.1371/journal.pone.0134441] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 06/08/2015] [Indexed: 01/20/2023] Open
Abstract
Following the resolution of a severe inflammatory injury in rodents, administration of mu-opioid receptor inverse agonists leads to reinstatement of pain hypersensitivity. The mechanisms underlying this form of latent pain sensitization (LS) likely contribute to the development of chronic pain, but LS has not yet been demonstrated in humans. Using a C57BL/6 mouse model of cutaneous mild heat injury (MHI) we demonstrated a dose-dependent reinstatement of pain sensitization, assessed as primary (P < 0.001) and secondary hyperalgesia (P < 0.001) by naloxone (0.3–10 mg/kg), 168 hrs after the induction of MHI. Forward-translating the dose data to a human MHI model (n = 12) we could show that LS does indeed occur after naloxone 2 mg/kg, 168 hrs after a MHI. Our previous unsuccessful efforts to demonstrate unmasking of LS in humans are thus likely explained by an insufficient naloxone dose (0.021 mg/kg). However, while LS was consistently demonstrated in 21/24 mice, LS was only seen in 4/12 subjects. This difference is likely due to selection bias since the C57BL/6 mouse strain exhibits markedly enhanced pain sensitivity in assays of acute thermal nociception. Future exploratory studies in humans should prioritize inclusion of “high-sensitizers” prone to develop LS and use post-surgical models to elucidate markers of vulnerability to chronic postsurgical pain.
Collapse
Affiliation(s)
- Manuel P. Pereira
- Department of Anaesthesia, Centre of Head and Orthopaedics, Rigshospitalet, Copenhagen, Denmark
| | - Renee R. Donahue
- Department of Physiology, University of Kentucky, Lexington, KY, United States of America
| | - Jørgen B. Dahl
- Department of Anaesthesia, Centre of Head and Orthopaedics, Rigshospitalet, Copenhagen, Denmark
| | - Marianne Werner
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark
| | - Bradley K. Taylor
- Multidisciplinary Pain Center, Neuroscience Center, Rigshospitalet, Copenhagen, Denmark
| | - Mads U. Werner
- Multidisciplinary Pain Center, Neuroscience Center, Rigshospitalet, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
45
|
Kitanaka J, Kitanaka N, Hall FS, Fujii M, Goto A, Kanda Y, Koizumi A, Kuroiwa H, Mibayashi S, Muranishi Y, Otaki S, Sumikawa M, Tanaka KI, Nishiyama N, Uhl GR, Takemura M. Memory impairment and reduced exploratory behavior in mice after administration of systemic morphine. J Exp Neurosci 2015; 9:27-35. [PMID: 25987850 PMCID: PMC4428380 DOI: 10.4137/jen.s25057] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 03/16/2015] [Accepted: 03/19/2015] [Indexed: 01/28/2023] Open
Abstract
In the present study, the effects of morphine were examined on tests of spatial memory, object exploration, locomotion, and anxiety in male ICR mice. Administration of morphine (15 or 30 mg/kg, intraperitoneally (i.p.)) induced a significant decrease in Y-maze alternations compared to saline vehicle-treated mice. The reduced Y-maze alternations induced by morphine were completely blocked by naloxone (15 mg/kg) or β-funaltrexamine (5 mg/kg) but not by norbinaltorphimine (5 mg/kg) or naltrindole (5 mg/kg), suggesting that the morphine-induced spatial memory impairment was mediated predominantly by μ-opioid receptors (MOPs). Significant spatial memory retrieval impairments were observed in the Morris water maze (MWM) in mice treated with morphine (15 mg/kg) or scopolamine (1 mg/kg), but not with naloxone or morphine plus naloxone. Reduced exploratory time was observed in mice after administration of morphine (15 mg/kg), in a novel-object exploration test, without any changes in locomotor activity. No anxiolytic-like behavior was observed in morphine-treated mice in the elevated plus maze. A significant reduction in buried marbles was observed in morphine-treated mice measured in the marble-burying test, which was blocked by naloxone. These observations suggest that morphine induces impairments in spatial short-term memory and retrieval, and reduces exploratory behavior, but that these effects are not because of overall changes in locomotion or anxiety.
Collapse
Affiliation(s)
- Junichi Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Nobue Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - F Scott Hall
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Mei Fujii
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Akiko Goto
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Yusuke Kanda
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Akira Koizumi
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | | | - Satoko Mibayashi
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Yumi Muranishi
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Soichiro Otaki
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Minako Sumikawa
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Koh-Ichi Tanaka
- Division of Pharmacology, Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Hyogo, Japan
| | - Nobuyoshi Nishiyama
- Division of Pharmacology, Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Hyogo, Japan. ; The Office of the Dean, School of Pharmacy, Hyogo University of Health Sciences, Hyogo, Japan
| | - George R Uhl
- Molecular Neurobiology Branch, National Institute on Drug Abuse-Intramural Research Program, Baltimore, MD, USA
| | | |
Collapse
|
46
|
Marvizon JC, Walwyn W, Minasyan A, Chen W, Taylor BK. Latent sensitization: a model for stress-sensitive chronic pain. ACTA ACUST UNITED AC 2015; 71:9.50.1-9.50.14. [PMID: 25829356 DOI: 10.1002/0471142301.ns0950s71] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Latent sensitization is a rodent model of chronic pain that reproduces both its episodic nature and its sensitivity to stress. It is triggered by a wide variety of injuries ranging from injection of inflammatory agents to nerve damage. It follows a characteristic time course in which a hyperalgesic phase is followed by a phase of remission. The hyperalgesic phase lasts between a few days to several months, depending on the triggering injury. Injection of μ-opioid receptor inverse agonists (e.g., naloxone or naltrexone) during the remission phase induces reinstatement of hyperalgesia. This indicates that the remission phase does not represent a return to the normal state, but rather an altered state in which hyperalgesia is masked by constitutive activity of opioid receptors. Importantly, stress also triggers reinstatement. Here we describe in detail procedures for inducing and following latent sensitization in its different phases in rats and mice.
Collapse
Affiliation(s)
- Juan Carlos Marvizon
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Wendy Walwyn
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ani Minasyan
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Wenling Chen
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Bradley K Taylor
- Department of Physiology, School of Medicine, University of Kentucky Medical Center, Lexington, Kentucky
| |
Collapse
|
47
|
Taylor BK, Corder G. Endogenous analgesia, dependence, and latent pain sensitization. Curr Top Behav Neurosci 2014; 20:283-325. [PMID: 25227929 PMCID: PMC4464817 DOI: 10.1007/7854_2014_351] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Endogenous activation of µ-opioid receptors (MORs) provides relief from acute pain. Recent studies have established that tissue inflammation produces latent pain sensitization (LS) that is masked by spinal MOR signaling for months, even after complete recovery from injury and re-establishment of normal pain thresholds. Disruption with MOR inverse agonists reinstates pain and precipitates cellular, somatic, and aversive signs of physical withdrawal; this phenomenon requires N-methyl-D-aspartate receptor-mediated activation of calcium-sensitive adenylyl cyclase type 1 (AC1). In this review, we present a new conceptual model of the transition from acute to chronic pain, based on the delicate balance between LS and endogenous analgesia that develops after painful tissue injury. First, injury activates pain pathways. Second, the spinal cord establishes MOR constitutive activity (MORCA) as it attempts to control pain. Third, over time, the body becomes dependent on MORCA, which paradoxically sensitizes pain pathways. Stress or injury escalates opposing inhibitory and excitatory influences on nociceptive processing as a pathological consequence of increased endogenous opioid tone. Pain begets MORCA begets pain vulnerability in a vicious cycle. The final result is a silent insidious state characterized by the escalation of two opposing excitatory and inhibitory influences on pain transmission: LS mediated by AC1 (which maintains the accelerator) and pain inhibition mediated by MORCA (which maintains the brake). This raises the prospect that opposing homeostatic interactions between MORCA analgesia and latent NMDAR-AC1-mediated pain sensitization creates a lasting vulnerability to develop chronic pain. Thus, chronic pain syndromes may result from a failure in constitutive signaling of spinal MORs and a loss of endogenous analgesic control. An overarching long-term therapeutic goal of future research is to alleviate chronic pain by either (a) facilitating endogenous opioid analgesia, thus restricting LS within a state of remission, or (b) extinguishing LS altogether.
Collapse
Affiliation(s)
- Bradley K Taylor
- Department of Physiology, School of Medicine, University of Kentucky Medical Center, Lexington, KY, 40536-0298, USA,
| | | |
Collapse
|
48
|
Werner MU. Management of persistent postsurgical inguinal pain. Langenbecks Arch Surg 2014; 399:559-69. [DOI: 10.1007/s00423-014-1211-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/05/2014] [Indexed: 01/27/2023]
|
49
|
Werner MU, Bischoff JM. Persistent postsurgical pain: evidence from breast cancer surgery, groin hernia repair, and lung cancer surgery. Curr Top Behav Neurosci 2014; 20:3-29. [PMID: 24523139 DOI: 10.1007/7854_2014_285] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The prevalences of severe persistent postsurgical pain (PPP) following breast cancer surgery (BCS), groin hernia repair (GHR), and lung cancer surgery (LCS) are 13, 2, and 4-12 %, respectively. Estimates indicate that 80,000 patients each year in the U.S.A. are affected by severe pain and debilitating impairment in the aftermath of BCS, GHR, and LCS. Data across the three surgical procedures indicate a 35-65 % decrease in prevalence of PPP at 4-6 years follow-up. However, this is outweighed by late-onset PPP, which appears following a pain-free interval. The consequences of PPP include severe impairments of physical, psychological, and socioeconomic aspects of life. The pathophysiology underlying PPP consists of a continuing inflammatory response, a neuropathic component, and/or a late reinstatement of postsurgical inflammatory pain. While the sensory profiles of PPP-patients and pain-free controls are comparable with hypofunction on the surgical side, this seems to be accentuated in PPP-patients. In BCS-patients and GHR-patients, the sensory profiles indicate inflammatory and neuropathic components with contribution of central sensitization. A number of surgical factors including increased duration of surgery, repeat surgery, more invasive surgical techniques, and intraoperative nerve lesion have been associated with PPP. One of the most consistent predictive factors for PPP is high intensity acute postsurgical pain, but also psychological factors including anxiety, catastrophizing trait, depression, and psychological vulnerability have been identified as significant predictors of PPP. The quest to identify improved surgical and anesthesiological techniques to prevent severe pain and functional impairment in patients after surgery continues.
Collapse
Affiliation(s)
- Mads Utke Werner
- Multidisciplinary Pain Center 7612, Neuroscience Center, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark,
| | | |
Collapse
|
50
|
The selective μ opioid receptor antagonist β-funaltrexamine attenuates methamphetamine-induced stereotypical biting in mice. Brain Res 2013; 1522:88-98. [DOI: 10.1016/j.brainres.2013.05.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 04/23/2013] [Accepted: 05/19/2013] [Indexed: 11/22/2022]
|