1
|
Smith KM, Nguyen E, Ross SE. The Delta-Opioid Receptor Bidirectionally Modulates Itch. THE JOURNAL OF PAIN 2023; 24:264-272. [PMID: 36464136 PMCID: PMC10866011 DOI: 10.1016/j.jpain.2022.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 12/05/2022]
Abstract
Opioid signaling has been shown to be critically important in the neuromodulation of sensory circuits in the superficial spinal cord. Agonists of the mu-opioid receptor (MOR) elicit itch, whereas agonists of the kappa-opioid receptor (KOR) have been shown to inhibit itch. Despite the clear roles of MOR and KOR for the modulation itch, whether the delta-opioid receptor (DOR) is involved in the regulation of itch remained unknown. Here, we show that intrathecal administration of DOR agonists suppresses chemical itch and that intrathecal application of DOR antagonists is sufficient to evoke itch. We identify that spinal enkephalin neurons co-express neuropeptide Y (NPY), a peptide previously implicated in the inhibition of itch. In the spinal cord, DOR overlapped with both the NPY receptor (NPY1R) and KOR, suggesting that DOR neurons represent a site for convergent itch information in the dorsal horn. Lastly, we found that neurons co-expressing DOR and KOR showed significant Fos induction following pruritogen-evoked itch. These results uncover a role for DOR in the modulation of itch in the superficial dorsal horn. PERSPECTIVE: This article reveals the role of the delta-opioid receptor in itch. Intrathecal administration of delta agonists suppresses itch whereas the administration of delta antagonists is sufficient to induce itch. These studies highlight the importance of delta-opioid signaling for the modulation of itch behaviors, which may represent new targets for the management of itch disorders.
Collapse
Affiliation(s)
- Kelly M Smith
- University of Pittsburgh School of Medicine, Department of Neurobiology,Pittsburgh, Pennsylvania; University of Pittsburgh, Pittsburgh Center for Pain Research, Pittsburgh, Pennsylvania
| | - Eileen Nguyen
- University of Pittsburgh School of Medicine, Department of Neurobiology,Pittsburgh, Pennsylvania; University of Pittsburgh, Pittsburgh Center for Pain Research, Pittsburgh, Pennsylvania; University of Pittsburgh School of Medicine, Medical Scientist Training Program, Pittsburgh, Pennsylvania
| | - Sarah E Ross
- University of Pittsburgh School of Medicine, Department of Neurobiology,Pittsburgh, Pennsylvania; University of Pittsburgh, Pittsburgh Center for Pain Research, Pittsburgh, Pennsylvania.
| |
Collapse
|
2
|
Atigari DV, Paton KF, Uprety R, Váradi A, Alder AF, Scouller B, Miller JH, Majumdar S, Kivell BM. The mixed kappa and delta opioid receptor agonist, MP1104, attenuates chemotherapy-induced neuropathic pain. Neuropharmacology 2021; 185:108445. [PMID: 33383089 PMCID: PMC8344368 DOI: 10.1016/j.neuropharm.2020.108445] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/20/2020] [Accepted: 12/20/2020] [Indexed: 01/04/2023]
Abstract
Effective treatments for chronic pain without abuse liability are urgently needed. One in 5 adults suffer chronic pain and half of these patients report inefficient treatment. Mu opioid receptor agonists (MOP), including oxycodone, tramadol and morphine, are often prescribed to treat chronic pain, however, use of drugs targeting MOP can lead to drug dependency, tolerance and overdose deaths. Kappa opioid receptor (KOP) agonists have antinociceptive effects without abuse potential; however, they have not been utilised clinically due to dysphoria and sedation. We hypothesise that mixed opioid receptor agonists targeting the KOP and delta opioid receptor (DOP) would have a wider therapeutic index, with the rewarding effects of DOP negating the negative effects of KOP. MP1104, an analogue of 3-Iodobenzoyl naltrexamine, is a novel mixed opioid receptor agonist with potent antinociceptive effects mediated via KOP and DOP in mice without rewarding or aversive effects. In this study, we show MP1104 has potent, long-acting antinociceptive effects in the warm-water tail-withdrawal assay in male and female mice and rats; and is longer acting than morphine. In the paclitaxel-induced neuropathic pain model in mice, MP1104 reduced both mechanical and cold allodynia and unlike morphine, did not produce tolerance when administered daily for 23 days. Moreover, MP1104 did not induce sedative effects in the open-field locomotor activity test, respiratory depression in mice using whole-body plethysmography, or have cross-tolerance with morphine. This data supports the therapeutic development of mixed opioid receptor agonists, particularly mixed KOP/DOP agonists, as non-addictive pain medications with reduced tolerance.
Collapse
Affiliation(s)
- Diana Vivian Atigari
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Kelly Frances Paton
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Rajendra Uprety
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA
| | - András Váradi
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA
| | - Amy Frances Alder
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Brittany Scouller
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - John H Miller
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Susruta Majumdar
- Center of Clinical Pharmacology, St. Louis College of Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
| | - Bronwyn Maree Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|
3
|
Piotrowska A, Starnowska-Sokół J, Makuch W, Mika J, Witkowska E, Tymecka D, Ignaczak A, Wilenska B, Misicka A, Przewłocka B. Novel bifunctional hybrid compounds designed to enhance the effects of opioids and antagonize the pronociceptive effects of nonopioid peptides as potent analgesics in a rat model of neuropathic pain. Pain 2021; 162:432-445. [PMID: 32826750 PMCID: PMC7808367 DOI: 10.1097/j.pain.0000000000002045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/20/2020] [Accepted: 07/29/2020] [Indexed: 12/27/2022]
Abstract
ABSTRACT The purpose of our work was to determine the role of nonopioid peptides derived from opioid prohormones in sensory hypersensitivity characteristics of neuropathic pain and to propose a pharmacological approach to restore the balance of these endogenous opioid systems. Nonopioid peptides may have a pronociceptive effect and therefore contribute to less effective opioid analgesia in neuropathic pain. In our study, we used unilateral chronic constriction injury (CCI) of the sciatic nerve as a neuropathic pain model in rats. We demonstrated the pronociceptive effects of proopiomelanocortin- and proenkephalin-derived nonopioid peptides assessed by von Frey and cold plate tests, 7 to 14 days after injury. The concentration of proenkephalin-derived pronociceptive peptides was increased more robustly than that of Met-enkephalin in the ipsilateral lumbar spinal cord of CCI-exposed rats, as shown by mass spectrometry, and the pronociceptive effect of one of these peptides was blocked by an antagonist of the melanocortin 4 (MC4) receptor. The above results confirm our hypothesis regarding the possibility of creating an analgesic drug for neuropathic pain based on enhancing opioid activity and blocking the pronociceptive effect of nonopioid peptides. We designed and synthesized bifunctional hybrids composed of opioid (OP) receptor agonist and MC4 receptor antagonist (OP-linker-MC4). Moreover, we demonstrated that they have potent and long-lasting antinociceptive effects after a single administration and a delayed development of tolerance compared with morphine after repeated intrathecal administration to rats subjected to CCI. We conclude that the bifunctional hybrids OP-linker-MC4 we propose are important prototypes of drugs for use in neuropathic pain.
Collapse
Affiliation(s)
- Anna Piotrowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Joanna Starnowska-Sokół
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Wioletta Makuch
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Joanna Mika
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Ewa Witkowska
- Faculty of Chemistry, Biological, and Chemistry Research Centre, University of Warsaw, Warsaw, Poland
| | - Dagmara Tymecka
- Faculty of Chemistry, Biological, and Chemistry Research Centre, University of Warsaw, Warsaw, Poland
| | - Angelika Ignaczak
- Faculty of Chemistry, Biological, and Chemistry Research Centre, University of Warsaw, Warsaw, Poland
| | - Beata Wilenska
- Faculty of Chemistry, Biological, and Chemistry Research Centre, University of Warsaw, Warsaw, Poland
| | - Aleksandra Misicka
- Faculty of Chemistry, Biological, and Chemistry Research Centre, University of Warsaw, Warsaw, Poland
| | - Barbara Przewłocka
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| |
Collapse
|
4
|
Zhou W, Li Y, Meng X, Liu A, Mao Y, Zhu X, Meng Q, Jin Y, Zhang Z, Tao W. Switching of delta opioid receptor subtypes in central amygdala microcircuits is associated with anxiety states in pain. J Biol Chem 2021; 296:100277. [PMID: 33428940 PMCID: PMC7948800 DOI: 10.1016/j.jbc.2021.100277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 11/25/2022] Open
Abstract
Anxiety is often comorbid with pain. Delta opioid receptors (DORs) are promising targets for the treatment of pain and mental disorders with little addictive potential. However, their roles in anxiety symptoms at different stages of pain are unclear. In the current study, mice with inflammatory pain at the fourth hour following complete Freund’s adjuvant (CFA) injection displayed significant anxiety-like behavior, which disappeared at the seventh day. Combining electrophysiology, optogenetics, and pharmacology, we found that activation of delta opioid receptor 1 (DOR1) in the central nucleus amygdala (CeA) inhibited both the anxiolytic excitatory input from the basolateral amygdala (BLA) and the anxiogenic excitatory input from the parabrachial nucleus (PBN). In contrast, activation of delta opioid receptor 2 (DOR2) did not affect CeA excitatory synaptic transmission in normal and 4-h CFA mice but inhibited the excitatory projection from the PBN rather than the BLA in 7-day CFA mice. Furthermore, the function of both DOR1 and DOR2 was downregulated to the point of not being detectable in the CeA of mice at the 21st day following CFA injection. Taken together, these results suggest that functional switching of DOR1 and DOR2 is associated with anxiety states at different stages of pain via modulating the activity of specific pathways (BLA-CeA and PBN-CeA).
Collapse
Affiliation(s)
- Wenjie Zhou
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China
| | - Yanhua Li
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China
| | - Xiaojing Meng
- Department of Science and Education, Affiliated Psychological Hospital of Anhui Medical University, Hefei, China
| | - An Liu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yu Mao
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China; Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xia Zhu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China
| | - Qian Meng
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China; Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yan Jin
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China
| | - Zhi Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China.
| | - Wenjuan Tao
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China; Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
5
|
Zhao B, Li W, Sun L, Fu W. The Use of Computational Approaches in the Discovery and Mechanism Study of Opioid Analgesics. Front Chem 2020; 8:335. [PMID: 32500054 PMCID: PMC7242749 DOI: 10.3389/fchem.2020.00335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022] Open
Abstract
Opioid receptors that belong to class A G protein-coupled receptors (GPCRs) are vital in pain control. In the past few years, published high-resolution crystal structures of opioid receptor laid a solid basis for both experimental and computational studies. Computer-aided drug design (CADD) has been established as a powerful tool for discovering novel lead compounds and for understanding activation mechanism of target receptors. Herein, we reviewed the computational-guided studies on opioid receptors for the discovery of new analgesics, the structural basis of receptor subtype selectivity, agonist interaction mechanism, and biased signaling mechanism.
Collapse
Affiliation(s)
- Bangyi Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Lijie Sun
- Shijiazhuang No. 4 Pharmaceutical Co., Ltd., Shijiazhuang Economic and Technological Development Zone, Shijiazhuang, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Kremer M, Megat S, Bohren Y, Wurtz X, Nexon L, Ceredig RA, Doridot S, Massotte D, Salvat E, Yalcin I, Barrot M. Delta opioid receptors are essential to the antiallodynic action of Β 2-mimetics in a model of neuropathic pain. Mol Pain 2020; 16:1744806920912931. [PMID: 32208806 PMCID: PMC7097867 DOI: 10.1177/1744806920912931] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The adrenergic system, because of its reported implication in pain mechanisms, may be a potential target for chronic pain treatment. We previously demonstrated that β2-adrenoceptors (β2-ARs) are essential for neuropathic pain treatment by antidepressant drugs, and we showed that agonists of β2-ARs, that is, β2-mimetics, had an antiallodynic effect per se following chronic administration. To further explore the downstream mechanism of this action, we studied here the role of the opioid system. We used behavioral, genetic, and pharmacological approaches to test whether opioid receptors were necessary for the antiallodynic action of a short acting (terbutaline) and a long-acting (formoterol) β2-mimetic. Using the Cuff model of neuropathic pain in mice, we showed that chronic treatments with terbutaline (intraperitoneal) or formoterol (orally) alleviated mechanical hypersensitivity. We observed that these β2-mimetics remained fully effective in μ-opioid and in κ-opioid receptor deficient mice, but lost their antiallodynic action in δ-opioid receptor deficient mice, either female or male. Accordingly, we showed that the δ-opioid receptor antagonist naltrindole induced an acute relapse of allodynia in mice with neuropathic pain chronically treated with the β2-mimetics. Such relapse was also observed following administration of the peripheral opioid receptor antagonist naloxone methiodide. These data demonstrate that the antiallodynic effect of long-term β2-mimetics in a context of neuropathic pain requires the endogenous opioid system, and more specifically peripheral δ-opioid receptors.
Collapse
Affiliation(s)
- Mélanie Kremer
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Salim Megat
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Yohann Bohren
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Xavier Wurtz
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Laurent Nexon
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Rhian Alice Ceredig
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Stéphane Doridot
- Centre National de la Recherche Scientifique, Université de Strasbourg, Chronobiotron, Strasbourg, France
| | - Dominique Massotte
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Eric Salvat
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France.,Hôpitaux Universitaires de Strasbourg, Centre d'Evaluation et de Traitement de la Douleur, Strasbourg, France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Michel Barrot
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| |
Collapse
|
7
|
Berthiaume S, Abdallah K, Blais V, Gendron L. Alleviating pain with delta opioid receptor agonists: evidence from experimental models. J Neural Transm (Vienna) 2020; 127:661-672. [PMID: 32189076 DOI: 10.1007/s00702-020-02172-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
The use of opioids for the relief of pain and headache disorders has been studied for years. Nowadays, particularly because of its ability to produce analgesia in various pain models, delta opioid receptor (DOPr) emerges as a promising target for the development of new pain therapies. Indeed, their potential to avoid the unwanted effects commonly observed with clinically used opioids acting at the mu opioid receptor (MOPr) suggests that DOPr agonists could be a therapeutic option. In this review, we discuss the use of opioids in the management of pain in addition to describing the evidence of the analgesic potency of DOPr agonists in animal models.
Collapse
Affiliation(s)
- Sophie Berthiaume
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
| | - Khaled Abdallah
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
| | - Véronique Blais
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
| | - Louis Gendron
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada.
| |
Collapse
|
8
|
Kissiwaa SA, Patel SD, Winters BL, Bagley EE. Opioids differentially modulate two synapses important for pain processing in the amygdala. Br J Pharmacol 2020; 177:420-431. [PMID: 31596498 PMCID: PMC6989950 DOI: 10.1111/bph.14877] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/04/2019] [Accepted: 08/09/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Pain is a subjective experience involving sensory discriminative and emotionally aversive components. Consistent with its role in pain processing and emotions, the amygdala modulates the aversive component of pain. The laterocapsular region of the central nucleus of the amygdala (CeLC) receives nociceptive information from the parabrachial nucleus (PB) and polymodal, including nociceptive, inputs from the basolateral nucleus of the amygdala (BLA). Opioids are strong analgesics and reduce both the sensory discriminative and the affective component of pain. However, it is unknown whether opioids regulate activity at the two nociceptive inputs to the amygdala. EXPERIMENTAL APPROACH Using whole-cell electrophysiology, optogenetics, and immunohistochemistry, we investigated whether opioids inhibit the rat PB-CeLC and BLA-CeLC synapses. KEY RESULTS Opioids inhibited glutamate release at the PB-CeLC and BLA-CeLC synapses. Opioid inhibition is via the μ-receptor at the PB-CeLC synapse, while at the BLA-CeLC synapse, inhibition is via μ-receptors in all neurons and via δ-receptors and κ-receptors in a subset of neurons. CONCLUSIONS AND IMPLICATIONS Agonists of μ-receptors inhibited two of the synaptic inputs carrying nociceptive information into the laterocapsular amygdala. Therefore, μ-receptor agonists, such as morphine, will inhibit glutamate release from PB and BLA in the CeLC, and this could serve as a mechanism through which opioids reduce the affective component of pain and pain-induced associative learning. The lower than expected regulation of BLA synaptic outputs by δ-receptors does not support the proposal that opioid receptor subtypes segregate into subnuclei of brain regions.
Collapse
MESH Headings
- Amygdala/drug effects
- Amygdala/metabolism
- Amygdala/physiopathology
- Analgesics, Opioid/pharmacology
- Animals
- Glutamic Acid/metabolism
- Male
- Neural Inhibition/drug effects
- Nociception/drug effects
- Nociceptive Pain/metabolism
- Nociceptive Pain/physiopathology
- Nociceptive Pain/prevention & control
- Optogenetics
- Pain Perception/drug effects
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Synapses/drug effects
- Synapses/metabolism
Collapse
Affiliation(s)
- Sarah A. Kissiwaa
- Discipline of Pharmacology and Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| | - Sahil D. Patel
- Discipline of Pharmacology and Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| | - Bryony L. Winters
- Pain Management Research Institute, Kolling Institute of Medical ResearchThe University of Sydney, Royal North Shore HospitalSt LeonardsNSWAustralia
| | - Elena E. Bagley
- Discipline of Pharmacology and Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| |
Collapse
|
9
|
Vicario N, Pasquinucci L, Spitale FM, Chiechio S, Turnaturi R, Caraci F, Tibullo D, Avola R, Gulino R, Parenti R, Parenti C. Simultaneous Activation of Mu and Delta Opioid Receptors Reduces Allodynia and Astrocytic Connexin 43 in an Animal Model of Neuropathic Pain. Mol Neurobiol 2019; 56:7338-7354. [PMID: 31030416 DOI: 10.1007/s12035-019-1607-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/12/2019] [Indexed: 12/27/2022]
Abstract
Neuropathic pain is a chronic condition triggered by lesions to the somatosensory nervous system in which pain stimuli occur spontaneously or as pathologically amplified responses. In this scenario, the exchange of signaling molecules throughout cell-to-cell and cell-to-extracellular environment communications plays a key role in the transition from acute to chronic pain. As such, connexin 43 (Cx43), the core glial gap junction and hemichannel-forming protein, is considered a triggering factor for disease chronicization in the central nervous system (CNS). Drugs targeting μ opioid receptors (MOR) are currently used for moderate to severe pain conditions, but their use in chronic pain is limited by the tolerability profile. δ opioid receptors (DOR) have become attractive targets for the treatment of persistent pain and have been associated with the inhibition of pain-sustaining factors. Moreover, it has been shown that simultaneous targeting of MOR and DOR leads to an improved pharmacological fingerprint. Herein, we aimed to study the effects of the benzomorphan ligand LP2, a multitarget MOR/DOR agonist, in an experimental model of neuropathic pain induced by the unilateral sciatic nerve chronic constriction injury (CCI) on male Sprague-Dawley rats. Results showed that LP2 significantly ameliorated mechanical allodynia from the early phase of treatment up to 21 days post-ligatures. We additionally showed that LP2 prevented CCI-induced Cx43 alterations and pro-apoptotic signaling in the CNS. These findings increase the knowledge of neuropathic pain development and the role of spinal astrocytic Cx43, suggesting new approaches for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123, Catania, Italy
| | - Lorella Pasquinucci
- Department of Drug Sciences, Section of Medicinal Chemistry, University of Catania, 95125, Catania, Italy
| | - Federica M Spitale
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123, Catania, Italy
| | - Santina Chiechio
- Department of Drug Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125, Catania, Italy.,Oasi Research Institute-IRCCS, 94018, Troina, Italy
| | - Rita Turnaturi
- Department of Drug Sciences, Section of Medicinal Chemistry, University of Catania, 95125, Catania, Italy
| | - Filippo Caraci
- Department of Drug Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125, Catania, Italy.,Oasi Research Institute-IRCCS, 94018, Troina, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123, Catania, Italy
| | - Roberto Avola
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123, Catania, Italy
| | - Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123, Catania, Italy.
| | - Carmela Parenti
- Department of Drug Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125, Catania, Italy
| |
Collapse
|
10
|
Beaudeau JL, Blais V, Holleran BJ, Bergeron A, Piñeyro G, Guérin B, Gendron L, Dory YL. N-Guanidyl and C-Tetrazole Leu-Enkephalin Derivatives: Efficient Mu and Delta Opioid Receptor Agonists with Improved Pharmacological Properties. ACS Chem Neurosci 2019; 10:1615-1626. [PMID: 30614675 DOI: 10.1021/acschemneuro.8b00550] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Leu-enkephalin and d-Ala2-Leu-enkephalin were modified at their N- and C-termini with guanidyl and tetrazole groups. The resulting molecules were prepared in solution or by solid phase peptide synthesis. The affinity of the different analogues at mu (MOP) and delta opioid receptors (DOP) was then assessed by competitive binding in stably transfected DOP and MOP HEK293 cells. Inhibition of cAMP production and recruitment of β-arrestin were also investigated. Finally, lipophilicity (logD7.4) and plasma stability of each compound were measured. Compared to the native ligands, we found that the replacement of the terminal carboxylate by a tetrazole slightly decreased both the affinity at mu and delta opioid receptors as well as the half-life. By contrast, replacing the ammonium at the N-terminus with a guanidyl significantly improved the affinity, the potency, as well as the lipophilicity and the stability of the resulting peptides. Replacing the glycine residue with a d-alanine in position 2 consistently improved the potency as well as the stability of the analogues. The best peptidomimetic of the whole series, guanidyl-Tyr-d-Ala-Gly-Phe-Leu-tetrazole, displayed sub-nanomolar affinity and an increased lipophilicity. Moreover, it proved to be stable in plasma for up to 24 h, suggesting that the modifications are protecting the compound against protease degradation.
Collapse
Affiliation(s)
| | | | | | | | - Graciela Piñeyro
- Département de Psychiatrie, Centre de Recherche du CHU Ste-Justine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | | | | | | |
Collapse
|
11
|
Wu B, Liu Y, Liu F, Deng Q, Wang J, Han R, Zhang D, Chen J, Wei J. The antinociceptive effects and molecular mechanisms of ghrelin(1–7)-NH2 at the supraspinal level in acute pain in mice. Brain Res Bull 2019; 146:112-123. [DOI: 10.1016/j.brainresbull.2018.12.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/07/2018] [Accepted: 12/26/2018] [Indexed: 01/01/2023]
|
12
|
Rojewska E, Wawrzczak-Bargiela A, Szucs E, Benyhe S, Starnowska J, Mika J, Przewlocki R, Przewlocka B. Alterations in the Activity of Spinal and Thalamic Opioid Systems in a Mice Neuropathic Pain Model. Neuroscience 2018; 390:293-302. [DOI: 10.1016/j.neuroscience.2018.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 01/29/2023]
|
13
|
Van Elstraete A, Sitbon P, Hamdi L, Juarez-Perez V, Mazoit JX, Benhamou D, Rougeot C. The Opiorphin Analog STR-324 Decreases Sensory Hypersensitivity in a Rat Model of Neuropathic Pain. Anesth Analg 2018; 126:2102-2111. [DOI: 10.1213/ane.0000000000002413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
14
|
Abstract
Nowadays, the delta opioid receptor (DOPr) represents a promising target for the treatment of chronic pain and emotional disorders. Despite the fact that they produce limited antinociceptive effects in healthy animals and in most acute pain models, DOPr agonists have shown efficacy in various chronic pain models. In this chapter, we review the progresses that have been made over the last decades in understanding the role played by DOPr in the control of pain. More specifically, the distribution of DOPr within the central nervous system and along pain pathways is presented. We also summarize the literature supporting a role for DOPr in acute, tonic, and chronic pain models, as well as the mechanisms regulating its activity under specific conditions. Finally, novel compounds that have make their way to clinical trials are discussed.
Collapse
Affiliation(s)
- Khaled Abdallah
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada
- Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de recherche du CHUS, Sherbrooke, QC, Canada
| | - Louis Gendron
- Département de pharmacologie-physiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Centre de recherche du CHUS, Sherbrooke, QC, Canada.
- Département d'anesthésiologie, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Quebec Pain Research Network, Sherbrooke, QC, Canada.
| |
Collapse
|
15
|
Karad SN, Pal M, Crowley RS, Prisinzano TE, Altman RA. Synthesis and Opioid Activity of Tyr 1 -ψ[(Z)CF=CH]-Gly 2 and Tyr 1 -ψ[(S)/(R)-CF 3 CH-NH]-Gly 2 Leu-enkephalin Fluorinated Peptidomimetics. ChemMedChem 2017; 12:571-576. [PMID: 28296145 PMCID: PMC5486982 DOI: 10.1002/cmdc.201700103] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/13/2017] [Indexed: 12/16/2022]
Abstract
We describe the design, synthesis, and opioid activity of fluoroalkene (Tyr1 -ψ[(Z)CF=CH]-Gly2 ) and trifluoroethylamine (Tyr1 -ψ[(S)/(R)-CF3 CH-NH]-Gly2 ) analogues of the endogenous opioid neuropeptide, Leu-enkephalin. The fluoroalkene peptidomimetic exhibited low nanomolar functional activity (5.0±2 nm and 60±15 nm for δ- and μ-opioid receptors, respectively) with a μ/δ-selectivity ratio that mimics that of the natural peptide. However, the trifluoroethylamine peptidomimetics, irrespective of stereochemistry, did not activate the opioid receptors, which suggest that bulky CF3 substituents are not tolerated at this position.
Collapse
Affiliation(s)
- Somnath Narayan Karad
- Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas, 66045, USA
| | - Mohan Pal
- Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas, 66045, USA
| | - Rachel S Crowley
- Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas, 66045, USA
| | - Thomas E Prisinzano
- Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas, 66045, USA
| | - Ryan A Altman
- Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas, 66045, USA
| |
Collapse
|
16
|
Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 2017; 68:631-700. [PMID: 27343248 DOI: 10.1124/pr.114.008979] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Opioids are among the most effective analgesics available and are the first choice in the treatment of acute severe pain. However, partial efficacy, a tendency to produce tolerance, and a host of ill-tolerated side effects make clinically available opioids less effective in the management of chronic pain syndromes. Given that most therapeutic opioids produce their actions via µ-opioid receptors (MOPrs), other targets are constantly being explored, among which δ-opioid receptors (DOPrs) are being increasingly considered as promising alternatives. This review addresses DOPrs from the perspective of cellular and molecular determinants of their pharmacological diversity. Thus, DOPr ligands are examined in terms of structural and functional variety, DOPrs' capacity to engage a multiplicity of canonical and noncanonical G protein-dependent responses is surveyed, and evidence supporting ligand-specific signaling and regulation is analyzed. Pharmacological DOPr subtypes are examined in light of the ability of DOPr to organize into multimeric arrays and to adopt multiple active conformations as well as differences in ligand kinetics. Current knowledge on DOPr targeting to the membrane is examined as a means of understanding how these receptors are especially active in chronic pain management. Insight into cellular and molecular mechanisms of pharmacological diversity should guide the rational design of more effective, longer-lasting, and better-tolerated opioid analgesics for chronic pain management.
Collapse
Affiliation(s)
- Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Catherine M Cahill
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Mark von Zastrow
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Peter W Schiller
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Graciela Pineyro
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| |
Collapse
|
17
|
Pirisedigh A, Blais V, Ait-Mohand S, Abdallah K, Holleran BJ, Leduc R, Dory YL, Gendron L, Guérin B. Synthesis and Evaluation of a 64Cu-Conjugate, a Selective δ-Opioid Receptor Positron Emission Tomography Imaging Agent. Org Lett 2017; 19:2018-2021. [DOI: 10.1021/acs.orglett.7b00575] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Azadeh Pirisedigh
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Véronique Blais
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Samia Ait-Mohand
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Khaled Abdallah
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Brian J. Holleran
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Richard Leduc
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Yves L. Dory
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Louis Gendron
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Brigitte Guérin
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| |
Collapse
|
18
|
Zeng L, Alongkronrusmee D, van Rijn RM. An integrated perspective on diabetic, alcoholic, and drug-induced neuropathy, etiology, and treatment in the US. J Pain Res 2017; 10:219-228. [PMID: 28176937 PMCID: PMC5268333 DOI: 10.2147/jpr.s125987] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neuropathic pain (NeuP) is a syndrome that results from damaged nerves and/or aberrant regeneration. Common etiologies of neuropathy include chronic illnesses and medication use. Chronic disorders, such as diabetes and alcoholism, can cause neuronal injury and consequently NeuP. Certain medications with antineoplastic effects also carry an exquisitely high risk for neuropathy. These culprits are a few of many that are fueling the NeuP epidemic, which currently affects 7%-10% of the population. It has been estimated that approximately 10% and 7% of US adults carry a diagnosis of diabetes and alcohol disorder, respectively. Despite its pervasiveness, many physicians are unfamiliar with adequate treatment of NeuP, partly due to the few reviews that are available that have integrated basic science and clinical practice. In light of the recent Centers for Disease Control and Prevention guidelines that advise against the routine use of μ-opioid receptor-selective opioids for chronic pain management, such a review is timely. Here, we provide a succinct overview of the etiology and treatment options of diabetic and alcohol- and drug-induced neuropathy, three different and prevalent neuropathies fusing the combined clinical and preclinical pharmacological expertise in NeuP of the authors. We discuss the anatomy of pain and pain transmission, with special attention to key ion channels, receptors, and neurotransmitters. An understanding of pain neurophysiology will lead to a better understanding of the rationale for the effectiveness of current treatment options, and may lead to better diagnostic tools to help distinguish types of neuropathy. We close with a discussion of ongoing research efforts to develop additional treatments for NeuP.
Collapse
Affiliation(s)
- Lily Zeng
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Doungkamol Alongkronrusmee
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
19
|
Liu FY, Zhang MM, Zeng P, Liu WW, Wang JL, Yang B, Dai Q, Wei J. Study on the molecular mechanism of antinociception induced by ghrelin in acute pain in mice. Peptides 2016; 83:1-7. [PMID: 27474249 DOI: 10.1016/j.peptides.2016.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 01/04/2023]
Abstract
Ghrelin has been identified as the endogenous ligand for the GHS-R1α (growth hormone secretagogue receptor 1 alpha). Our previous experiments have indicated that ghrelin (i.c.v.) induces antinociceptive effects in acute pain in mice, and the effects were mediated through the central opioid receptors and GHS-R1α. However, which opioid receptor (OR) mediates the antinociceptive effects and the molecular mechanisms are also needed to be further explored. In the present study, the antinociceptive effects of ghrelin (i.c.v.) could be fully antagonized by δ-opioid receptor antagonist NTI. Furthermore, the mRNA and protein levels of δ-opioid peptide PENK and δ-opioid receptor OPRD were increased after i.c.v injection of ghrelin. Thus, it showed that the antinociception of ghrelin was correlated with the GHS-R1α and δ-opioid receptors. To explore which receptor was firstly activated by ghrelin, GHS-R1α antagonist [D-Lys(3)]-GHRP-6 was co-injection (i.c.v.) with deltorphin II (selective δ-opioid receptor agonist). Finally, the antinociception induced by deltorphin II wasn't blocked by the co-injection (i.c.v.) of [D-Lys(3)]-GHRP-6, indicating that the GHS-R1α isn't on the backward position of δ-opioid receptor. The results suggested that i.c.v. injection of ghrelin initially activated the GHS-R1α, which in turn increased the release of endogenous PENK to activation of OPRD to produce antinociception.
Collapse
Affiliation(s)
- Fu-Yan Liu
- Department of Physiology, Medical College of Nanchang University, Bayi Road 461, Nanchang, Jiangxi, 330006, China
| | - Min-Min Zhang
- Department of Physiology, Medical College of Nanchang University, Bayi Road 461, Nanchang, Jiangxi, 330006, China
| | - Ping Zeng
- Department of Physiology, Medical College of Nanchang University, Bayi Road 461, Nanchang, Jiangxi, 330006, China
| | - Wen-Wen Liu
- Department of Physiology, Medical College of Nanchang University, Bayi Road 461, Nanchang, Jiangxi, 330006, China
| | - Jing-Lei Wang
- Department of Physiology, Medical College of Nanchang University, Bayi Road 461, Nanchang, Jiangxi, 330006, China
| | - Bei Yang
- Department of Physiology, Medical College of Nanchang University, Bayi Road 461, Nanchang, Jiangxi, 330006, China
| | - Qun Dai
- Medical Experimental Teaching Department, Nanchang University, Nanchang 330031, China
| | - Jie Wei
- Department of Physiology, Medical College of Nanchang University, Bayi Road 461, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
20
|
Shen Q, Qian Y, Huang X, Xu X, Li W, Liu J, Fu W. Discovery of Potent and Selective Agonists of δ Opioid Receptor by Revisiting the "Message-Address" Concept. ACS Med Chem Lett 2016; 7:391-6. [PMID: 27096047 DOI: 10.1021/acsmedchemlett.5b00423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/10/2016] [Indexed: 01/10/2023] Open
Abstract
The classic "message-address" concept was proposed to address the binding of endogenous peptides to the opioid receptors and was later successfully applied in the discovery of the first nonpeptide δ opioid receptor (DOR) antagonist naltrindole. By revisiting this concept, and based on the structure of tramadol, we designed a series of novel compounds that act as highly potent and selective agonists of DOR among which (-)-6j showed the highest affinity (K i = 2.7 nM), best agonistic activity (EC50 = 2.6 nM), and DOR selectivity (more than 1000-fold over the other two subtype opioid receptors). Molecular docking studies suggest that the "message" part of (-)-6j interacts with residue Asp128(3.32) and a neighboring water molecule, and the "address" part of (-)-6j packs with hydrophobic residues Leu300(7.35), Val281(6.55), and Trp284(6.58), rendering DOR selectivity. The discovery of novel compound (-)-6j, and the obtained insights into DOR-agonist binding will help us design more potent and selective DOR agonists.
Collapse
Affiliation(s)
- Qing Shen
- Department
of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Yuanyuan Qian
- Department
of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Xiaoqin Huang
- Center
for Theoretical Biological Physics and Center for Research Computing, Rice University, Houston, Texas 77005, United States
| | - Xuejun Xu
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Wei Li
- Department
of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Jinggen Liu
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Wei Fu
- Department
of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
21
|
Zamponi GW, Han C, Waxman SG. Voltage-Gated Ion Channels as Molecular Targets for Pain. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
22
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 728] [Impact Index Per Article: 80.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
23
|
Mika J, Popiolek-Barczyk K, Rojewska E, Makuch W, Starowicz K, Przewlocka B. Delta-opioid receptor analgesia is independent of microglial activation in a rat model of neuropathic pain. PLoS One 2014; 9:e104420. [PMID: 25105291 PMCID: PMC4126741 DOI: 10.1371/journal.pone.0104420] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/09/2014] [Indexed: 12/15/2022] Open
Abstract
The analgesic effect of delta-opioid receptor (DOR) ligands in neuropathic pain is not diminished in contrast to other opioid receptor ligands, which lose their effectiveness as analgesics. In this study, we examine whether this effect is related to nerve injury-induced microglial activation. We therefore investigated the influence of minocycline-induced inhibition of microglial activation on the analgesic effects of opioid receptor agonists: morphine, DAMGO, U50,488H, DPDPE, Deltorphin II and SNC80 after chronic constriction injury (CCI) to the sciatic nerve in rats. Pre-emptive and repeated administration of minocycline (30 mg/kg, i.p.) over 7 days significantly reduced allodynia and hyperalgesia as measured on day 7 after CCI. The antiallodynic and antihyperalgesic effects of intrathecally (i.t.) administered morphine (10–20 µg), DAMGO (1–2 µg) and U50,488H (25–50 µg) were significantly potentiated in rats after minocycline, but no such changes were observed after DPDPE (10–20 µg), deltorphin II (1.5–15 µg) and SNC80 (10–20 µg) administration. Additionally, nerve injury-induced down-regulation of all types of opioid receptors in the spinal cord and dorsal root ganglia was not influenced by minocycline, which indicates that the effects of opioid ligands are dependent on other changes, presumably neuroimmune interactions. Our study of rat primary microglial cell culture using qRT-PCR, Western blotting and immunocytochemistry confirmed the presence of mu-opioid receptors (MOR) and kappa-opioid receptors (KOR), further we provide the first evidence for the lack of DOR on microglial cells. In summary, DOR analgesia is different from analgesia induced by MOR and KOR receptors because it does not dependent on injury-induced microglial activation. DOR agonists appear to be the best candidates for new drugs to treat neuropathic pain.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/therapeutic use
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/therapeutic use
- Animals
- Anti-Bacterial Agents/administration & dosage
- Anti-Bacterial Agents/therapeutic use
- Cells, Cultured
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use
- Gene Expression Regulation/drug effects
- Male
- Microglia/cytology
- Microglia/drug effects
- Microglia/metabolism
- Minocycline/administration & dosage
- Minocycline/therapeutic use
- Morphine/administration & dosage
- Morphine/therapeutic use
- Neuralgia/drug therapy
- Rats, Wistar
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
Collapse
Affiliation(s)
- Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
- * E-mail: (BP); (JM)
| | | | - Ewelina Rojewska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Starowicz
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Barbara Przewlocka
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
- * E-mail: (BP); (JM)
| |
Collapse
|
24
|
Chabot-Doré AJ, Schuster DJ, Stone LS, Wilcox GL. Analgesic synergy between opioid and α2 -adrenoceptors. Br J Pharmacol 2014; 172:388-402. [PMID: 24641506 DOI: 10.1111/bph.12695] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 03/08/2014] [Accepted: 03/12/2014] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Opioid and α2 -adrenoceptor agonists are potent analgesic drugs and their analgesic effects can synergize when co-administered. These supra-additive interactions are potentially beneficial clinically; by increasing efficacy and/or reducing the total drug required to produce sufficient pain relief, undesired side effects can be minimized. However, combination therapies of opioids and α2 -adrenoceptor agonists remain underutilized clinically, in spite of a large body of preclinical evidence describing their synergistic interaction. One possible obstacle to the translation of preclinical findings to clinical applications is a lack of understanding of the mechanisms underlying the synergistic interactions between these two drug classes. In this review, we provide a detailed overview of the interactions between different opioid and α2 -adrenoceptor agonist combinations in preclinical studies. These studies have identified the spinal cord as an important site of action of synergistic interactions, provided insights into which receptors mediate these interactions and explored downstream signalling events enabling synergy. It is now well documented that the activation of both μ and δ opioid receptors can produce synergy with α2 -adrenoceptor agonists and that α2 -adrenoceptor agonists can mediate synergy through either the α2A or the α2C adrenoceptor subtypes. Current hypotheses surrounding the cellular mechanisms mediating opioid-adrenoceptor synergy, including PKC signalling and receptor oligomerization, and the evidence supporting them are presented. Finally, the implications of these findings for clinical applications and drug discovery are discussed. 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)
- A-J Chabot-Doré
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | | | | |
Collapse
|
25
|
Antkowiak B, Paluch M, Ciechanowska M, Nawrocka M, Bańkowski K, Michalak O, Kocik J, Kowalczyk M, Izdebski J. Antinociceptive effect of D-Lys(2), Dab(4)N-(ureidoethyl)amide, a new cyclic 1-4 dermorphin/deltorphin analog. Pharmacol Rep 2014; 66:600-5. [PMID: 24948060 DOI: 10.1016/j.pharep.2014.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 11/22/2013] [Accepted: 01/30/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND A preliminary evaluation of antinociceptive activity of a new cyclic dermorphin/deltorphin tetrapeptide analog restricted via a urea bridge and containing C-terminal ureidoethylamid {[H-Tyr-d-Lys(&(1))-Phe-Dab(&(2))-CH2CH2NHCONH2][&(1)CO&(2)]} (cUP-1) revealed a significant and long-lasting increase of pain threshold to thermal stimulation after systemic application. The current studies were aimed at further evaluation of cUP-1 activity in animal models of somatic and visceral pain. The influence of cUP-1 on motor functions was also investigated. METHODS The influence of cUP-1 (0.5-2mgkg(-1), iv) on nociceptive threshold to mechanical pressure and analgesic efficacy in formalin and acetic acid-induced writhing tests were estimated. The antinociceptive effect of cUP-1 was compared to that of morphine (MF). The influence of cUP-1 (1, 4 and 8mgkg(-1), iv) on locomotor activity, motor coordination and muscle strength was estimated using open field and rota-rod tests and a grip strength measurement. RESULTS Administration of cUP-1 in doses of 1 and 2mgkg(-1) elicited a significant increase of nociceptive threshold to mechanical pressure. MF applied in the same doses induced an antinociceptive effect only at the higher dose (2mgkg(-1)). There were no marked differences between the effect of cUP-1 and MF at each dose, at relative time points. In the writhing test and both phases of the formalin test, cUP-1 showed a significant, dose-dependent antinociceptive effect which did not markedly differ from that of MF. cUP-1 did not significantly affect motor functions of mice. CONCLUSIONS Systemic application of cUP-1 elicited a dose-dependent antinociceptive effect. The analgesic efficacy of cUP-1 on mechanical nociception, visceral and formalin-induced pain was comparable to that of MF. cUP-1 did not impair motor functions of mice.
Collapse
Affiliation(s)
- Bożena Antkowiak
- Military Institute of Hygiene and Epidemiology, Department of Pharmacology and Toxicology, Warsaw, Poland.
| | - Małgorzata Paluch
- Military Institute of Hygiene and Epidemiology, Department of Pharmacology and Toxicology, Warsaw, Poland
| | - Magdalena Ciechanowska
- Military Institute of Hygiene and Epidemiology, Department of Pharmacology and Toxicology, Warsaw, Poland
| | - Małgorzata Nawrocka
- Military Institute of Hygiene and Epidemiology, Department of Pharmacology and Toxicology, Warsaw, Poland
| | | | | | - Janusz Kocik
- Military Institute of Hygiene and Epidemiology, Department of Pharmacology and Toxicology, Warsaw, Poland
| | - Marek Kowalczyk
- Military Institute of Hygiene and Epidemiology, Department of Pharmacology and Toxicology, Warsaw, Poland; Józef Piłsudski University of Physical Education, Warsaw, Poland
| | - Jan Izdebski
- Warsaw University, Department of Chemistry, Warsaw, Poland
| |
Collapse
|
26
|
Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW. Calcium-permeable ion channels in pain signaling. Physiol Rev 2014; 94:81-140. [PMID: 24382884 DOI: 10.1152/physrev.00023.2013] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The detection and processing of painful stimuli in afferent sensory neurons is critically dependent on a wide range of different types of voltage- and ligand-gated ion channels, including sodium, calcium, and TRP channels, to name a few. The functions of these channels include the detection of mechanical and chemical insults, the generation of action potentials and regulation of neuronal firing patterns, the initiation of neurotransmitter release at dorsal horn synapses, and the ensuing activation of spinal cord neurons that project to pain centers in the brain. Long-term changes in ion channel expression and function are thought to contribute to chronic pain states. Many of the channels involved in the afferent pain pathway are permeable to calcium ions, suggesting a role in cell signaling beyond the mere generation of electrical activity. In this article, we provide a broad overview of different calcium-permeable ion channels in the afferent pain pathway and their role in pain pathophysiology.
Collapse
|
27
|
Somvanshi RK, Kumar U. δ-opioid receptor and somatostatin receptor-4 heterodimerization: possible implications in modulation of pain associated signaling. PLoS One 2014; 9:e85193. [PMID: 24416361 PMCID: PMC3885706 DOI: 10.1371/journal.pone.0085193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 12/01/2013] [Indexed: 01/13/2023] Open
Abstract
Pain relief is the principal action of opioids. Somatostatin (SST), a growth hormone inhibitory peptide is also known to alleviate pain even in cases when opioids fail. Recent studies have shown that mice are prone to sustained pain and devoid of analgesic effect in the absence of somatostatin receptor 4 (SSTR4). In the present study, using brain slices, cultured neurons and HEK-293 cells, we showed that SSTR4 and δ-Opioid receptor (δOR) exist in a heteromeric complex and function in synergistic manner. SSTR4 and δOR co-expressed in cortical/striatal brain regions and spinal cord. Using cultured neuronal cells, we describe the heterogeneous complex formation of SSTR4 and δOR at neuronal cell body and processes. Cotransfected cells display inhibition of cAMP/PKA and co-activation of SSTR4 and δOR oppose receptor trafficking induced by individual receptor activation. Furthermore, downstream signaling pathways either associated with withdrawal or pain relief are modulated synergistically with a predominant role of SSTR4. Inhibition of cAMP/PKA and activation of ERK1/2 are the possible cellular adaptations to prevent withdrawal induced by chronic morphine use. Our results reveal direct intra-membrane interaction between SSTR4 and δOR and provide insights for the molecular mechanism for the anti-nociceptive property of SST in combination with opioids as a potential therapeutic approach to avoid undesirable withdrawal symptoms.
Collapse
Affiliation(s)
- Rishi K. Somvanshi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
| | - Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
28
|
Rochon K, Proteau-Gagné A, Bourassa P, Nadon JF, Côté J, Bournival V, Gobeil F, Guérin B, Dory YL, Gendron L. Preparation and evaluation at the delta opioid receptor of a series of linear leu-enkephalin analogues obtained by systematic replacement of the amides. ACS Chem Neurosci 2013; 4:1204-16. [PMID: 23650868 PMCID: PMC3750687 DOI: 10.1021/cn4000583] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/07/2013] [Indexed: 12/16/2022] Open
Abstract
Leu-enkephalin analogues, in which the amide bonds were sequentially and systematically replaced either by ester or N-methyl amide bonds, were prepared using classical organic chemistry as well as solid phase peptide synthesis (SPPS). The peptidomimetics were characterized using competition binding, ERK1/2 phosphorylation, receptor internalization, and contractility assays to evaluate their pharmacological profile over the delta opioid receptor (DOPr). The lipophilicity (LogD7.4) and plasma stability of the active analogues were also measured. Our results revealed that the last amide bond can be successfully replaced by either an ester or an N-methyl amide bond without significantly decreasing the biological activity of the corresponding analogues when compared to Leu-enkephalin. The peptidomimetics with an N-methyl amide function between residues Phe and Leu were found to be more lipophilic and more stable than Leu-enkephalin. Findings from the present study further revealed that the hydrogen-bond donor properties of the fourth amide of Leu-enkephalin are not important for its biological activity on DOPr. Our results show that the systematic replacement of amide bonds by isosteric functions represents an efficient way to design and synthesize novel peptide analogues with enhanced stability. Our findings further suggest that such a strategy can also be useful to study the biological roles of amide bonds.
Collapse
Affiliation(s)
- Kristina Rochon
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Arnaud Proteau-Gagné
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Philippe Bourassa
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Jean-François Nadon
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Jérome Côté
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Véronique Bournival
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Fernand Gobeil
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Brigitte Guérin
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Yves L. Dory
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Louis Gendron
- Département de Physiologie
et Biophysique, Laboratoire de Synthèse Supramoléculaire, Département
de Chimie, Département de pharmacologie, and Département de Médecine
Nucléaire et Radiobiologie, Institut de Pharmacologie, Université de Sherbrooke, 3001, 12 Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| |
Collapse
|
29
|
Systematic replacement of amides by 1,4-disubstituted[1,2,3]triazoles in Leu-enkephalin and the impact on the delta opioid receptor activity. Bioorg Med Chem Lett 2013; 23:5267-9. [PMID: 23988352 DOI: 10.1016/j.bmcl.2013.08.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 07/25/2013] [Accepted: 08/05/2013] [Indexed: 12/28/2022]
Abstract
Using Cu(I)-catalyzed azide-alkyne cycloaddition in a mixed classical organic phase and solid phase peptide synthesis approach, we synthesized four analogs of Leu-enkephalin to systematically replace amides by 1,4-disubstituted[1,2,3]triazoles. The peptidomimetics obtained were characterized by competitive binding, contractility assays and ERK1/2 phosphorylation. The present study reveals that the analog bearing a triazole between Phe and Leu retains some potency, more than all the others, suggesting that the hydrogen bond acceptor capacity of the last amide of Leu-enkephalin is essential for the biological activity of the peptide.
Collapse
|
30
|
Pharmacological traits of delta opioid receptors: pitfalls or opportunities? Psychopharmacology (Berl) 2013; 228:1-18. [PMID: 23649885 PMCID: PMC3679311 DOI: 10.1007/s00213-013-3129-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 04/15/2013] [Indexed: 12/11/2022]
Abstract
RATIONALE Delta opioid receptors (DORs) have been considered as a potential target to relieve pain as well as treat depression and anxiety disorders and are known to modulate other physiological responses, including ethanol and food consumption. A small number of DOR-selective drugs are in clinical trials, but no DOR-selective drugs have been approved by the Federal Drug Administration and some candidates have failed in phase II clinical trials, highlighting current difficulties producing effective delta opioid-based therapies. Recent studies have provided new insights into the pharmacology of the DOR, which is often complex and at times paradoxical. OBJECTIVE This review will discuss the existing literature focusing on four aspects: (1) Two DOR subtypes have been postulated based on differences in pharmacological effects of existing DOR-selective ligands. (2) DORs are expressed ubiquitously throughout the body and central nervous system and are, thus, positioned to play a role in a multitude of diseases. (3) DOR expression is often dynamic, with many reports of increased expression during exposure to chronic stimuli, such as stress, inflammation, neuropathy, morphine, or changes in endogenous opioid tone. (4) A large structural variety in DOR ligands implies potential different mechanisms of activating the receptor. CONCLUSION The reviewed features of DOR pharmacology illustrate the potential benefit of designing tailored or biased DOR ligands.
Collapse
|
31
|
Pettinger L, Gigout S, Linley JE, Gamper N. Bradykinin controls pool size of sensory neurons expressing functional δ-opioid receptors. J Neurosci 2013; 33:10762-71. [PMID: 23804098 PMCID: PMC3724994 DOI: 10.1523/jneurosci.0123-13.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 11/21/2022] Open
Abstract
Analgesics targeting the δ-opioid receptor (DOR) may lead to fewer side effects than conventional opioid drugs, which mainly act on μ-opioid receptors (MOR), because of the less abundant expression of DOR in the CNS compared with MOR. Analgesic potential of DOR agonists increases after inflammation, an effect that may be mediated by DOR expressed in the peripheral sensory fibers. However, the expression of functional DOR at the plasma membrane of sensory neurons is controversial. Here we have used patch-clamp recordings and total internal reflection fluorescence microscopy to study the functional expression of DOR in sensory neurons from rat trigeminal (TG) and dorsal root ganglia (DRG). Real-time total internal reflection fluorescence microscopy revealed that treatment of TG and DRG cultures with the inflammatory mediator bradykinin (BK) caused robust trafficking of heterologously expressed GFP-tagged DOR to the plasma membrane. By contrast, treatment of neurons with the DOR agonist [d-Ala(2), d-Leu(5)]-enkephalin (DADLE) caused a decrease in the membrane abundance of DOR, suggesting internalization of the receptor after agonist binding. Patch-clamp experiments revealed that DADLE inhibited voltage-gated Ca(2+) channels (VGCCs) in 23% of small-diameter TG neurons. Pretreatment with BK resulted in more than twice as many DADLE responsive neurons (54%) but did not affect the efficacy of VGCC inhibition by DADLE. Our data suggest that inflammatory mediator-induced membrane insertion of DOR into the plasma membrane of peripheral sensory neurons may underlie increased DOR analgesia in inflamed tissue. Furthermore, the majority of BK-responsive TG neurons may have a potential to become responsive to DOR ligands in inflammatory conditions.
Collapse
Affiliation(s)
- Louisa Pettinger
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Sylvain Gigout
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - John E. Linley
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Nikita Gamper
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| |
Collapse
|
32
|
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: 6.0] [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
|
33
|
Otis V, Sarret P, Gendron L. Spinal activation of delta opioid receptors alleviates cancer-related bone pain. Neuroscience 2011; 183:221-9. [PMID: 21458544 PMCID: PMC3848971 DOI: 10.1016/j.neuroscience.2011.03.052] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 03/21/2011] [Accepted: 03/24/2011] [Indexed: 11/28/2022]
Abstract
Over the past few years, significant progress has been made in cancer therapy. Indeed, the lifespan of cancer patients has significantly increased. Although patients live longer, cancer-related pain remains a daily problem affecting their quality of life, especially when metastases reach the bone. In patients coping with cancer-induced bone pain, morphine and NSAIDs, often used in combination with other medications, are the most commonly used drugs to alleviate pain. However, these drugs have dose-limiting side effects. Morphine and other routinely used opioids are mu opioid receptor (MOPR) agonists. The MOPR is responsible for most opioid-related adverse effects. In the present study, we revealed potent analgesic effects of an intrathecally-administered selective delta opioid receptor (DOPR) agonist, deltorphin II, in a recently developed rat bone cancer model. Indeed, we found that deltorphin II dose-dependently reversed mechanical allodynia 14 days post-surgery in this cancer pain model, which is based on the implantation of mammary MRMT-1 cells in the femur. This effect was DOPR-mediated as it was completely blocked by naltrindole, a selective DOPR antagonist. Using the complete Freund's adjuvant model of inflammatory pain, we further demonstrated that deltorphin II was equipotent at alleviating inflammatory and cancer pain (i.e. similar ED50 values). Altogether, the present results show, for the first time, that activation of spinal DOPRs causes significant analgesia at doses sufficient to reduce inflammatory pain in a rat bone cancer pain model. Our results further suggest that DOPR represents a potential target for the development of novel analgesic therapies to be used in the treatment of cancer-related pain.
Collapse
Affiliation(s)
- Valérie Otis
- Département de physiologie et biophysique, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Philippe Sarret
- Département de physiologie et biophysique, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institut de pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Louis Gendron
- Département de physiologie et biophysique, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institut de pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
34
|
Gaveriaux-Ruff C, Nozaki C, Nadal X, Hever XC, Weibel R, Matifas A, Reiss D, Filliol D, Nassar MA, Wood JN, Maldonado R, Kieffer BL. Genetic ablation of delta opioid receptors in nociceptive sensory neurons increases chronic pain and abolishes opioid analgesia. Pain 2011; 152:1238-1248. [PMID: 21295407 DOI: 10.1016/j.pain.2010.12.031] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 12/17/2010] [Accepted: 12/17/2010] [Indexed: 11/24/2022]
Abstract
Opioid receptors are major actors in pain control and are broadly distributed throughout the nervous system. A major challenge in pain research is the identification of key opioid receptor populations within nociceptive pathways, which control physiological and pathological pain. In particular, the respective contribution of peripheral vs. central receptors remains unclear, and it has not been addressed by genetic approaches. To investigate the contribution of peripheral delta opioid receptors in pain control, we created conditional knockout mice where delta receptors are deleted specifically in peripheral Na(V)1.8-positive primary nociceptive neurons. Mutant mice showed normal pain responses to acute heat and to mechanical and formalin stimuli. In contrast, mutant animals showed a remarkable increase of mechanical allodynia under both inflammatory pain induced by complete Freund adjuvant and neuropathic pain induced by partial sciatic nerve ligation. In these 2 models, heat hyperalgesia was virtually unchanged. SNC80, a delta agonist administered either systemically (complete Freund adjuvant and sciatic nerve ligation) or into a paw (sciatic nerve ligation), reduced thermal hyperalgesia and mechanical allodynia in control mice. However, these analgesic effects were absent in conditional mutant mice. In conclusion, this study reveals the existence of delta opioid receptor-mediated mechanisms, which operate at the level of Na(V)1.8-positive nociceptive neurons. Delta receptors in these neurons tonically inhibit mechanical hypersensitivity in both inflammatory and neuropathic pain, and they are essential to mediate delta opioid analgesia under conditions of persistent pain. This delta receptor population represents a feasible therapeutic target to alleviate chronic pain while avoiding adverse central effects. The conditional knockout of delta-opioid receptor in primary afferent Na(V)1.8 neurons augmented mechanical allodynia in persistent pain models and abolished delta opioid analgesia in these models.
Collapse
Affiliation(s)
- Claire Gaveriaux-Ruff
- IGBMC Institut de Génétique et de Biologie Moléculaire et Cellulaire, Neurobiology and Genetic Department, Illkirch F-67400, France INSERM, U964, Illkirch F-67400, France CNRS, UMR7104, Illkirch F-67400, France UdS Université de Strasbourg, Strasbourg F-67000, France Laboratori de Neurofarmacologica, Facultat de Ciencies de la Salut i de la vida, Universitat Pompeu Fabra, Parc de Recerca Biomedica de Barcelona, 08003 Barcelona, Spain Molecular Nociception, Wolfson Institute for Biomedical research, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Proteau-Gagné A, Bournival V, Rochon K, Dory YL, Gendron L. Exploring the Backbone of Enkephalins To Adjust Their Pharmacological Profile for the δ-Opioid Receptor. ACS Chem Neurosci 2010; 1:757-69. [PMID: 22778812 DOI: 10.1021/cn1000759] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 09/08/2010] [Indexed: 12/23/2022] Open
Abstract
The role of each of the four amide bonds in Leu(5)-enkephalin was investigated by systematically and sequentially replacing each with its corresponding trans-alkene. Six Leu(5)-enkephalin analogs based on six dipeptide surrogates and two Met(5)-enkephalin analogs were synthesized and thoroughly tested using a δ-opioid receptor internalization assay, an ERK1/2 activation assay, and a competition binding assay to evaluate their biological properties. We observed that an E-alkene can efficiently replace the first amide bond of Leu(5)- and Met(5)-enkephalin without significantly affecting biological activity. By contrast, the second amide bond was found to be highly sensitive to the same modification, suggesting that it is involved in biologically essential intra- or intermolecular interactions. Finally, we observed that the affinity and activity of analogs containing an E-alkene at either the third or fourth position were partially reduced, indicating that these amide bonds are less important for these intra- or intermolecular interactions. Overall, our study demonstrates that the systematic and sequential replacement of amide bonds by E-alkene represents an efficient way to explore peptide backbones.
Collapse
Affiliation(s)
| | | | | | - Yves L. Dory
- Laboratoire de synthèse supramoléculaire, Département de chimie,
| | | |
Collapse
|
36
|
Dubois D, Gendron L. Delta opioid receptor-mediated analgesia is not altered in preprotachykinin A knockout mice. Eur J Neurosci 2010; 32:1921-9. [DOI: 10.1111/j.1460-9568.2010.07466.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
37
|
Hervera A, Negrete R, Leánez S, Martín-Campos J, Pol O. The role of nitric oxide in the local antiallodynic and antihyperalgesic effects and expression of delta-opioid and cannabinoid-2 receptors during neuropathic pain in mice. J Pharmacol Exp Ther 2010; 334:887-96. [PMID: 20498253 DOI: 10.1124/jpet.110.167585] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Both delta-opioid receptor (DOPr) and cannabinoid-2 receptor (CB2R) agonists attenuate neuropathic pain, but the precise mechanism implicated in these effects is not completely elucidated. We investigated whether nitric oxide synthesized by neuronal (NOS1) or inducible (NOS2) nitric-oxide synthases could modulate DOPr and/or CB2R antiallodynic and antihyperalgesic effects through the peripheral nitric oxide-cGMP-protein kinase G (PKG) pathway activation and affect their expression during neuropathic pain. In wild-type (WT) mice at 21 days after chronic constriction of sciatic nerve, we evaluated the effects of [d-Pen(2),d-Pen(5)]-enkephalin (DPDPE); (2-methyl-1-propyl-1H-indol-3-yl)-1-naphthalenylmethanone (JWH-015); and a NOS1 [N-[(4S)-4-amino-5-[(2-aminoethyl)amino]pentyl]-N'-nitroguanidine tris(trifluoroacetate) salt; NANT], NOS2 [l-N(6)-(1-iminoethyl)-lysine; l-NIL], l-guanylate cyclase [1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one; ODQ], or PKG [(Rp)-8-(para-chlorophenylthio)guanosine-3',5'-cyclic monophosphorothioate; Rp-8-pCPT-cGMPs] inhibitor administered alone or combined. Expression of DOPr and CB2R mRNA in the spinal cord and dorsal root ganglia of naive and nerve-injured WT, NOS1-knockout (KO), and NOS2-KO mice, also was assessed. The subplantar administration of NANT, l-NIL, ODQ, or Rp-8-pCPT-cGMPs dose-dependently inhibited neuropathic pain and enhanced the local effects of DPDPE or JWH-015. Moreover, although the basal levels of DOPr and CB2R mRNA were similar between WT and NOS-KO animals, nerve injury only decreased (DOPr) or increased (CB2R) their expression in the dorsal root ganglia of WT and NOS2-KO mice, and not in NOS1-KO mice. Results suggest that inactivation of the nitric oxide-cGMP-PKG peripheral pathway triggered by NOS1 and NOS2 enhanced the peripheral actions of DOPr and CB2R agonists and that nitric oxide synthesized by NOS1 is implicated in the peripheral regulation of DOPr and CB2R gene transcription during neuropathic pain.
Collapse
Affiliation(s)
- Arnau Hervera
- Grup de Neurofarmacologia Molecular, Institut de Recerca de l'Hospital de la Sta Creu i Sant Pau and Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | | | | |
Collapse
|
38
|
Abstract
Similar to mu opioid receptors, kappa and delta opioid receptors reside in the periphery, the dorsal root ganglion, the spinal cord, and in supraspinal regions associated with pain modulation. Both delta and kappa opioid agonists have been shown to activate pain inhibitory pathways in the central nervous system. Yet, currently there are only a few pharmacologic agents that target kappa receptors, and none that target delta receptors. Spurred by the need for an efficacious analgesic without the unwanted side effects associated with the typical clinical profile of mu opioid agonists, new research has provided insight into why the development of effective kappa and delta opioid receptor agonists has remained elusive thus far, and importantly, how these obstacles may be overcome. For example, for delta opioid agonists to be effective, a state of inflammation may be required as this induces delta opioid receptors to migrate to the surface of neuronal cells and thereby become accessible to delta opioid agonists. Studies have shown that delta opioid agonists can provide relief of inflammatory pain and malignant bone pain. Meanwhile, peripherally restricted kappa opioid agonists have been developed to target kappa opioid receptors located on visceral and somatic afferent nerves for relief of inflammatory, visceral, and neuropathic chronic pain. The recently shown efficacy of these analgesics combined with a possible lower abuse potential and side effect burden than mu opioid receptor agonists makes delta and peripherally restricted kappa opioid receptor agonists promising targets for treating pain.
Collapse
|
39
|
Nonpeptidic Delta (δ) Opioid Agonists and Antagonists of the Diarylmethylpiperazine Class: What Have We Learned? Top Curr Chem (Cham) 2010; 299:121-40. [DOI: 10.1007/128_2010_83] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
40
|
Le Bourdonnec B, Windh RT, Leister LK, Zhou QJ, Ajello CW, Gu M, Chu GH, Tuthill PA, Barker WM, Koblish M, Wiant DD, Graczyk TM, Belanger S, Cassel JA, Feschenko MS, Brogdon BL, Smith SA, Derelanko MJ, Kutz S, Little PJ, DeHaven RN, DeHaven-Hudkins DL, Dolle RE. Spirocyclic delta opioid receptor agonists for the treatment of pain: discovery of N,N-diethyl-3-hydroxy-4-(spiro[chromene-2,4'-piperidine]-4-yl) benzamide (ADL5747). J Med Chem 2009; 52:5685-702. [PMID: 19694468 DOI: 10.1021/jm900773n] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Selective, nonpeptidic delta opioid receptor agonists have been the subject of great interest as potential novel analgesic agents. The discoveries of BW373U86 (1) and SNC80 (2) contributed to the rapid expansion of research in this field. However, poor drug-like properties and low therapeutic indices have prevented clinical evaluation of these agents. Doses of 1 and 2 similar to those required for analgesic activity produce convulsions in rodents and nonhuman primates. Recently, we described a novel series of potent, selective, and orally bioavailable delta opioid receptor agonists. The lead derivative, ADL5859 (4), is currently in phase II proof-of-concept studies for the management of pain. Further structure activity relationship exploration has led to the discovery of ADL5747 (36), which is approximately 50-fold more potent than 4 in an animal model of inflammatory pain. On the basis of its favorable efficacy, safety, and pharmacokinetic profile, 36 was selected as a clinical candidate for the treatment of pain.
Collapse
Affiliation(s)
- Bertrand Le Bourdonnec
- Departments of Chemistry, Adolor Corporation, 700 Pennsylvania Drive, Exton, Pennsylvania 19341, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pradhan AAA, Becker JAJ, Scherrer G, Tryoen-Toth P, Filliol D, Matifas A, Massotte D, Gavériaux-Ruff C, Kieffer BL. In vivo delta opioid receptor internalization controls behavioral effects of agonists. PLoS One 2009; 4:e5425. [PMID: 19412545 PMCID: PMC2672171 DOI: 10.1371/journal.pone.0005425] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Accepted: 03/30/2009] [Indexed: 11/19/2022] Open
Abstract
Background GPCRs regulate a remarkable diversity of biological functions, and are thus often targeted for drug therapies. Stimulation of a GPCR by an extracellular ligand triggers receptor signaling via G proteins, and this process is highly regulated. Receptor activation is typically accompanied by desensitization of receptor signaling, a complex feedback regulatory process of which receptor internalization is postulated as a key event. The in vivo significance of GPCR internalization is poorly understood. In fact, the majority of studies have been performed in transfected cell systems, which do not adequately model physiological environments and the complexity of integrated responses observed in the whole animal. Methods and Findings In this study, we used knock-in mice expressing functional fluorescent delta opioid receptors (DOR-eGFP) in place of the native receptor to correlate receptor localization in neurons with behavioral responses. We analyzed the pain-relieving effects of two delta receptor agonists with similar signaling potencies and efficacies, but distinct internalizing properties. An initial treatment with the high (SNC80) or low (AR-M100390) internalizing agonist equally reduced CFA-induced inflammatory pain. However, subsequent drug treatment produced highly distinct responses. Animals initially treated with SNC80 showed no analgesic response to a second dose of either delta receptor agonist. Concomitant receptor internalization and G-protein uncoupling were observed throughout the nervous system. This loss of function was temporary, since full DOR-eGFP receptor responses were restored 24 hours after SNC80 administration. In contrast, treatment with AR-M100390 resulted in retained analgesic response to a subsequent agonist injection, and ex vivo analysis showed that DOR-eGFP receptor remained G protein-coupled on the cell surface. Finally SNC80 but not AR-M100390 produced DOR-eGFP phosphorylation, suggesting that the two agonists produce distinct active receptor conformations in vivo which likely lead to differential receptor trafficking. Conclusions Together our data show that delta agonists retain full analgesic efficacy when receptors remain on the cell surface. In contrast, delta agonist-induced analgesia is abolished following receptor internalization, and complete behavioral desensitization is observed. Overall these results establish that, in the context of pain control, receptor localization fully controls receptor function in vivo. This finding has both fundamental and therapeutic implications for slow-recycling GPCRs.
Collapse
MESH Headings
- Animals
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Benzamides/pharmacology
- Biological Transport, Active/drug effects
- Cell Membrane/metabolism
- Cells, Cultured
- Green Fluorescent Proteins/chemistry
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- In Vitro Techniques
- Ligands
- Mice
- Mice, Transgenic
- Neurons/drug effects
- Neurons/physiology
- Pain/drug therapy
- Pain/physiopathology
- Phosphorylation
- Piperazines/pharmacology
- Piperidines/pharmacology
- Protein Conformation
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/physiology
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Amynah A. A. Pradhan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
| | - Jérôme A. J. Becker
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
| | - Grégory Scherrer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
| | - Petra Tryoen-Toth
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
| | - Dominique Filliol
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
| | - Audrey Matifas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
| | - Dominique Massotte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
| | - Claire Gavériaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
| | - Brigitte L. Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur, Illkirch, France
- * E-mail:
| |
Collapse
|
42
|
Beaudry H, Proteau-Gagné A, Li S, Dory Y, Chavkin C, Gendron L. Differential noxious and motor tolerance of chronic delta opioid receptor agonists in rodents. Neuroscience 2009; 161:381-91. [PMID: 19328839 DOI: 10.1016/j.neuroscience.2009.03.053] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 03/19/2009] [Accepted: 03/20/2009] [Indexed: 01/28/2023]
Abstract
In the present study, we asked whether multiple intrathecal injections of deltorphin II, a selective delta opioid receptor (DOPR) agonist, induced DOPR tolerance in three behavioral assays. Unilateral inflammation caused by complete Freund's adjuvant (CFA) injection into the rat or mouse hind paw (CFA model) induced thermal hyperalgesic response that was transiently and dose-dependently reduced by intrathecal administration of deltorphin II or morphine. In both rodent species, the effect of deltorphin II was not modified by a single prior administration of deltorphin II, suggesting an absence of acute tolerance in this paradigm. Repeated administration of intrathecal deltorphin II or s.c. SB-235863 (five consecutive injections over 60 h) also failed to impair the antihyperalgesic response to delta opioid receptor agonist, whereas repeated intrathecal or s.c. injections of morphine induced a significant decrease in the subsequent thermal antihyperalgesic response to morphine. In mice, deltorphin II also induced a rapid, transient motor incoordination/ataxia-like behavior as tested with the accelerating rotarod. In contrast to the antihyperalgesic responses, tolerance to the motoric effect of deltorphin II was evident in mice previously exposed to multiple intrathecal agonist injections, but not multiple saline administrations. Using the tail flick antinociceptive test, we found that DOPR-mediated analgesia was significantly reduced by repeated exposure to deltorphin II. Altogether, these observations suggest that repeated injections of DOPR agonists induce differential tolerance effects on antihyperalgesic, antinociceptive, and motor incoordination/ataxia-like behaviors related to DOPR activation by deltorphin II.
Collapse
Affiliation(s)
- H Beaudry
- Department of Physiology and Biophysics, Université de Sherbrooke, Faculty of Medicine, 3001, 12e Avenue Nord, Sherbrooke, QC, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Buesa I, Urrutia A, Aira Z, Salgueiro M, Bilbao J, Mozas M, Aguilera L, Zimmermann M, Azkue JJ. Depression of C fibre-evoked spinal field potentials by the spinal δ opioid receptor is enhanced in the spinal nerve ligation model of neuropathic pain: Involvement of the μ-subtype. Neuropharmacology 2008; 55:1376-82. [DOI: 10.1016/j.neuropharm.2008.08.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 07/23/2008] [Accepted: 08/25/2008] [Indexed: 10/21/2022]
|
44
|
Le Bourdonnec B, Windh RT, Ajello CW, Leister LK, Gu M, Chu GH, Tuthill PA, Barker WM, Koblish M, Wiant DD, Graczyk TM, Belanger S, Cassel JA, Feschenko MS, Brogdon BL, Smith SA, Christ DD, Derelanko MJ, Kutz S, Little PJ, DeHaven RN, DeHaven-Hudkins DL, Dolle RE. Potent, orally bioavailable delta opioid receptor agonists for the treatment of pain: discovery of N,N-diethyl-4-(5-hydroxyspiro[chromene-2,4'-piperidine]-4-yl)benzamide (ADL5859). J Med Chem 2008; 51:5893-6. [PMID: 18788723 DOI: 10.1021/jm8008986] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Selective delta opioid receptor agonists are promising potential therapeutic agents for the treatment of various types of pain conditions. A spirocyclic derivative was identified as a promising hit through screening. Subsequent lead optimization identified compound 20 (ADL5859) as a potent, selective, and orally bioavailable delta agonist. Compound 20 was selected as a clinical candidate for the treatment of pain.
Collapse
|
45
|
Radzievsky AA, Gordiienko OV, Alekseev S, Szabo I, Cowan A, Ziskin MC. Electromagnetic millimeter wave induced hypoalgesia: frequency dependence and involvement of endogenous opioids. Bioelectromagnetics 2008; 29:284-95. [PMID: 18064600 DOI: 10.1002/bem.20389] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Millimeter wave treatment (MMWT) is based on the systemic biological effects that develop following local skin exposure to low power electromagnetic waves in the millimeter range. In the present set of experiments, the hypoalgesic effect of this treatment was analyzed in mice. The murine nose area was exposed to MMW of "therapeutic" frequencies: 42.25, 53.57, and 61.22 GHz. MMWT-induced hypoalgesia was shown to be frequency dependent in two experimental models: (1) the cold water tail-flick test (chronic non-neuropathic pain), and (2) the wire surface test (chronic neuropathic pain following unilateral constriction injury to the sciatic nerve). Maximum hypoalgesic effect was obtained when the frequency was 61.22 GHz. Other exposure parameters were: incident power density = 13.3 mW/cm(2), duration of each exposure = 15 min. Involvement of delta and kappa endogenous opioids in the MMWT-induced hypoalgesia was demonstrated using selective blockers of delta- and kappa-opioid receptors and the direct ELISA measurement of endogenous opioids in CNS tissue. Possible mechanisms of the effect and the perspectives of the clinical application of MMWT are discussed.
Collapse
Affiliation(s)
- A A Radzievsky
- Center for Biomedical Physics, Temple University Medical School, Philadelphia, Pennsylvania 19140, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Benbouzid M, Gavériaux-Ruff C, Yalcin I, Waltisperger E, Tessier LH, Muller A, Kieffer BL, Freund-Mercier MJ, Barrot M. Delta-opioid receptors are critical for tricyclic antidepressant treatment of neuropathic allodynia. Biol Psychiatry 2008; 63:633-6. [PMID: 17693391 DOI: 10.1016/j.biopsych.2007.06.016] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 06/15/2007] [Accepted: 06/18/2007] [Indexed: 11/20/2022]
Abstract
BACKGROUND The therapeutic effect of antidepressant drugs against depression usually necessitates a chronic treatment. A large body of clinical evidence indicates that antidepressant drugs can also be highly effective against chronic neuropathic pain. However, the mechanism by which these drugs alleviate pain is still unclear. METHODS We used a murine model of neuropathic pain induced by sciatic nerve constriction to study the antiallodynic properties of a chronic treatment with the tricyclic antidepressants nortriptyline and amitriptyline. Using knockout and pharmacological approaches in mice, we determined the influence of delta-opioid receptors in the therapeutic action of chronic antidepressant treatment. RESULTS In our model, a chronic treatment by tricyclic antidepressant drugs totally suppresses the mechanical allodynia in neuropathic C57Bl/6J mice. This therapeutic effect can be acutely reversed by an injection of the delta-opioid receptor antagonist naltrindole. Moreover, the antiallodynic property of antidepressant treatment is absent in mice deficient for the delta-opioid receptor gene. CONCLUSIONS The antiallodynic effect of chronic antidepressant treatment is mediated by a recruitment of the endogenous opioid system acting through delta-opioid receptors.
Collapse
Affiliation(s)
- Malika Benbouzid
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de Recherche Scientifique/Université Louis Pasteur, Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Essential role of mu opioid receptor in the regulation of delta opioid receptor-mediated antihyperalgesia. Neuroscience 2007; 150:807-17. [PMID: 17997230 DOI: 10.1016/j.neuroscience.2007.09.060] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 09/27/2007] [Accepted: 10/04/2007] [Indexed: 11/21/2022]
Abstract
Analgesic effects of delta opioid receptor (DOR) -selective agonists are enhanced during persistent inflammation and arthritis. Although the underlying mechanisms are still unknown, membrane density of DOR was shown to be increased 72 h after induction of inflammation, an effect abolished in mu opioid receptor (MOR) -knockout (KO) mice [Morinville A, Cahill CM, Kieffer B, Collier B, Beaudet A (2004b) Mu-opioid receptor knockout prevents changes in delta-opioid receptor trafficking induced by chronic inflammatory pain. Pain 109:266-273]. In this study, we demonstrated a crucial role of MOR in DOR-mediated antihyperalgesia. Intrathecal administration of the DOR selective agonist deltorphin II failed to induce antihyperalgesic effects in MOR-KO mice, whereas it dose-dependently reversed thermal hyperalgesia in wild-type mice. The antihyperalgesic effects of deltorphin II were blocked by naltrindole but not d-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2) (CTOP) suggesting that this agonist was mainly acting through DOR. SNC80-induced antihyperalgesic effects in MOR-KO mice were also attenuated as compared with littermate controls. In contrast, kappa opioid receptor knockout did not affect deltorphin II-induced antihyperalgesia. As evaluated using mice lacking endogenous opioid peptides, the regulation of DOR's effects was also independent of beta-endorphin, enkephalins, or dynorphin opioids known to be released during persistent inflammation. We therefore conclude that DOR-mediated antihyperalgesia is dependent on MOR expression but that activation of MOR by endogenous opioids is probably not required.
Collapse
|
48
|
Aceto MD, May EL, Harris LS, Bowman ER, Cook CD. Pharmacological studies with a nonpeptidic, delta-opioid (-)-(1R,5R,9R)-5,9-dimethyl-2'-hydroxy-2-(6-hydroxyhexyl)-6,7-benzomorphan hydrochloride ((-)-NIH 11082). Eur J Pharmacol 2007; 566:88-93. [PMID: 17434480 PMCID: PMC2994320 DOI: 10.1016/j.ejphar.2007.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 02/28/2007] [Accepted: 03/06/2007] [Indexed: 11/25/2022]
Abstract
In the search for a selective delta-opioid receptor agonist, (-)-(1R,5R,9R)-5,9-dimethyl-2'-hydroxy-2-(6-hydroxyhexyl)-6,7-benzomorphan hydrochloride ((-)-NIH 11082) and the (+)-enantiomer were synthesized and tested. (-)-NIH 11082 displayed antinociceptive activity in the paraphenylquinone test (PPQ test) in male ICR mice [ED50=1.9 (0.7-5.3) mg/kg, s.c.] and showed little, if any, activity in the tail-flick and hot-plate assays. The (+)-enantiomer was essentially inactive indicating stereoselectivity. Opioid receptor subtype characterization studies indicated that naltrindole, a delta-opioid receptor antagonist, was potent versus the ED80 of (-)-NIH 11082 in the PPQ test [AD50=0.75 (0.26-2.20) mg/kg, s.c]. beta-Funaltrexamine and nor-binaltorphimine, selective mu- and kappa-receptor antagonists, respectively, were inactive versus the ED80 of (-)-NIH 11082. In rats with inflammation-induced pain, (-)-NIH 11082 produced antihyperalgesic effects that were attenuated by naltrindole. In morphine-dependent rhesus monkeys of both sexes, (-)-NIH 11082 neither substituted for morphine nor exacerbated withdrawal signs in the dose range of 4.0 to 32.0 mg/kg, s.c. Neither convulsions nor other overt behavioral signs were observed in any of the species tested. The results indicate that (-)-NIH 11082 has delta-opioid receptor properties.
Collapse
Affiliation(s)
- Mario D Aceto
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0613, USA.
| | | | | | | | | |
Collapse
|
49
|
Gendron L, Esdaile MJ, Mennicken F, Pan H, O'Donnell D, Vincent JP, Devi LA, Cahill CM, Stroh T, Beaudet A. Morphine priming in rats with chronic inflammation reveals a dichotomy between antihyperalgesic and antinociceptive properties of deltorphin. Neuroscience 2007; 144:263-74. [PMID: 17055663 DOI: 10.1016/j.neuroscience.2006.08.077] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Revised: 08/26/2006] [Accepted: 08/29/2006] [Indexed: 10/24/2022]
Abstract
We previously showed that prolonged morphine treatment and chronic inflammation both enhanced delta opioid receptor (deltaOR) cell surface density in lumbar spinal cord neurons. Here, we sought to determine whether administration of morphine to rats with chronic inflammation would further increase the bio-availability of deltaOR, and thereby the analgesic properties of the deltaOR agonist deltorphin, over that produced by inflammation alone. We found that chronic inflammation produced by injection of complete Freund's adjuvant (CFA) into the hind paw resulted in a bilateral increase in the binding and internalization of fluorescent deltorphin in neurons of the lumbar spinal cord as did prolonged morphine treatment [Morinville A, Cahill CM, Aibak H, Rymar VV, Pradhan A, Hoffert C, Mennicken F, Stroh T, Sadikot AF, O'Donnell D, Clarke PB, Collier B, Henry JL, Vincent JP, Beaudet A (2004a) Morphine-induced changes in delta opioid receptor trafficking are linked to somatosensory processing in the rat spinal cord. J Neurosci 24:5549-5559]. This effect was accompanied by an increase in the antinociceptive efficacy of intrathecal deltorphin as measured using the tail-flick test. Treatment of CFA-injected rats with morphine decreased the cell surface availability of deltaOR in neurons of the dorsal horn of the lumbar spinal cord as compared with treatment with CFA alone. Behaviorally, it significantly enhanced the antihyperalgesic effects of deltorphin (plantar test; % maximum possible antihyperalgesic effect (MPAHE)=113.5%+/-32.4% versus 26.1%+/-11.6% in rats injected with CFA alone) but strongly reduced the antinociceptive efficacy of the drug (tail-flick test; % maximum possible antinociceptive effect (MPE)=29.6%+/-3.6% versus 66.6%+/-6.3% in rats injected with CFA alone) suggesting that the latter, but not the former, is linked to the deltaOR trafficking events observed neuroanatomically. These results demonstrate that in chronic inflammation, the antihyperalgesic effects of deltaOR agonists may be enhanced by morphine pre-treatment. They also reveal a dichotomy between mechanisms underlying antihyperalgesic and antinociceptive effects of deltaOR agonists.
Collapse
MESH Headings
- Analgesics
- Analgesics, Opioid/pharmacology
- Animals
- Behavior, Animal/drug effects
- Chronic Disease
- Dose-Response Relationship, Drug
- Enkephalin, Methionine/metabolism
- Freund's Adjuvant
- Hyperalgesia/drug therapy
- Hyperalgesia/etiology
- Hyperalgesia/psychology
- Inflammation/chemically induced
- Inflammation/complications
- Male
- Morphine/pharmacology
- Oligopeptides/administration & dosage
- Oligopeptides/pharmacology
- Pain Threshold/drug effects
- Rats
- Rats, Sprague-Dawley
- Reaction Time/drug effects
- Receptors, Cell Surface/drug effects
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Spinal Cord/drug effects
- Spinal Cord/metabolism
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- L Gendron
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Room 896, 3801 University Street, McGill University, Montreal, Quebec, Canada H3A 2B4
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Holdridge SV, Cahill CM. Spinal administration of a delta opioid receptor agonist attenuates hyperalgesia and allodynia in a rat model of neuropathic pain. Eur J Pain 2006; 11:685-93. [PMID: 17175187 DOI: 10.1016/j.ejpain.2006.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Revised: 09/14/2006] [Accepted: 10/30/2006] [Indexed: 12/17/2022]
Abstract
Neuropathic (NP) pain is a debilitating chronic pain disorder considered by some to be inherently resistant to therapy with traditional analgesics. Indeed, micro opioid receptor (OR) agonists show reduced therapeutic benefit and their long term use is hindered by the high incidence of adverse effects. However, pharmacological and physiological evidence increasingly suggests a role for deltaOR agonists in modulating NP pain symptoms. In this study, we examined the antihyperalgesic and antiallodynic effects of the spinally administered deltaOR agonist, d-[Ala(2), Glu(4)]deltorphin II (deltorphin II), as well as the changes in deltaOR expression, in rats following chronic constriction injury (CCI) of the sciatic nerve. Rats with CCI exhibited cold hyperalgesia and mechanical allodynia over a 14-day testing period. Intrathecal administration of deltorphin II reversed cold hyperalgesia on day 14 and dose-dependently attenuated mechanical allodynia. The effects of deltorphin II were mediated via activation of the deltaOR as the effect was antagonized by co-treatment with the delta-selective antagonist, naltrindole. Western blotting experiments revealed no changes in deltaOR protein in the dorsal spinal cord following CCI. Taken together, these data demonstrate the antihyperalgesic and antiallodynic effectiveness of a spinally administered deltaOR agonist following peripheral nerve injury and support further investigation of deltaORs as potential therapeutic targets in the treatment of NP pain.
Collapse
Affiliation(s)
- Sarah V Holdridge
- Department of Pharmacology and Toxicology, Queen's University, Kingston, Ont, Canada
| | | |
Collapse
|