1
|
Fujita W. [MOPr-DOPr heteromer: the meaning and possibility as novel therapeutic target for pain control]. Nihon Yakurigaku Zasshi 2021; 156:134-138. [PMID: 33952839 DOI: 10.1254/fpj.20103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Many studies suggest opioid receptor (OPr) dimerization modulates the pharmacological properties of opiates. Specifically, heteromerization between OPr types has been reported to lead to changes in intracellular signaling. Thus, ligands targeting heteromers are expected to be novel therapeutic targets with reduced side effects. The heteromers of mu (MOPr) and delta (DOPr) are detected in brain regions involved in pain processing. The bivalent ligand or small molecule were identified as a MOPr-DOPr targeting ligand. These ligands exhibit antinociceptive properties similar to that of morphine with lesser antinociceptive tolerance as compared to morphine. Studies exploring the in vivo regulation of MOPr-DOPr heteromers, showed chronic morphine administration leads to an upregulation of these heteromers in select brain regions. Exploration of mechanisms underlying this phenomenon led us to the G protein-coupled receptor chaperone, RTP4, that is induced by chronic morphine and facilitates the heteromerization of MOPr and DOPr. In this review, I will introduce the simulated structure or property of MOPr-DOPr heteromer, its targeting ligands, and its intracellular regulatory mechanism that include a key molecule like RTP4 that could serve as a scaffold for the development of novel therapeutic drugs with reduced adverse effects, and hence may take place of the conventional clinical opioids.
Collapse
Affiliation(s)
- Wakako Fujita
- Department of Medical Pharmacology, Nagasaki University Graduate School of Biomedical Sciences
| |
Collapse
|
2
|
Dual Enkephalinase Inhibitors and Their Role in Chronic Pain Management. Curr Pain Headache Rep 2021; 25:29. [PMID: 33761014 DOI: 10.1007/s11916-021-00949-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Dual enkephalinase inhibitors (DENKIs) are pain medications that indirectly activate opioid receptors and can be used as an alternative to traditional opioids. Understanding the physiology of enkephalins and their inhibitors and the pharmacology of these drugs will allow for proper clinical application for chronic pain patients in the future. RECENT FINDINGS DENKIs can be used as an alternative mode of analgesia for patients suffering from chronic pain by preventing the degradation of endogenous opioid ligands. By inhibiting the two major enkephalin-degrading enzymes (neprilysin and aminopeptidase N), DENKIs can provide analgesia with less adverse effects than nonendogenous opioids. The purpose of this paper is to review the current literature investigating DENKIs and explore their contribution to chronic pain management.
Collapse
|
3
|
Chang C, Liu HK, Yeh CB, Yang ML, Liao WC, Liu CH, Tseng TJ. Cross-Talk of Toll-Like Receptor 5 and Mu-Opioid Receptor Attenuates Chronic Constriction Injury-Induced Mechanical Hyperalgesia through a Protein Kinase C Alpha-Dependent Signaling. Int J Mol Sci 2021; 22:1891. [PMID: 33673008 PMCID: PMC7918001 DOI: 10.3390/ijms22041891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/30/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Recently, Toll-like receptors (TLRs), a family of pattern recognition receptors, are reported as potential modulators for neuropathic pain; however, the desired mechanism is still unexplained. Here, we operated on the sciatic nerve to establish a pre-clinical rodent model of chronic constriction injury (CCI) in Sprague-Dawley rats, which were assigned into CCI and Decompression groups randomly. In Decompression group, the rats were performed with nerve decompression at post-operative week 4. Mechanical hyperalgesia and mechanical allodynia were obviously attenuated after a month. Toll-like receptor 5 (TLR5)-immunoreactive (ir) expression increased in dorsal horn, particularly in the inner part of lamina II. Additionally, substance P (SP) and isolectin B4 (IB4)-ir expressions, rather than calcitonin-gene-related peptide (CGRP)-ir expression, increased in their distinct laminae. Double immunofluorescence proved that increased TLR5-ir expression was co-expressed mainly with IB4-ir expression. Through an intrathecal administration with FLA-ST Ultrapure (a TLR5 agonist, purified flagellin from Salmonella Typhimurium, only the CCI-induced mechanical hyperalgesia was attenuated dose-dependently. Moreover, we confirmed that mu-opioid receptor (MOR) and phospho-protein kinase Cα (pPKCα)-ir expressions but not phospho-protein kinase A RII (pPKA RII)-ir expression, increased in lamina II, where they mostly co-expressed with IB4-ir expression. Go 6976, a potent protein kinase C inhibitor, effectively reversed the FLA-ST Ultrapure- or DAMGO-mediated attenuated trend towards mechanical hyperalgesia by an intrathecal administration in CCI rats. In summary, our current findings suggest that nerve decompression improves CCI-induced mechanical hyperalgesia that might be through the cross-talk of TLR5 and MOR in a PKCα-dependent manner, which opens a novel opportunity for the development of analgesic therapeutics in neuropathic pain.
Collapse
Affiliation(s)
- Ching Chang
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
| | - Hung-Kai Liu
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
| | - Chao-Bin Yeh
- Department of Emergency Medicine, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan;
- Department of Emergency Medicine, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan
| | - Ming-Lin Yang
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan
| | - Wen-Chieh Liao
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan
| | - Chiung-Hui Liu
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan
| | - To-Jung Tseng
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan
| |
Collapse
|
4
|
Fujita W. The Possible Role of MOPr-DOPr Heteromers and Its Regulatory Protein RTP4 at Sensory Neurons in Relation to Pain Perception. Front Cell Neurosci 2020; 14:609362. [PMID: 33304244 PMCID: PMC7693438 DOI: 10.3389/fncel.2020.609362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Heteromers between mu opioid receptor (MOPr) and delta opioid receptor (DOPr) (i.e., MOPr-DOPr heteromer) have been found to be expressed in different brain regions, in the spinal cord, and in dorsal root ganglia. Recent studies on this heteromer reveal its important pathophysiological function in pain regulation including neuropathic pain; this suggests a role as a novel therapeutic target in chronic pain management. In addition, receptor transporter protein 4 (RTP4) has been shown to be involved in the intracellular maturation of the MOPr-DOPr heteromers. RTP4 appears to have unique distribution in vivo being highly expressed in sensory neurons and also macrophages; the latter are effector cells of the innate immune system that phagocytose foreign substances and secrete both pro-inflammatory and antimicrobial mediators; this suggests a possible contribution of RTP4 to neuronal immune-related pathological conditions such as neuropathic pain. Although RTP4 could be considered as an important therapeutic target in the management of pain via MOPr-DOPr heteromer, a few reports have supported this. This review will summarize the possible role or functions of the MOPr-DOPr heteromer and its regulatory molecule RTP4 in pain modulation at sensory neurons.
Collapse
Affiliation(s)
- Wakako Fujita
- Department of Medical Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
5
|
Uhelski ML, Bruce D, Speltz R, Wilcox GL, Simone DA. Topical Application of Loperamide/Oxymorphindole, Mu and Delta Opioid Receptor Agonists, Reduces Sensitization of C-fiber Nociceptors that Possess Na V1.8. Neuroscience 2020; 446:102-112. [PMID: 32858141 DOI: 10.1016/j.neuroscience.2020.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 06/27/2020] [Accepted: 08/17/2020] [Indexed: 01/09/2023]
Abstract
It was recently shown that local injection, systemic administration or topical application of the peripherally-restricted mu-opioid receptor (MOR) agonist loperamide (Lo) and the delta-opioid receptor (DOR) agonist oxymorphindole (OMI) synergized to produce highly potent anti-hyperalgesia that was dependent on both MOR and DOR located in the periphery. We assessed peripheral mechanisms by which this Lo/OMI combination produces analgesia in mice expressing the light-sensitive protein channelrhodopsin2 (ChR2) in neurons that express NaV1.8 voltage-gated sodium channels. These mice (NaV1.8-ChR2+) enabled us to selectively target and record electrophysiological activity from these neurons (the majority of which are nociceptive) using blue light stimulation of the hind paw. We assessed the effect of Lo/OMI on nociceptor activity in both naïve mice and mice treated with complete Freund's adjuvant (CFA) to induce chronic inflammation of the hind paw. Teased fiber recording of tibial nerve fibers innervating the plantar hind paw revealed that the Lo/OMI combination reduced responses to light stimulation in naïve mice and attenuated spontaneous activity (SA) as well as responses to light and mechanical stimuli in CFA-treated mice. These results show that Lo/OMI reduces activity of C-fiber nociceptors that express NaV1.8 and corroborate recent behavioral studies demonstrating the potent analgesic effects of this drug combination. Because of its peripheral site of action, Lo/OMI might produce effective analgesia without the side effects associated with activation of opioid receptors in the central nervous system.
Collapse
Affiliation(s)
- Megan L Uhelski
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel Bruce
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rebecca Speltz
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - George L Wilcox
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Dermatology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Donald A Simone
- Department of Diagnostic & Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
6
|
Tiwari V, He SQ, Huang Q, Liang L, Yang F, Chen Z, Tiwari V, Fujita W, Devi LA, Dong X, Guan Y, Raja SN. Activation of µ-δ opioid receptor heteromers inhibits neuropathic pain behavior in rodents. Pain 2020; 161:842-855. [PMID: 31815916 PMCID: PMC7085422 DOI: 10.1097/j.pain.0000000000001768] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Several reports support the idea that µ- and δ-opioid receptors (ORs) may exist as heterodimers in brain regions involved in pain signaling. The unique pharmacology of these heteromers may present a novel analgesic target. However, the role of µ-δ heteromers in sensory neurons involved in pain and opioid analgesia remains unclear, particularly during neuropathic pain. We examined the effects of spinal nerve injury on µ-δ heteromer expression in dorsal root ganglion (DRG) neurons and the effects of a µ-δ heteromer-targeting agonist, CYM51010, on neuropathic pain behavior in rats and mice. An L5 spinal nerve ligation (SNL) in rats significantly decreased µ-δ heteromer expression in L5 DRG but increased heteromer levels in uninjured L4 DRG. Importantly, in SNL rats, subcutaneous injection of CYM51010 inhibited mechanical hypersensitivity in a dose-related manner (EC50: 1.09 mg/kg) and also reversed heat hyperalgesia and attenuated ongoing pain (2 mg/kg, subcutaneously). HEK-293T cell surface-labeled with µ- and δ-ORs internalized both receptors after exposure to CYM51010. By contrast, in cells transfected with µ-OR alone, CYM51010 was significantly less effective at inducing receptor internalization. Electrophysiologic studies showed that CYM51010 inhibited the C-component and windup phenomenon in spinal wide dynamic range neurons of SNL rats. The pain inhibitory effects of CYM51010 persisted in morphine-tolerant rats but was markedly attenuated in µ-OR knockout mice. Our studies show that spinal nerve injury may increase µ-δ heterodimerization in uninjured DRG neurons, and that µ-δ heteromers may be a potential therapeutic target for relieving neuropathic pain, even under conditions of morphine tolerance.
Collapse
Affiliation(s)
- Vinod Tiwari
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Shao-Qiu He
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Qian Huang
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Lingli Liang
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Fei Yang
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Zhiyong Chen
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Vineeta Tiwari
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Wakako Fujita
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lakshmi A. Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
- Howard Hughes Medical Institute, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
- Department of Neurological Surgery, the Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Srinivasa N. Raja
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| |
Collapse
|
7
|
In Vitro Nociceptor Neuroplasticity Associated with In Vivo Opioid-Induced Hyperalgesia. J Neurosci 2019; 39:7061-7073. [PMID: 31300521 DOI: 10.1523/jneurosci.1191-19.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 11/21/2022] Open
Abstract
Opioid-induced hyperalgesia (OIH) is a serious adverse event produced by opioid analgesics. Lack of an in vitro model has hindered study of its underlying mechanisms. Recent evidence has implicated a role of nociceptors in OIH. To investigate the cellular and molecular mechanisms of OIH in nociceptors, in vitro, subcutaneous administration of an analgesic dose of fentanyl (30 μg/kg, s.c.) was performed in vivo in male rats. Two days later, when fentanyl was administered intradermally (1 μg, i.d.), in the vicinity of peripheral nociceptor terminals, it produced mechanical hyperalgesia (OIH). Additionally, 2 d after systemic fentanyl, rats had also developed hyperalgesic priming (opioid-primed rats), long-lasting nociceptor neuroplasticity manifested as prolongation of prostaglandin E2 (PGE2) hyperalgesia. OIH was reversed, in vivo, by intrathecal administration of cordycepin, a protein translation inhibitor that reverses priming. When fentanyl (0.5 nm) was applied to dorsal root ganglion (DRG) neurons, cultured from opioid-primed rats, it induced a μ-opioid receptor (MOR)-dependent increase in [Ca2+]i in 26% of small-diameter neurons and significantly sensitized (decreased action potential rheobase) weakly IB4+ and IB4- neurons. This sensitizing effect of fentanyl was reversed in weakly IB4+ DRG neurons cultured from opioid-primed rats after in vivo treatment with cordycepin, to reverse of OIH. Thus, in vivo administration of fentanyl induces nociceptor neuroplasticity, which persists in culture, providing evidence for the role of nociceptor MOR-mediated calcium signaling and peripheral protein translation, in the weakly IB4-binding population of nociceptors, in OIH.SIGNIFICANCE STATEMENT Clinically used μ-opioid receptor agonists such as fentanyl can produce hyperalgesia and hyperalgesic priming. We report on an in vitro model of nociceptor neuroplasticity mediating this opioid-induced hyperalgesia (OIH) and priming induced by fentanyl. Using this model, we have found qualitative and quantitative differences between cultured nociceptors from opioid-naive and opioid-primed animals, and provide evidence for the important role of nociceptor μ-opioid receptor-mediated calcium signaling and peripheral protein translation in the weakly IB4-binding population of nociceptors in OIH. These findings provide information useful for the design of therapeutic strategies to alleviate OIH, a serious adverse event of opioid analgesics.
Collapse
|
8
|
Synergistic efficacy of tramadol and meloxicam on alleviation of pain and selected immunological variables after sciatic nerve ligation in rats. Int J Vet Sci Med 2019. [DOI: 10.1016/j.ijvsm.2013.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
9
|
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
|
10
|
Liu B, Liu Y, Li N, Zhang J, Zhang X. Oxycodone regulates incision-induced activation of neurotrophic factors and receptors in an acute post-surgery pain rat model. J Pain Res 2018; 11:2663-2674. [PMID: 30464584 PMCID: PMC6214342 DOI: 10.2147/jpr.s180396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background Oxycodone, which is one of the most commonly used opiates in postoperative pain management, has a different affinity for μ-opioid receptors (MOR), κ-opioid receptors (KOR), and δ-opioid receptors (DOR). Accumulating research has suggested that neurotrophins (NTs) are involved in opioid analgesia. In the current exploratory study, we aimed to investigate the underlying mechanisms of the analgesic effects of oxycodone on post-surgery pain in rats and to determine whether neurotrophic factors and receptors were involved in these effects. Methods Mechanical and thermal sensitivity tests were used to evaluate the validity of the postoperative pain rat model and to determine the analgesic effect of oxycodone. Quantitative PCR and Western blot analysis were used to detect the changes in the expression of three types of opioid receptors and NTs and their high-affinity receptors in the spinal cord after surgery and oxycodone administration. Results Oxycodone showed an analgesic effect on plantar incision (PI)-induced hyperalgesia, especially thermal hyperalgesia. We detected an obvious increase in MOR expression levels but insignificant changes in KOR and DOR levels in the spinal cord after PI. Moreover, we found that oxycodone was able to reverse the increased expression of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), tyrosine kinase receptor (TrK) A, and TrkB and the decreased expression of NT-3 and TrkC, after PI. Pretreatment with oxycodone also altered the expression of these mediators. Conclusion Based on the results, possible underlying mechanisms for the antinociceptive properties of oxycodone in acute postoperative pain include the activation of MOR downstream signaling and the regulation of NTs and receptor expression through attenuation of glial activation and fortification of antinociceptive mediators in the spinal cord. This study may provide new insights into the molecular mechanisms underlying the analgesic action of oxycodone.
Collapse
Affiliation(s)
- Baowen Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| | - Yi Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| | - Ningbo Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| | - Jin Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| | - Xianwei Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| |
Collapse
|
11
|
Guerrero-Alba R, Valdez-Morales EE, Jiménez-Vargas NN, Bron R, Poole D, Reed D, Castro J, Campaniello M, Hughes PA, Brierley SM, Bunnett N, Lomax AE, Vanner S. Co-expression of μ and δ opioid receptors by mouse colonic nociceptors. Br J Pharmacol 2018; 175:2622-2634. [PMID: 29579315 DOI: 10.1111/bph.14222] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE To better understand opioid signalling in visceral nociceptors, we examined the expression and selective activation of μ and δ opioid receptors by dorsal root ganglia (DRG) neurons innervating the mouse colon. EXPERIMENTAL APPROACH DRG neurons projecting to the colon were identified by retrograde tracing. δ receptor-GFP reporter mice, in situ hybridization, single-cell RT-PCR and μ receptor-specific antibodies were used to characterize expression of μ and δ receptors. Voltage-gated Ca2+ currents and neuronal excitability were recorded in small diameter nociceptive neurons (capacitance <30 pF) by patch clamp and ex vivo single-unit afferent recordings were obtained from the colon. KEY RESULTS In situ hybridization of oprm1 expression in Fast Blue-labelled DRG neurons was observed in 61% of neurons. μ and δ receptors were expressed by 36-46% of colon DRG neurons, and co-expressed by ~25% of neurons. μ and δ receptor agonists inhibited Ca2+ currents in DRG, effects blocked by opioid antagonists. One or both agonists inhibited action potential firing by colonic afferent endings. Incubation of neurons with supernatants from inflamed colon segments inhibited Ca2+ currents and neuronal excitability. Antagonists of μ, but not δ receptors, inhibited the effects of these supernatant on Ca2+ currents, whereas both antagonists inhibited their actions on neuronal excitability. CONCLUSIONS AND IMPLICATIONS A significant number of small diameter colonic nociceptors co-express μ and δ receptors and are inhibited by agonists and endogenous opioids in inflamed tissues. Thus, opioids that act at μ or δ receptors, or their heterodimers may be effective in treating visceral pain.
Collapse
Affiliation(s)
- Raquel Guerrero-Alba
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | | | | | - Romke Bron
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Daniel Poole
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - David Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - Joel Castro
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Melissa Campaniello
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Patrick A Hughes
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Nigel Bunnett
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC, Australia.,Departments of Surgery and Pharmacology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - Stephen Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, ON, Canada
| |
Collapse
|
12
|
Raffa RB, Pergolizzi JV, Taylor R, Ossipov MH. Indirect-acting strategy of opioid action instead of direct receptor activation: dual-acting enkephalinase inhibitors (DENKIs). J Clin Pharm Ther 2018; 43:443-449. [DOI: 10.1111/jcpt.12687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/19/2018] [Indexed: 01/26/2023]
Affiliation(s)
- R. B. Raffa
- University of Arizona College of Pharmacy; Tucson AZ USA
- Temple University School of Pharmacy; Philadelphia PA USA
| | | | | | - M. H. Ossipov
- University of Arizona College of Medicine; Tucson AZ USA
| | | |
Collapse
|
13
|
Hsieh YL, Kan HW, Chiang H, Lee YC, Hsieh ST. Distinct TrkA and Ret modulated negative and positive neuropathic behaviors in a mouse model of resiniferatoxin-induced small fiber neuropathy. Exp Neurol 2017; 300:87-99. [PMID: 29106982 DOI: 10.1016/j.expneurol.2017.10.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/21/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022]
Abstract
Neurotrophic factors and their corresponding receptors play key roles in the maintenance of different phenotypic dorsal root ganglion (DRG) neurons, the axons of which degenerate in small fiber neuropathy, leading to various neuropathic manifestations. Mechanisms underlying positive and negative symptoms of small fiber neuropathy have not been systematically explored. This study investigated the molecular basis of these seemingly paradoxical neuropathic behaviors according to the profiles of TrkA and Ret with immunohistochemical and pharmacological interventions in a mouse model of resiniferatoxin (RTX)-induced small fiber neuropathy. Mice with RTX neuropathy exhibited thermal hypoalgesia and mechanical allodynia, reduced skin innervation, and altered DRG expression profiles with decreased TrkA(+) neurons and increased Ret(+) neurons. RTX neuropathy induced the expression of activating transcription factor 3 (ATF3), and ATF3(+) neurons were colocalized with Ret but not with TrkA (P<0.001). As a neuroprotectant, 4-Methylcatechol (4MC) promoted skin reinnervation partially with correlated reversal of the neuropathic behaviors and altered neurochemical expression. Gambogic amide, a selective TrkA agonist, normalized thermal hypoalgesia, and GW441756, a TrkA kinase inhibitor, induced thermal hypoalgesia, which was already reversed by 4MC. Mechanical allodynia was reversed by a Ret kinase inhibitor, AST487, which induced thermal hyperalgesia in naïve mice. The activation of Ret signaling by XIB4035 induced mechanical allodynia and thermal hypoalgesia in RTX neuropathy mice in which the neuropathic behaviors were previously normalized by 4MC. Distinct neurotrophic factor receptors, TrkA and Ret, accounted for negative and positive neuropathic behaviors in RTX-induced small fiber neuropathy, respectively: TrkA for thermal hypoalgesia and Ret for mechanical allodynia and thermal hypoalgesia.
Collapse
Affiliation(s)
- Yu-Lin Hsieh
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| | - Hung-Wei Kan
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Hao Chiang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan; Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02115, USA; Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | - Yi-Chen Lee
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan; Department of Neurology, National Taiwan University Hospital, Taipei 10002, Taiwan; Graduate Institute of Brain and Mind Science, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
14
|
François A, Scherrer G. Delta Opioid Receptor Expression and Function in Primary Afferent Somatosensory Neurons. Handb Exp Pharmacol 2017; 247:87-114. [PMID: 28993838 DOI: 10.1007/164_2017_58] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The functional diversity of primary afferent neurons of the dorsal root ganglia (DRG) generates a variety of qualitatively and quantitatively distinct somatosensory experiences, from shooting pain to pleasant touch. In recent years, the identification of dozens of genetic markers specifically expressed by subpopulations of DRG neurons has dramatically improved our understanding of this diversity and provided the tools to manipulate their activity and uncover their molecular identity and function. Opioid receptors have long been known to be expressed by discrete populations of DRG neurons, in which they regulate cell excitability and neurotransmitter release. We review recent insights into the identity of the DRG neurons that express the delta opioid receptor (DOR) and the ion channel mechanisms that DOR engages in these cells to regulate sensory input. We highlight recent findings derived from DORGFP reporter mice and from in situ hybridization and RNA sequencing studies in wild-type mice that revealed DOR presence in cutaneous mechanosensory afferents eliciting touch and implicated in tactile allodynia. Mechanistically, we describe how DOR modulates opening of voltage-gated calcium channels (VGCCs) to control glutamatergic neurotransmission between somatosensory neurons and postsynaptic neurons in the spinal cord dorsal horn. We additionally discuss other potential signaling mechanisms, including those involving potassium channels, which DOR may engage to fine tune somatosensation. We conclude by discussing how this knowledge may explain the analgesic properties of DOR agonists against mechanical pain and uncovers an unanticipated specialized function for DOR in cutaneous mechanosensation.
Collapse
Affiliation(s)
- Amaury François
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA. .,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA. .,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
15
|
Yaksh TL, Fisher CJ, Hockman TM, Wiese AJ. Current and Future Issues in the Development of Spinal Agents for the Management of Pain. Curr Neuropharmacol 2017; 15:232-259. [PMID: 26861470 PMCID: PMC5412694 DOI: 10.2174/1570159x14666160307145542] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 12/02/2015] [Accepted: 02/05/2016] [Indexed: 11/22/2022] Open
Abstract
Targeting analgesic drugs for spinal delivery reflects the fact that while the conscious experience of pain is mediated supraspinally, input initiated by high intensity stimuli, tissue injury and/or nerve injury is encoded at the level of the spinal dorsal horn and this output informs the brain as to the peripheral environment. This encoding process is subject to strong upregulation resulting in hyperesthetic states and downregulation reducing the ongoing processing of nociceptive stimuli reversing the hyperesthesia and pain processing. The present review addresses the biology of spinal nociceptive processing as relevant to the effects of intrathecally-delivered drugs in altering pain processing following acute stimulation, tissue inflammation/injury and nerve injury. The review covers i) the major classes of spinal agents currently employed as intrathecal analgesics (opioid agonists, alpha 2 agonists; sodium channel blockers; calcium channel blockers; NMDA blockers; GABA A/B agonists; COX inhibitors; ii) ongoing developments in the pharmacology of spinal therapeutics focusing on less studied agents/targets (cholinesterase inhibition; Adenosine agonists; iii) novel intrathecal targeting methodologies including gene-based approaches (viral vectors, plasmids, interfering RNAs); antisense, and toxins (botulinum toxins; resniferatoxin, substance P Saporin); and iv) issues relevant to intrathecal drug delivery (neuraxial drug distribution), infusate delivery profile, drug dosing, formulation and principals involved in the preclinical evaluation of intrathecal drug safety.
Collapse
Affiliation(s)
- Tony L. Yaksh
- University of California, San Diego, Anesthesia Research Lab 0818, 9500 Gilman Dr. LaJolla, CA 92093, USA
| | - Casey J. Fisher
- University of California, San Diego, Anesthesia Research Lab 0818, 9500 Gilman Dr. LaJolla, CA 92093, USA
| | - Tyler M. Hockman
- University of California, San Diego, Anesthesia Research Lab 0818, 9500 Gilman Dr. LaJolla, CA 92093, USA
| | - Ashley J. Wiese
- University of California, San Diego, Anesthesia Research Lab 0818, 9500 Gilman Dr. LaJolla, CA 92093, USA
| |
Collapse
|
16
|
Auh QS, Chun YH, Melemedjian OK, Zhang Y, Ro JY. Peripheral interactions between cannabinoid and opioid receptor agonists in a model of inflammatory mechanical hyperalgesia. Brain Res Bull 2016; 125:211-7. [PMID: 27450703 DOI: 10.1016/j.brainresbull.2016.07.009] [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: 05/06/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 01/28/2023]
Abstract
Activation of opioid and cannabinoid receptors expressed in nociceptors induces effective antihyperalgesia. In this study, we examined whether combinations of opioid and cannabinoid receptor agonists directed at the injured site would enhance therapeutic effectiveness. Behavioral pharmacology experiments were performed to compare the effects of DAMGO, a selective agonist for μ-opioid receptor (MOR), ACPA, a specific agonist for CB1, and combinations of DAMGO and ACPA in attenuating complete Freund's adjuvant (CFA)-induced mechanical hyperalgesia in the rat hindpaw. DAMGO (1μg-1mg) or ACPA (1μg-2mg) was administered into the inflamed paw when mechanical hyperalgesia was fully developed. When administered individually, DAMGO and ACPA dose-dependently reversed the mechanical hyperalgesia. DAMGO displayed a lower ED50 value (57.4±2.49μg) than ACPA (111.6±2.18μg), but ACPA produced longer lasting antihyperalgesic effects. Combinations of DAMGO and ACPA also dose-dependently attenuated mechanical hyperalgesia, but the antihyperalgesic effects were partial and transient even at high doses. Using isobolographic analysis, we determined that combined treatment with DAMGO and ACPA produced antagonistic effects with the observed ED50 of 128.4±2.28μg. Our findings showed that MOR and CB1 agonists directed at the inflamed site effectively attenuate mechanical hyperalgesia when administered individually, but exert opposing effects when administered together. The antagonistic interactions between the two classes of drugs at the inflamed site suggest distinct mechanisms unique to peripheral nociceptors or inflamed tissue, and therefore require further studies to investigate whether the therapeutic utility of the combined drug treatments in chronic pain conditions can be optimized.
Collapse
Affiliation(s)
- Q-Schick Auh
- Kyung Hee University, School of Dentistry, Department of Oral Medicine, 1 Hoegi Dong, Dongdaemun Gu, Seoul, Republic of Korea
| | - Yang Hyun Chun
- Kyung Hee University, School of Dentistry, Department of Oral Medicine, 1 Hoegi Dong, Dongdaemun Gu, Seoul, Republic of Korea
| | - Ohannes K Melemedjian
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore St., Baltimore, MD 21201, USA
| | - Youping Zhang
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore St., Baltimore, MD 21201, USA
| | - Jin Y Ro
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore St., Baltimore, MD 21201, USA; Kyung Hee University, School of Dentistry, Department of Oral Medicine, 1 Hoegi Dong, Dongdaemun Gu, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Yao P, Ding Y, Wang Z, Ma J, Hong T, Zhu Y, Li H, Pan S. Impacts of anti-nerve growth factor antibody on pain-related behaviors and expressions of opioid receptor in spinal dorsal horn and dorsal root ganglia of rats with cancer-induced bone pain. Mol Pain 2016; 12:12/0/1744806916644928. [PMID: 27118770 PMCID: PMC4955994 DOI: 10.1177/1744806916644928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/20/2016] [Indexed: 11/15/2022] Open
Abstract
Objective To investigate the impacts of anti-nerve growth factor antibody on pain-related behaviors and expressions of μ-opioid receptor in spinal dorsal horn and dorsal root ganglia of rats with cancer-induced bone pain. Methods The rats were randomly grouped and then injected with 10 μl of phosphate buffer saline or Walker256 tumor cells into the upper segment of left tibia. Thirteen days after the injection, the intrathecal catheterization was performed, followed by the injection of saline, anti-nerve growth factor, nerve growth factor, and naloxone twice a day. The pain ethological changes were measured at the set time points; the expression changes of μ-opioid receptor protein and mRNA in spinal dorsal horn and dorsal root ganglia were detected on the 18th day. Results After the tumor cells were injected into the tibia, hyperalgesia appeared and the expression of μ-opioid receptor protein and mRNA in spinal dorsal horn and dorsal root ganglia was increased, compared with the sham group; after intrathecally injected anti-nerve growth factor, the significant antinociceptive effects appeared, and the μ-opioid receptor expression was increased, compared with the cancer pain group; the μ-opioid receptor expressions in the other groups showed no statistical significance. The naloxone pretreatment could mostly inverse the antinociception effects of anti-nerve growth factor. Conclusions Anti-nerve growth factor could reduce hyperalgesia in the cancer-induced bone pain rats, and the antinociceptive effects were related with the upregulation of μ-opioid receptor.
Collapse
Affiliation(s)
- Peng Yao
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Ding
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhibin Wang
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiaming Ma
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Hong
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongqiang Zhu
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongxi Li
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shinong Pan
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Xiao J, Zeng S, Wang X, Babazada H, Li Z, Liu R, Yu W. Neurokinin 1 and opioid receptors: relationships and interactions in nervous system. TRANSLATIONAL PERIOPERATIVE AND PAIN MEDICINE 2016; 1:11-21. [PMID: 28409174 PMCID: PMC5388438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Opioid receptors and neurokinin 1 receptor (NK1R) are found highly expressed in the central nervous system. The co-localization of these two kinds of receptors suggests that they might interact with each other in both the transmission and modulation of the pain signal. In this review, we explore the relationships between opioid receptors and NK1R. Substance P (SP) plays a modulatory role in the pain transmission by activating the NK1R. Opioid receptor activation can inhibit SP release. NK1R is found participating in the mechanisms of the side effects of the opioids, including opioid analgesic tolerance, hyperalgesia, anxiety behaviors of morphine reward and opioids related respiratory depression. A series of compounds such as NK1R antagonists and ligands works on both mu/delta opioid receptor (MOR/DOR) and NK1R were synthesized as novel analgesics that enhance the clinical pain management efficacy and reduce the dosage and side effects. The current status of these novel ligands and the limitations are discussed in this review. Although the working mechanisms of these ligands remained unclear, they could be used as research tool for developing novel analgesic drugs in the future.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania
| | - Si Zeng
- Department of Anesthesiology, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, Electronic Science and Technology University
| | - Xiangrui Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Hasan Babazada
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania
| | | | - Renyu Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| |
Collapse
|
19
|
Merighi A. Targeting the glial-derived neurotrophic factor and related molecules for controlling normal and pathologic pain. Expert Opin Ther Targets 2015; 20:193-208. [PMID: 26863504 DOI: 10.1517/14728222.2016.1085972] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Glial-derived neurotrophic factor (GDNF) and its family of ligands (GFLs) have several functions in the nervous system. As a survival factor for dopaminergic neurons, GDNF was used in clinical trials for Parkinson's disease. GFLs and their receptors are also potential targets for new pain-controlling drugs. Although molecules with analgesic activities in rodents mostly failed to be effective in translational studies, this potential should not be underestimated. AREAS COVERED The circuitry, molecular, and cellular mechanisms by which GFLs control nociception and their intervention in inflammatory and neuropathic pain are considered first. The problems related to effective GDNF delivery to the brain and the possibility to target the GFL receptor complex rather than its ligands are then discussed, also considering the use of non-peptidyl agonists. EXPERT OPINION In nociceptive pathways, an ideal drug should either: i) target the release of endogenous GFLs from large dense-cored vesicles (LGVs) by acting, for example, onto the phosphatidylinositol-3-phosphate [PtdIns(3)P] pool, which is sensitive to Ca(2+) modulation, or ii) target the GFL receptor complex. Besides XIB403, a tiol molecule that enhances GFRα family receptor signaling, existing drugs such as retinoic acid and amitriptyline should be considered for effective targeting of GDNF, at least in neuropathic pain. The approach of pain modeling in experimental animals is discussed.
Collapse
Affiliation(s)
- Adalberto Merighi
- a University of Turin, Department of Veterinary Sciences , Grugliasco, TO, Italy ;
| |
Collapse
|
20
|
Wang F, Cai B, Li KC, Hu XY, Lu YJ, Wang Q, Bao L, Zhang X. FXYD2, a γ subunit of Na⁺, K⁺-ATPase, maintains persistent mechanical allodynia induced by inflammation. Cell Res 2015; 25:318-34. [PMID: 25633594 DOI: 10.1038/cr.2015.12] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 10/09/2014] [Accepted: 12/15/2014] [Indexed: 01/08/2023] Open
Abstract
Na⁺, K⁺-ATPase (NKA) is required to generate the resting membrane potential in neurons. Nociceptive afferent neurons express not only the α and β subunits of NKA but also the γ subunit FXYD2. However, the neural function of FXYD2 is unknown. The present study shows that FXYD2 in nociceptive neurons is necessary for maintaining the mechanical allodynia induced by peripheral inflammation. FXYD2 interacted with α1NKA and negatively regulated the NKA activity, depolarizing the membrane potential of nociceptive neurons. Mechanical allodynia initiated in FXYD2-deficient mice was abolished 4 days after inflammation, whereas it persisted for at least 3 weeks in wild-type mice. Importantly, the FXYD2/α1NKA interaction gradually increased after inflammation and peaked on day 4 post inflammation, resulting in reduction of NKA activity, depolarization of neuron membrane and facilitation of excitatory afferent neurotransmission. Thus, the increased FXYD2 activity may be a fundamental mechanism underlying the persistent hypersensitivity to pain induced by inflammation.
Collapse
Affiliation(s)
- Feng Wang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bing Cai
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kai-Cheng Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu-Ye Hu
- Shanghai Clinical Center, Chinese Academy of Sciences/XuHui Central Hospital, Shanghai, China
| | - Ying-Jin Lu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiong Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lan Bao
- 1] State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China [2] School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Xu Zhang
- 1] Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China [2] School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| |
Collapse
|
21
|
Schuster DJ, Metcalf MD, Kitto KF, Messing RO, Fairbanks CA, Wilcox GL. Ligand requirements for involvement of PKCε in synergistic analgesic interactions between spinal μ and δ opioid receptors. Br J Pharmacol 2014; 172:642-53. [PMID: 24827408 DOI: 10.1111/bph.12774] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE We recently found that PKCε was required for spinal analgesic synergy between two GPCRs, δ opioid receptors and α2 A adrenoceptors, co-located in the same cellular subpopulation. We sought to determine if co-delivery of μ and δ opioid receptor agonists would similarly result in synergy requiring PKCε. EXPERIMENTAL APPROACH Combinations of μ and δ opioid receptor agonists were co-administered intrathecally by direct lumbar puncture to PKCε-wild-type (PKCε-WT) and -knockout (PKCε-KO) mice. Antinociception was assessed using the hot-water tail-flick assay. Drug interactions were evaluated by isobolographic analysis. KEY RESULTS All agonists produced comparable antinociception in both PKCε-WT and PKCε-KO mice. Of 19 agonist combinations that produced analgesic synergy, only 3 required PKCε for a synergistic interaction. In these three combinations, one of the agonists was morphine, although not all combinations involving morphine required PKCε. Morphine + deltorphin II and morphine + deltorphin I required PKCε for synergy, whereas a similar combination, morphine + deltorphin, did not. Additionally, morphine + oxymorphindole required PKCε for synergy, whereas a similar combination, morphine + oxycodindole, did not. CONCLUSIONS AND IMPLICATIONS We discovered biased agonism for a specific signalling pathway at the level of spinally co-delivered opioid agonists. As the bias is only revealed by an appropriate ligand combination and cannot be accounted for by a single drug, it is likely that the receptors these agonists act on are interacting with each other. Our results support the existence of μ and δ opioid receptor heteromers at the spinal level in vivo. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- D J Schuster
- Department of Neuroscience, Medical School, University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | | |
Collapse
|
22
|
Zhang X, Bao L, Li S. Opioid receptor trafficking and interaction in nociceptors. Br J Pharmacol 2014; 172:364-74. [PMID: 24611685 DOI: 10.1111/bph.12653] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/29/2014] [Accepted: 02/17/2014] [Indexed: 01/15/2023] Open
Abstract
UNLABELLED Opiate analgesics such as morphine are often used for pain therapy. However, antinociceptive tolerance and dependence may develop with long-term use of these drugs. It was found that μ-opioid receptors can interact with δ-opioid receptors, and morphine antinociceptive tolerance can be reduced by blocking δ-opioid receptors. Recent studies have shown that μ- and δ-opioid receptors are co-expressed in a considerable number of small neurons in the dorsal root ganglion. The interaction of μ-opioid receptors with δ-opioid receptors in the nociceptive afferents is facilitated by the stimulus-induced cell-surface expression of δ-opioid receptors, and contributes to morphine tolerance. Further analysis of the molecular, cellular and neural circuit mechanisms that regulate the trafficking and interaction of opioid receptors and related signalling molecules in the pain pathway would help to elucidate the mechanism of opiate analgesia and improve pain therapy. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- X Zhang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai, China
| | | | | |
Collapse
|
23
|
Gendron L, Mittal N, Beaudry H, Walwyn W. Recent advances on the δ opioid receptor: from trafficking to function. Br J Pharmacol 2014; 172:403-19. [PMID: 24665909 DOI: 10.1111/bph.12706] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Within the opioid family of receptors, δ (DOPrs) and μ opioid receptors (MOPrs) are typical GPCRs that activate canonical second-messenger signalling cascades to influence diverse cellular functions in neuronal and non-neuronal cell types. These receptors activate well-known pathways to influence ion channel function and pathways such as the map kinase cascade, AC and PI3K. In addition new information regarding opioid receptor-interacting proteins, downstream signalling pathways and resultant functional effects has recently come to light. In this review, we will examine these novel findings focusing on the DOPr and, in doing so, will contrast and compare DOPrs with MOPrs in terms of differences and similarities in function, signalling pathways, distribution and interactions. We will also discuss and clarify issues that have recently surfaced regarding the expression and function of DOPrs in different cell types and analgesia. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- Louis Gendron
- Département de physiologie et biophysique, Institut de pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|
24
|
Viet CT, Dang D, Ye Y, Ono K, Campbell RR, Schmidt BL. Demethylating drugs as novel analgesics for cancer pain. Clin Cancer Res 2014; 20:4882-4893. [PMID: 24963050 DOI: 10.1158/1078-0432.ccr-14-0901] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE In this study, we evaluated the analgesic potential of demethylating drugs on oral cancer pain. Although demethylating drugs could affect expression of many genes, we focused on the mu-opioid receptor (OPRM1) gene pathway, because of its role in pain processing. We determined the antinociceptive effect of OPRM1 re-expression in a mouse oral cancer model. EXPERIMENTAL DESIGN Using a mouse oral cancer model, we determined whether demethylating drugs produced antinociception through re-expression of OPRM1. We then re-expressed OPRM1 with adenoviral transduction and determined if, and by what mechanism, OPRM1 re-expression produced antinociception. To determine the clinical significance of OPRM1 on cancer pain, we quantified OPRM1 methylation in painful cancer tissues and nonpainful contralateral normal tissues of patients with oral cancer, and nonpainful dysplastic tissues of patients with oral dysplasia. RESULTS We demonstrated that OPRM1 was methylated in cancer tissue, but not normal tissue, of patients with oral cancer, and not in dysplastic tissues from patients with oral dysplasia. Treatment with demethylating drugs resulted in mechanical and thermal antinociception in the mouse cancer model. This behavioral change correlated with OPRM1 re-expression in the cancer and associated neurons. Similarly, adenoviral-mediated OPRM1 re-expression on cancer cells resulted in naloxone-reversible antinociception. OPRM1 re-expression on oral cancer cells in vitro increased β-endorphin secretion from the cancer, and decreased activation of neurons that were treated with cancer supernatant. CONCLUSION Our study establishes the regulatory role of methylation in cancer pain. OPRM1 re-expression in cancer cells produces antinociception through cancer-mediated endogenous opioid secretion. Demethylating drugs have an analgesic effect that involves OPRM1.
Collapse
Affiliation(s)
- Chi T Viet
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Dongmin Dang
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Yi Ye
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Kentaro Ono
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Ronald R Campbell
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Brian L Schmidt
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| |
Collapse
|
25
|
Bardoni R, Tawfik VL, Wang D, François A, Solorzano C, Shuster SA, Choudhury P, Betelli C, Cassidy C, Smith K, de Nooij JC, Mennicken F, O'Donnell D, Kieffer BL, Woodbury CJ, Basbaum AI, MacDermott AB, Scherrer G. Delta opioid receptors presynaptically regulate cutaneous mechanosensory neuron input to the spinal cord dorsal horn. Neuron 2014; 81:1312-1327. [PMID: 24583022 DOI: 10.1016/j.neuron.2014.01.044] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2014] [Indexed: 11/24/2022]
Abstract
Cutaneous mechanosensory neurons detect mechanical stimuli that generate touch and pain sensation. Although opioids are generally associated only with the control of pain, here we report that the opioid system in fact broadly regulates cutaneous mechanosensation, including touch. This function is predominantly subserved by the delta opioid receptor (DOR), which is expressed by myelinated mechanoreceptors that form Meissner corpuscles, Merkel cell-neurite complexes, and circumferential hair follicle endings. These afferents also include a small population of CGRP-expressing myelinated nociceptors that we now identify as the somatosensory neurons that coexpress mu and delta opioid receptors. We further demonstrate that DOR activation at the central terminals of myelinated mechanoreceptors depresses synaptic input to the spinal dorsal horn, via the inhibition of voltage-gated calcium channels. Collectively our results uncover a molecular mechanism by which opioids modulate cutaneous mechanosensation and provide a rationale for targeting DOR to alleviate injury-induced mechanical hypersensitivity.
Collapse
Affiliation(s)
- Rita Bardoni
- Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Dong Wang
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Amaury François
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Carlos Solorzano
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Scott A Shuster
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Papiya Choudhury
- Department of Physiology and Cellular Biophysics, Department of Neuroscience, Columbia University, New York, NY 10032, USA
| | - Chiara Betelli
- Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Colleen Cassidy
- Graduate Program in Neuroscience, University of Wyoming, Laramie, WY 82071, USA; Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | - Kristen Smith
- Graduate Program in Neuroscience, University of Wyoming, Laramie, WY 82071, USA; Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | - Joriene C de Nooij
- Departments of Neuroscience and Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Françoise Mennicken
- AstraZeneca R&D Montreal, Department of Translational Science, Montreal, QC H4S 1Z9, Canada
| | - Dajan O'Donnell
- AstraZeneca R&D Montreal, Department of Translational Science, Montreal, QC H4S 1Z9, Canada
| | - Brigitte L Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR7104 CNRS/Université de Strasbourg, U964 INSERM, 67400 Illkirch, France
| | - C Jeffrey Woodbury
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | - Allan I Basbaum
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Amy B MacDermott
- Department of Physiology and Cellular Biophysics, Department of Neuroscience, Columbia University, New York, NY 10032, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA.
| |
Collapse
|
26
|
Ye Y, Bae SS, Viet CT, Troob S, Bernabé D, Schmidt BL. IB4(+) and TRPV1(+) sensory neurons mediate pain but not proliferation in a mouse model of squamous cell carcinoma. Behav Brain Funct 2014; 10:5. [PMID: 24524628 PMCID: PMC3942073 DOI: 10.1186/1744-9081-10-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 01/30/2014] [Indexed: 11/20/2022] Open
Abstract
Background Cancer pain severely limits function and significantly reduces quality of life. Subtypes of sensory neurons involved in cancer pain and proliferation are not clear. Methods We produced a cancer model by inoculating human oral squamous cell carcinoma (SCC) cells into the hind paw of athymic mice. We quantified mechanical and thermal nociception using the paw withdrawal assays. Neurotoxins isolectin B4-saporin (IB4-SAP), or capsaicin was injected intrathecally to selectively ablate IB4(+) neurons or TRPV1(+) neurons, respectively. JNJ-17203212, a TRPV1 antagonist, was also injected intrathecally. TRPV1 protein expression in the spinal cord was quantified with western blot. Paw volume was measured by a plethysmometer and was used as an index for tumor size. Ki-67 immunostaining in mouse paw sections was performed to evaluate cancer proliferation in situ. Results We showed that mice with SCC exhibited both mechanical and thermal hypersensitivity. Selective ablation of IB4(+) neurons by IB4-SAP decreased mechanical allodynia in mice with SCC. Selective ablation of TRPV1(+) neurons by intrathecal capsaicin injection, or TRPV1 antagonism by JNJ-17203212 in the IB4-SAP treated mice completely reversed SCC-induced thermal hyperalgesia, without affecting mechanical allodynia. Furthermore, TRPV1 protein expression was increased in the spinal cord of SCC mice compared to normal mice. Neither removal of IB4(+) or TRPV1(+) neurons affected SCC proliferation. Conclusions We show in a mouse model that IB4(+) neurons play an important role in cancer-induced mechanical allodynia, while TRPV1 mediates cancer-induced thermal hyperalgesia. Characterization of the sensory fiber subtypes responsible for cancer pain could lead to the development of targeted therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Brian L Schmidt
- Bluestone Center for Clinical Research, New York University, New York, USA.
| |
Collapse
|
27
|
Kouchek M, Takasusuki T, Terashima T, Yaksh TL, Xu Q. Effects of intrathecal SNC80, a delta receptor ligand, on nociceptive threshold and dorsal horn substance p release. J Pharmacol Exp Ther 2013; 347:258-64. [PMID: 23978562 PMCID: PMC3807063 DOI: 10.1124/jpet.113.206573] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/26/2013] [Indexed: 12/22/2022] Open
Abstract
Delta-opioid receptors (DOR) are present in the superficial dorsal horn and are believed to regulate the release of small afferent transmitters as evidenced by the effects of spinally delivered delta-opioid preferring peptides. Here we examined the effects of intrathecal SNC80 [(+)-4-[α(R)-α-[(2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl]-3-(methoxybenzyl)-N,N-diethylbenzamide], a selective nonpeptidic DOR agonist, in three preclinical pain models, acute thermal escape, intraplantar carrageenan-tactile allodynia, and intraplantar formalin flinches, and on the evoked release of substance P (SP) from small primary afferents. Rats with chronic intrathecal catheters received intrathecal vehicle or SNC80 (100 or 200 μg). Intrathecal SNC80 did not change acute thermal latencies or carrageenan-induced thermal hyperalgesia. However, SNC80 attenuated carrageenan-induced tactile allodynia and significantly reduced both phase 1 and phase 2 formalin-induced paw flinches, as assessed by an automatic flinch counting device. These effects were abolished by naltrindole (3 mg/kg i.p.), a selective DOR antagonist, but not CTOP (10 µg i.t.), a selective MOR antagonist. Furthermore, intrathecal SNC80 (200 μg) blocked formalin-induced substance P release otherwise evoked in the ispilateral superficial dorsal horn as measured by NK1 receptor internalization. In conclusion, intrathecal SNC80 alleviated pain hypersensitivity after peripheral inflammation in a fashion paralleling its ability to block peptide transmitter release from small peptidergic afferents, which by its pharmacology appears to represent an effect mediated by a spinal DOR.
Collapse
Affiliation(s)
- Milad Kouchek
- Department of Anesthesiology, University of California San Diego, La Jolla, California (M.K., T.L.Y., Q.X.); Department of Pharmacology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden (M.K.); and Department of Anesthesiology, Dokkyo Medical University, School of Medicine, Mibu, Tochigi Prefecture, Japan (T.Ta., T.Te.)
| | | | | | | | | |
Collapse
|
28
|
Iwaszkiewicz KS, Schneider JJ, Hua S. Targeting peripheral opioid receptors to promote analgesic and anti-inflammatory actions. Front Pharmacol 2013; 4:132. [PMID: 24167491 PMCID: PMC3807052 DOI: 10.3389/fphar.2013.00132] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/04/2013] [Indexed: 01/25/2023] Open
Abstract
Mechanisms of endogenous pain control are significant. Increasing studies have clearly produced evidence for the clinical usefulness of opioids in peripheral analgesia. The immune system uses mechanisms of cell migration not only to fight pathogens but also to control pain and inflammation within injured tissue. It has been demonstrated that peripheral inflammatory pain can be effectively controlled by an interaction of immune cell-derived opioid peptides with opioid receptors on peripheral sensory nerve terminals. Experimental and clinical studies have clearly shown that activation of peripheral opioid receptors with exogenous opioid agonists and endogenous opioid peptides are able to produce significant analgesic and anti-inflammatory effects, without central opioid mediated side effects (e.g., respiratory depression, sedation, tolerance, dependence). This article will focus on the role of opioids in peripheral inflammatory conditions and the clinical implications of targeting peripheral opioid receptors.
Collapse
Affiliation(s)
- Katerina S Iwaszkiewicz
- The School of Biomedical Sciences and Pharmacy, The University of Newcastle Callaghan, NSW, Australia
| | | | | |
Collapse
|
29
|
Weibel R, Reiss D, Karchewski L, Gardon O, Matifas A, Filliol D, Becker JAJ, Wood JN, Kieffer BL, Gaveriaux-Ruff C. Mu opioid receptors on primary afferent nav1.8 neurons contribute to opiate-induced analgesia: insight from conditional knockout mice. PLoS One 2013; 8:e74706. [PMID: 24069332 PMCID: PMC3771900 DOI: 10.1371/journal.pone.0074706] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 08/05/2013] [Indexed: 12/26/2022] Open
Abstract
Opiates are powerful drugs to treat severe pain, and act via mu opioid receptors distributed throughout the nervous system. Their clinical use is hampered by centrally-mediated adverse effects, including nausea or respiratory depression. Here we used a genetic approach to investigate the potential of peripheral mu opioid receptors as targets for pain treatment. We generated conditional knockout (cKO) mice in which mu opioid receptors are deleted specifically in primary afferent Nav1.8-positive neurons. Mutant animals were compared to controls for acute nociception, inflammatory pain, opiate-induced analgesia and constipation. There was a 76% decrease of mu receptor-positive neurons and a 60% reduction of mu-receptor mRNA in dorsal root ganglia of cKO mice. Mutant mice showed normal responses to heat, mechanical, visceral and chemical stimuli, as well as unchanged morphine antinociception and tolerance to antinociception in models of acute pain. Inflammatory pain developed similarly in cKO and controls mice after Complete Freund's Adjuvant. In the inflammation model, however, opiate-induced (morphine, fentanyl and loperamide) analgesia was reduced in mutant mice as compared to controls, and abolished at low doses. Morphine-induced constipation remained intact in cKO mice. We therefore genetically demonstrate for the first time that mu opioid receptors partly mediate opiate analgesia at the level of Nav1.8-positive sensory neurons. In our study, this mechanism operates under conditions of inflammatory pain, but not nociception. Previous pharmacology suggests that peripheral opiates may be clinically useful, and our data further demonstrate that Nav1.8 neuron-associated mu opioid receptors are feasible targets to alleviate some forms of persistent pain.
Collapse
Affiliation(s)
- Raphaël Weibel
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
| | - David Reiss
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
| | - Laurie Karchewski
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
| | - Olivier Gardon
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
| | - Audrey Matifas
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
| | - Dominique Filliol
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
| | - Jérôme A. J. Becker
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical research, University College London, London, United Kingdom
| | - Brigitte L. Kieffer
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
| | - Claire Gaveriaux-Ruff
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Translational Medicine and Neurogenetic Programme, UdS Université de Strasbourg, INSERM U964, CNRS UMR7104, Illkirch, France
- ESBS, École Supérieure de Biotechnologie de Strasbourg, UdS Université de Strasbourg, Strasbourg, France
- * E-mail:
| |
Collapse
|
30
|
Systemic morphine treatment induces changes in firing patterns and responses of nociceptive afferent fibers in mouse glabrous skin. Pain 2013; 154:2297-2309. [PMID: 23711478 DOI: 10.1016/j.pain.2013.05.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/26/2013] [Accepted: 05/17/2013] [Indexed: 01/22/2023]
Abstract
Patients receiving opioids for pain may experience decreased effectiveness of the drug and even abnormal pain sensitivity-hyperalgesia and/or allodynia. We hypothesized that peripheral nociceptor hyperexcitability contributes to opioid-induced hyperalgesia and tested this using an in vitro mouse glabrous skin-nerve preparation. Mice were injected intraperitoneally with escalating doses of morphine (5, 8, 10, 15 mg/kg) or saline every 12 hours for 48 hours and killed approximately 12 hours after the last injection. Receptive fields of nociceptors were tested for mechanical, heat, and cold sensitivity. Activity was also measured during an initial 2-minute period and during 5-minute periods between stimuli. Aberrant activity was common in fibers from morphine-treated mice but rare in saline-treated mice. Resting background activity was elevated in C-fibers from morphine-treated mice. Both C- and Aδ-fibers had afterdischarge in response to mechanical, heat, and/or cold stimulation of the skin as well as spontaneous, unevoked activity. Compared to saline, morphine treatment increased the proportion of fibers displaying polymodal rather than mechanical-only responses. A significant increase in Aδ-mechanoreceptive fibers responding to cold accounted for most of this change. In agreement with this, morphine-treated mice showed increased sensitivity in the cold tail flick test. In morphine-treated mice, aberrant activity and hyperexcitability of nociceptors could contribute to increased pain sensitivity. Importantly, this activity is likely driving central sensitization, a phenomenon contributing to abnormal sensory processing and chronic pain. If similar changes occur in human patients, aberrant nociceptor activity is likely to be interpreted as pain and could contribute to opioid-induced hyperalgesia.
Collapse
|
31
|
Interaction and regulatory functions of μ- and δ-opioid receptors in nociceptive afferent neurons. Neurosci Bull 2012; 28:121-30. [PMID: 22466123 DOI: 10.1007/s12264-012-1206-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
μ-opioid receptor (MOR) agonists such as morphine are powerful analgesics used for pain therapy. However, the use of these drugs is limited by their side-effects, which include antinociceptive tolerance and dependence. Earlier studies reported that MOR analgesic tolerance is reduced by blockade of δ-opioid receptors (DORs) that interact with MORs. Recent studies show that the MOR/DOR interaction in nociceptive afferent neurons in the dorsal root ganglion may contribute to morphine analgesic tolerance. Further analysis of the mechanisms for regulating the trafficking of receptors, ion channels and signaling molecules in nociceptive afferent neurons would help to understand the nociceptive mechanisms and improve pain therapy.
Collapse
|
32
|
Modulating pain in the periphery: gene-based therapies to enhance peripheral opioid analgesia: Bonica lecture, ASRA 2010. Reg Anesth Pain Med 2012; 37:210-4. [PMID: 22189620 DOI: 10.1097/aap.0b013e31823b145f] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
This article provides a brief overview of earlier work of our group on the peripheral signaling of pain, summarizes more recent studies on the role of opioids in chronic neuropathic pain, and speculates on the future of gene-based therapies as novel strategies to enhance the peripheral modulation of pain. Neurophysiologic and psychophysical studies have revealed features of primary afferent activity from somatic tissue that led to improved understanding of the physiology and pathophysiology of pain signaling by nociceptive and nonnociceptive fibers. The demonstration of peripheral opioid mechanisms in neuropathic pain suggests a potential role for these receptors in the modulation of pain at its initiation site. Our work has focused on characterizing this peripheral opioid analgesia in chronic neuropathic pain such that it can be exploited to develop novel and potent peripheral analgesics for its treatment. Ongoing research on virus-mediated gene transfer strategies to enhance peripheral opioid analgesia is presented.
Collapse
|
33
|
Interaction between Mu and Delta Opioid Receptor Agonists in an Assay of Capsaicin-Induced Thermal Allodynia in Rhesus Monkeys. PAIN RESEARCH AND TREATMENT 2012; 2012:867067. [PMID: 22666579 PMCID: PMC3361312 DOI: 10.1155/2012/867067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 02/28/2012] [Indexed: 01/25/2023]
Abstract
Delta opioid agonists enhance antinociceptive effects of mu-opioid agonists in many preclinical assays of acute nociception, but delta/mu interactions in preclinical models of inflammation-associated pain have not been examined. This study examined interactions between the delta agonist SNC80 [(+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide] and the mu agonist analgesics methadone, morphine, and nalbuphine in an assay of capsaicin-induced thermal allodynia in rhesus monkeys. Thermal allodynia was produced by topical application of capsaicin to the tail. Antiallodynic effects of methadone, morphine, and nalbuphine were evaluated alone or in combination with fixed proportions of SNC80 identical to proportions previously shown to enhance acute thermal antinociceptive effects of these mu agonists in rhesus monkeys (0.9 : 1 SNC80/methadone; 0.29 : 1 SNC80/morphine; 3.6 : 1 SNC80/nalbuphine). Methadone, morphine, and nalbuphine each produced dose-dependent antiallodynia. SNC80 produced partial antiallodynia up to the highest dose tested (5.6 mg/kg). SNC80 produced a modest, enantioselective, and naltrindole-reversible enhancement of methadone-induced antiallodynia. However, SNC80 did not enhance morphine antiallodynia and only weakly enhanced nalbuphine antiallodynia. Overall, SNC80 produced modest or no enhancement of the antiallodynic effects of the three mu agonists evaluated. These results suggest that delta agonist-induced enhancement of mu agonist antiallodynia may be weaker and less reliable than previously demonstrated enhancement of mu agonist acute thermal nociception.
Collapse
|
34
|
Ye Y, Dang D, Viet CT, Dolan JC, Schmidt BL. Analgesia targeting IB4-positive neurons in cancer-induced mechanical hypersensitivity. THE JOURNAL OF PAIN 2012; 13:524-31. [PMID: 22483679 DOI: 10.1016/j.jpain.2012.01.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 01/02/2012] [Accepted: 01/16/2012] [Indexed: 12/11/2022]
Abstract
UNLABELLED Cancer patients often suffer from pain and most will be prescribed μ-opioids. μ-opioids are not satisfactory in treating cancer pain and are associated with multiple debilitating side effects. Recent studies show that μ and δ opioid receptors are separately expressed on IB4 (-) and IB4 (+) neurons, which control thermal and mechanical pain, respectively. In this study we investigated IB4 (+) and IB4 (-) neurons in mechanical and thermal hypersensitivity in an orthotopic mouse oral cancer model. We used a δ opioid receptor agonist and a P2X(3) antagonist to target IB4 (+) neurons and to demonstrate that this subset plays a key role in cancer-induced mechanical allodynia, but not in thermal hyperalgesia. Moreover, selective removal of IB4 (+) neurons using IB4-saporin impacts cancer-induced mechanical but not thermal hypersensitivity. Our results demonstrate that peripherally administered pharmacological agents targeting IB4 (+) neurons, such as a selective δ-opioid receptor agonist or P2X(3) antagonist, might be useful in treating oral cancer pain. PERSPECTIVE To clarify the mechanisms of oral cancer pain, we examined the differential role of IB4 (+) and IB4 (-) neurons. Characterization of these 2 subsets of putative nociceptors is important for further development of effective clinical cancer pain relief.
Collapse
Affiliation(s)
- Yi Ye
- Bluestone Center for Clinic Research, New York University, New York, NY, USA
| | | | | | | | | |
Collapse
|
35
|
Inhibiting the breakdown of endogenous opioids and cannabinoids to alleviate pain. Nat Rev Drug Discov 2012; 11:292-310. [DOI: 10.1038/nrd3673] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Liu XJ, Zhang FX, Liu H, Li KC, Lu YJ, Wu QF, Li JY, Wang B, Wang Q, Lin LB, Zhong YQ, Xiao HS, Bao L, Zhang X. Activin C expressed in nociceptive afferent neurons is required for suppressing inflammatory pain. Brain 2012; 135:391-403. [DOI: 10.1093/brain/awr350] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Xing-Jun Liu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fang-Xiong Zhang
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Liu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kai-Cheng Li
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying-Jin Lu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qing-Feng Wu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jia-Yin Li
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Wang
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiong Wang
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li-Bo Lin
- 3 National Engineering Centre for Biochip at Shanghai, Shanghai 201203, China
| | - Yan-Qing Zhong
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua-Sheng Xiao
- 3 National Engineering Centre for Biochip at Shanghai, Shanghai 201203, China
| | - Lan Bao
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu Zhang
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
37
|
Delta opioid receptor analgesia: recent contributions from pharmacology and molecular approaches. Behav Pharmacol 2011; 22:405-14. [PMID: 21836459 DOI: 10.1097/fbp.0b013e32834a1f2c] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Delta opioid receptors represent a promising target for the development of novel analgesics. A number of tools have been developed recently that have significantly improved our knowledge of δ receptor function in pain control. These include several novel δ agonists with potent analgesic properties, and genetic mouse models with targeted mutations in the δ opioid receptor gene. Also, recent findings have further documented the regulation of δ receptor function at cellular level, which impacts on the pain-reducing activity of the receptor. These regulatory mechanisms occur at transcriptional and post-translational levels, along agonist-induced receptor activation, signaling and trafficking, or in interaction with other receptors and neuromodulatory systems. All these tools for in-vivo research, and proposed mechanisms at molecular level, have tremendously increased our understanding of δ receptor physiology, and contribute to designing innovative strategies for the treatment of chronic pain and other diseases such as mood disorders.
Collapse
|
38
|
Abstract
This paper is the thirty-third consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2010 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, USA.
| |
Collapse
|