1
|
Wang J, Li Z, Tu Y, Gao F. The Dopaminergic System in the Ventral Tegmental Area Contributes to Morphine Analgesia and Tolerance. Neuroscience 2023; 527:74-83. [PMID: 37286162 DOI: 10.1016/j.neuroscience.2023.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 05/17/2023] [Accepted: 05/27/2023] [Indexed: 06/09/2023]
Abstract
Morphine has a strong analgesic effect and is suitable for various types of pain, so it is widely used. But long-term usage of morphine can lead to drug tolerance, which limits its clinical application. The complex mechanisms underlying the development of morphine analgesia into tolerance involve multiple nuclei in the brain. Recent studies reveal the signaling at the cellular and molecular levels as well as neural circuits contributing to morphine analgesia and tolerance in the ventral tegmental area (VTA), which is traditionally considered a critical center of opioid reward and addiction. Existing studies show that dopamine receptors and μ-opioid receptors participate in morphine tolerance through the altered activities of dopaminergic and/or non-dopaminergic neurons in the VTA. Several neural circuits related to the VTA are also involved in the regulation of morphine analgesia and the development of drug tolerance. Reviewing specific cellular and molecular targets and related neural circuits may provide novel precautionary strategies for morphine tolerance.
Collapse
Affiliation(s)
- Jihong Wang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Tu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Parishar P, Sehgal N, Iyengar S. The expression of delta opioid receptor mRNA in adult male zebra finches (Taenopygia guttata). PLoS One 2021; 16:e0256599. [PMID: 34464410 PMCID: PMC8407588 DOI: 10.1371/journal.pone.0256599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022] Open
Abstract
The endogenous opioid system is evolutionarily conserved across reptiles, birds and mammals and is known to modulate varied brain functions such as learning, memory, cognition and reward. To date, most of the behavioral and anatomical studies in songbirds have mainly focused on μ-opioid receptors (ORs). Expression patterns of δ-ORs in zebra finches, a well-studied species of songbird have not yet been reported, possibly due to the high sequence similarity amongst different opioid receptors. In the present study, a specific riboprobe against the δ-OR mRNA was used to perform fluorescence in situ hybridization (FISH) on sections from the male zebra finch brain. We found that δ-OR mRNA was expressed in different parts of the pallium, basal ganglia, cerebellum and the hippocampus. Amongst the song control and auditory nuclei, HVC (abbreviation used as a formal name) and NIf (nucleus interfacialis nidopallii) strongly express δ-OR mRNA and stand out from the surrounding nidopallium. Whereas the expression of δ-OR mRNA is moderate in LMAN (lateral magnocellular nucleus of the anterior nidopallium), it is low in the MSt (medial striatum), Area X, DLM (dorsolateral nucleus of the medial thalamus), RA (robust nucleus of the arcopallium) of the song control circuit and Field L, Ov (nucleus ovoidalis) and MLd (nucleus mesencephalicus lateralis, pars dorsalis) of the auditory pathway. Our results suggest that δ-ORs may be involved in modulating singing, song learning as well as spatial learning in zebra finches.
Collapse
Affiliation(s)
- Pooja Parishar
- National Brain Research Centre, Gurugram, Haryana, India
| | - Neha Sehgal
- National Brain Research Centre, Gurugram, Haryana, India
| | - Soumya Iyengar
- National Brain Research Centre, Gurugram, Haryana, India
| |
Collapse
|
3
|
Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 2016; 68:631-700. [PMID: 27343248 PMCID: PMC4931872 DOI: 10.1124/pr.114.008979] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [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
|
4
|
Peppin JF, Raffa RB. Delta opioid agonists: a concise update on potential therapeutic applications. J Clin Pharm Ther 2015; 40:155-66. [PMID: 25726896 DOI: 10.1111/jcpt.12244] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/24/2014] [Indexed: 01/23/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE The endogenous opioid system co-evolved with chemical defences, or at times symbiotic relationships, between plants and other autotrophs and heterotrophic predators - thus, it is not surprising that endogenous opioid ligands and exogenous mimetic ligands produce diverse physiological effects. Among the endogenous opioid peptides (endomorphins, enkephalins, dynorphins and nociception/orphanin FQ) derived from the precursors encoded by four genes (PNOC, PENK, PDYN and POMC) are the pentapeptides Met-enkephalin (Tyr-Gly-Gly-Phe-Met) and Leu-enkephalin (Tyr-Gly-Gly-Phe-Leu). The physiological effects of the enkephalins are mediated via 7-transmembrane G protein-coupled receptors, including delta opioid receptor (DOR). We present a concise update on the status of progress and opportunities of this approach. METHODS A literature search of the PUBMED database and a combination of keywords including delta opioid receptor, analgesia, mood and individual compounds identified therein, from industry and other source, and from www.clinicaltrials.com. RESULTS AND DISCUSSION DOR agonist and antagonist ligands have been developed with ever increasing affinity and selectivity for DOR over other opioid receptor subtypes and studied for therapeutic utility, primarily for pain relief, but also for other clinical endpoints. WHAT IS NEW AND CONCLUSION Selective DOR agonists have been designed with a large increase in therapeutic window for a variety of potential CNS applications including pain, depression, and learning and memory among others.
Collapse
Affiliation(s)
- J F Peppin
- Center for Bioethics, Pain Management and Medicine, University City, MO, USA; Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | | |
Collapse
|
5
|
Lee CWS, Ho IK. Pharmacological Profiles of Oligomerized μ-Opioid Receptors. Cells 2013; 2:689-714. [PMID: 24709876 PMCID: PMC3972655 DOI: 10.3390/cells2040689] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/30/2013] [Accepted: 10/09/2013] [Indexed: 01/21/2023] Open
Abstract
Opioids are widely prescribed pain relievers with multiple side effects and potential complications. They produce analgesia via G-protein-protein coupled receptors: μ-, δ-, κ-opioid and opioid receptor-like 1 receptors. Bivalent ligands targeted to the oligomerized opioid receptors might be the key to developing analgesics without undesired side effects and obtaining effective treatment for opioid addicts. In this review we will update the biological effects of μ-opioids on homo- or hetero-oligomerized μ-opioid receptor and discuss potential mechanisms through which bivalent ligands exert beneficial effects, including adenylate cyclase regulation and receptor-mediated signaling pathways.
Collapse
Affiliation(s)
- Cynthia Wei-Sheng Lee
- Center for Drug Abuse and Addiction, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Ing-Kang Ho
- Center for Drug Abuse and Addiction, China Medical University Hospital, Taichung 40447, Taiwan.
| |
Collapse
|
6
|
Kwok CHT, Devonshire IM, Bennett AJ, Hathway GJ. Postnatal maturation of endogenous opioid systems within the periaqueductal grey and spinal dorsal horn of the rat. Pain 2013; 155:168-178. [PMID: 24076162 PMCID: PMC3894430 DOI: 10.1016/j.pain.2013.09.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/03/2013] [Accepted: 09/20/2013] [Indexed: 12/31/2022]
Abstract
Significant opioid-dependent changes occur during the fourth postnatal week in supraspinal sites (rostroventral medulla [RVM], periaqueductal grey [PAG]) that are involved in the descending control of spinal excitability via the dorsal horn (DH). Here we report developmentally regulated changes in the opioidergic signalling within the PAG and DH, which further increase our understanding of pain processing during early life. Microinjection of the μ-opioid receptor (MOR) agonist DAMGO (30 ng) into the PAG of Sprague-Dawley rats increased spinal excitability and lowered mechanical threshold to noxious stimuli in postnatal day (P)21 rats, but had inhibitory effects in adults and lacked efficacy in P10 pups. A tonic opioidergic tone within the PAG was revealed in adult rats by intra-PAG microinjection of CTOP (120 ng, MOR antagonist), which lowered mechanical thresholds and increased spinal reflex excitability. Spinal adminstration of DAMGO inhibited spinal excitability in all ages, yet the magnitude of this was greater in younger animals than in adults. The expression of MOR and related peptides were also investigated using TaqMan real-time polymerase chain reaction and immunohistochemistry. We found that pro-opiomelanocortin peaked at P21 in the ventral PAG, and MOR increased significantly in the DH as the animals aged. Enkephalin mRNA transcripts preceded the increase in enkephalin immunoreactive fibres in the superficial dorsal horn from P21 onwards. These results illustrate that profound differences in the endogenous opioidergic signalling system occur throughout postnatal development.
Collapse
Affiliation(s)
- Charlie H T Kwok
- Laboratory of Developmental Nociception, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK FRAME Laboratory, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | | | | | | |
Collapse
|
7
|
Ballesta JJ, Cremades J, Rodríguez-Muñoz M, Garzón J, Faura CC. Sensitivity to μ-opioid receptor-mediated anti-nociception is determined by cross-regulation between μ- and δ-opioid receptors at supraspinal level. Br J Pharmacol 2012; 166:309-26. [PMID: 22023056 DOI: 10.1111/j.1476-5381.2011.01750.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The perception of pain and its inhibition varies considerably between individuals, and this variability is still unexplained. The aim of the present study is to determine whether functional interactions between opioid receptors are involved in the inter-individual variability in the sensitivity to μ-opioid receptor agonists. EXPERIMENTAL APPROACH Anti-nociceptive tests, radioligand binding, stimulation of [(35) S]GTP-γ-S binding, inhibition of cAMP production and co-immunoprecipitation experiments were performed in two strains of rat (Sprague-Dawley bred at our university - SDU - and Wistar) that differ in their sensitivity to opioids. KEY RESULTS The increased anti-nociceptive potency of µ-opioid receptor agonists in SDU rats was reversed by the δ-opioid receptor antagonist, naltrindole. Inhibition of the binding of [(3) H] naltrindole by µ-opioid receptor agonists was different in brain membranes from SDU and Wistar rats. Differences were also evident in the effect of δ-opioid receptor ligands on the binding of [(35) S]GTP-γ-S stimulated by µ-opioid receptors agonists. No strain-related differences were detected in spinal cord membranes. The potency of morphine to inhibit cAMP production in brain membranes varied between the strains, in the presence of deltorphin II and naltrindole. Co-immunoprecipitation experiments demonstrated that δ-opioid receptors were associated with μ-opioid receptors to a higher extent in brain synaptosomal fractions from SDU than in those from Wistar rats. CONCLUSIONS AND IMPLICATIONS There was increased supraspinal cross-talk between μ and δ-opioid receptors in SDU, as compared with Wistar rats. This was related to an enhanced sensitivity to anti-nociception induced by µ-opioid receptor agonists.
Collapse
Affiliation(s)
- J J Ballesta
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernandez, Alicante, Spain
| | | | | | | | | |
Collapse
|
8
|
Abstract
Opiates such as morphine and fentanyl, a major class of analgesics used in the clinical management of pain, exert their effects through the activation of opioid receptors. Opioids are among the most commonly prescribed and frequently abused drugs in the USA; however, the prolonged use of opiates often leads to the development of tolerance and addiction. Although blockade of opioid receptors with antagonists such as naltrexone and naloxone can lessen addictive impulses and facilitate recovery from overdose, systemic disruption of endogenous opioid receptor signalling through the use of these antagonistic drugs can have severe side effects. In the light of these challenges, current efforts have focused on identifying new therapeutic targets that selectively and specifically modulate opioid receptor signalling and function so as to achieve analgesia without the adverse effects associated with chronic opiate use. We have previously reported that opioid receptors interact with each other to form heteromeric complexes and that these interactions affect morphine signalling. Since chronic morphine administration leads to an enhanced level of these heteromers, these opioid receptor heteromeric complexes represent novel therapeutic targets for the treatment of pain and opiate addiction. In this review, we discuss the role of heteromeric opioid receptor complexes with a focus on mu opioid receptor (MOR) and delta opioid receptor (DOR) heteromers. We also highlight the evidence for altered pharmacological properties of opioid ligands and changes in ligand function resulting from the heteromer formation.
Collapse
|
9
|
Kumar K, Goyal R, Mudgal A, Mohan A, Pasha S. YFa and analogs: Investigation of opioid receptors in smooth muscle contraction. World J Gastroenterol 2011; 17:4523-31. [PMID: 22110284 PMCID: PMC3218144 DOI: 10.3748/wjg.v17.i40.4523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/16/2011] [Accepted: 06/23/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the pharmacological profile and inhibition of smooth muscle contraction by YFa and its analogs in conjunction with their receptor selectivity.
METHODS: The effects of YFa and its analogs (D-Ala2) YFa, Y (D-Ala2) GFMKKKFMRF amide and Des-Phe-YGGFMKKKFMR amide in guinea pig ileum (GPI) and mouse vas deferens (MVD) motility were studied using an isolated tissue organ bath system, and morphine and DynA (1-13) served as controls. Acetylcholine was used for muscle stimulation. The observations were validated by specific antagonist pretreatment experiments using naloxonazine, naltrindole and norbinaltorphimine norBNI.
RESULTS: YFa did not demonstrate significant inhibition of GPI muscle contraction as compared with morphine (15% vs 62%, P = 0.0002), but moderate inhibition of MVD muscle contraction, indicating the role of κ opioid receptors in the contraction. A moderate inhibition of GPI muscles by (Des-Phe) YFa revealed the role of anti-opiate receptors in the smooth muscle contraction. (D-Ala-2) YFa showed significant inhibition of smooth muscle contraction, indicating the involvement of mainly δ receptors in MVD contraction. These results were supported by specific antagonist pretreatment assays.
CONCLUSION: YFa revealed its side-effect-free analgesic properties with regard to arrest of gastrointestinal transit. The study provides evidences for the involvement of κ and anti-opioid receptors in smooth muscle contraction.
Collapse
|
10
|
Abstract
Twin and triplet drugs are defined as compounds that contain respectively two and three pharmacophore components exerting pharmacological effects in a molecule. The twin drug bearing the same pharmacophores is a "symmetrical twin drug", whereas that possessing different pharmacophores is a "nonsymmetrical twin drug." In general, the symmetrical twin drug is expected to produce more potent and/or selective pharmacological effects, whereas the nonsymmetrical twin drug is anticipated to show both pharmacological activities stemming from the individual pharmacophores (dual action). On the other hand, nonsymmetrical triplet drugs, which have two of the same pharmacophores and one different moiety, are expected to elicit both increased pharmacological action and dual action. The two identical portions could bind the same receptor sites simultaneously while the third portion could bind a different receptor site or enzyme. This review will mainly focus on the twin and triplet drugs with an evaluation of their in vivo pharmacological effects, and will also include a description of their pharmacology and synthesis.
Collapse
Affiliation(s)
- Hideaki Fujii
- School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
11
|
D'ALAGNI MARIA, GULLÁ PATRIZIA, RODA LGIORGIO, ROSCETTI GIANNA. In vitvo interactions of opioid peptides with phospholipids. ACTA ACUST UNITED AC 2009. [DOI: 10.1111/j.1399-3011.1990.tb00057.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
12
|
JUDD AMRITK, LAWSON JOHNA, OLSEN CRISM, TOLL LAWRENCER, POLGAR WILLMAE, UYENO EDWARDT, KEYS CHRISTOPHERJ, LOEW GILDAH. Novel μ-selective Met-enkephalinamide analogs with antagonist activity. ACTA ACUST UNITED AC 2009. [DOI: 10.1111/j.1399-3011.1987.tb03336.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Rutherford JM, Wang J, Xu H, Dersch CM, Partilla JS, Rice KC, Rothman RB. Evidence for a mu-delta opioid receptor complex in CHO cells co-expressing mu and delta opioid peptide receptors. Peptides 2008; 29:1424-31. [PMID: 18472184 DOI: 10.1016/j.peptides.2008.03.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Revised: 03/21/2008] [Accepted: 03/25/2008] [Indexed: 11/25/2022]
Abstract
Based on non-competitive binding interactions we suggested that mu and delta receptors associate as a mu/delta receptor complex in rat brain. We hypothesized that the same non-competitive binding interactions observed in rat brain will be seen in CHO cells that co-express mu and delta receptors, but not in cells that express just mu or delta receptors. We used CHO cells expressing the cloned human mu receptor, cloned human delta receptor, or cloned mouse delta/human mu ("dimer cell"). Cell membranes were prepared from intact cells pretreated with 100nM SUPERFIT. [(3)H][d-Ala(2),d-Leu(5)]enkephalin binding assays followed published procedures. SUPERFIT, a delta-selective irreversible ligand, decreased [(3)H][d-Ala(2),d-Leu(5)]enkephalin binding to delta receptors by approximately 75% and to mu receptors by approximately 50% in dimer cells. SUPERFIT treatment did not decrease [(3)H][d-Ala(2),d-Leu(5)]enkephalin binding to mu cells. The IC(50) values observed in SUPERFIT-treated dimer cells were: [d-Pen(2),d-Pen(5)]enkephalin (1820nM) and morphine (171nM). Saturation binding experiments with SUPERFIT-treated dimer cells showed that [d-Pen(2),d-Pen(5)]enkephalin (5000nM) was a competitive inhibitor. In contrast, morphine (1000nM) lowered the B(max) from 1944fmol/mg to 1276fmol/mg protein (35% decrease). Both [d-Pen(2),d-Pen(5)]enkephalin and morphine competitively inhibited [(3)H][d-Ala(2),d-Leu(5)]enkephalin binding to SUPERFIT-treated mu cells. The results indicate that the mu-delta opioid receptor complex defined on the basis of non-competitive binding interactions in rat brain over 20 years ago likely occurs as a consequence of the formation of mu-delta heterodimers. SUPERFIT-treated dimer cells may provide a useful model to study the properties of mu-delta heterodimers.
Collapse
MESH Headings
- Animals
- Binding Sites
- CHO Cells
- Cricetinae
- Cricetulus
- Dimerization
- Enkephalin, Leucine-2-Alanine/pharmacology
- Female
- Humans
- Inhibitory Concentration 50
- Ligands
- Mice
- Ovary/cytology
- Ovary/drug effects
- Ovary/metabolism
- Receptor Aggregation/drug effects
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
Collapse
Affiliation(s)
- John M Rutherford
- Clinical Psychopharmacology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, DHHS, Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
|
15
|
Raut A, Iglewski M, Ratka A. Differential effects of impaired mitochondrial energy production on the function of mu and delta opioid receptors in neuronal SK-N-SH cells. Neurosci Lett 2006; 404:242-6. [PMID: 16808998 DOI: 10.1016/j.neulet.2006.05.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Revised: 05/17/2006] [Accepted: 05/31/2006] [Indexed: 10/24/2022]
Abstract
Oxidative stress contributes to changes in neurosensory processing, including pain, that occur during aging and neurodegeneration. The effects of neuronal oxidation on the opioid system are poorly understood. In this in vitro study, oxidative stress was induced by 3-nitroproprionic acid (3-NPA) in opioid-responsive differentiated SK-N-SH cells. Changes in the inhibitory effects of opioid receptor agonists on intracellular cAMP were used as a marker of the function of mu and delta opioid receptors (MOR and DOR, respectively). Cells were treated with morphine and selective MOR and DOR agonists and antagonists to characterize the function of each receptor subtype. Cyclic AMP (cAMP) was measured by enzyme immunoassay. Levels of reactive oxygen species (ROS) were assessed using the 2',7'-dichlorofluorescin diacetate assay. Exposure of cells to 3-NPA resulted in an increase in ROS. After 3-NPA exposure, there was a significant attenuation of the inhibitory effect of morphine and DAMGO but not of DPDPE on cAMP. In cells pretreated with CTOP, 3-NPA did not change the inhibitory effect on cAMP. These findings demonstrate for the first time that under conditions of mitochondrial damage, the function of MOR is significantly decreased, while the function of DOR does not change, suggesting that the effect of 3-NPA on opioid receptors is subtype-specific.
Collapse
MESH Headings
- Cell Line, Tumor
- Energy Metabolism/physiology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Humans
- Mitochondria/metabolism
- Neuroblastoma
- Neurons/physiology
- Nitro Compounds/pharmacology
- Propionates/pharmacology
- Reactive Oxygen Species/metabolism
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/physiology
Collapse
Affiliation(s)
- Atul Raut
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, 76107, USA
| | | | | |
Collapse
|
16
|
Yamada H, Shimoyama N, Sora I, Uhl GR, Fukuda Y, Moriya H, Shimoyama M. Morphine can produce analgesia via spinal kappa opioid receptors in the absence of mu opioid receptors. Brain Res 2006; 1083:61-9. [PMID: 16530171 DOI: 10.1016/j.brainres.2006.01.095] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2005] [Revised: 01/23/2006] [Accepted: 01/25/2006] [Indexed: 11/24/2022]
Abstract
Previous studies have demonstrated the virtual lack of analgesia in mu opioid receptor knockout mice after systemic administration of morphine. Thus, it has been suggested that analgesic actions of morphine are produced via the mu opioid receptor, despite its ability to bind to kappa and delta receptors in vitro. However, it is not clear whether the results of these studies reflect the effect of morphine in the spinal cord. In the present study, we report study of the analgesic actions of spinally-administered morphine and other opioid receptor agonists in mu opioid receptor knockout and wild type mice. Morphine produced a dose-dependent antinociceptive effect in the tail flick test in the knockout mice, although higher doses were needed to produce antinociception than in wild type mice. The antinociceptive effect of morphine was completely blocked by naloxone (a non-selective opioid antagonist) and nor-binaltorphimine (nor-BNI, a selective kappa-opioid receptor antagonist), but not by naltrindole (a selective delta-opioid receptor antagonist). U-50,488H (a selective kappa-opioid receptor agonist) also produced a dose-dependent antinociceptive effect in knockout mice but presented lower analgesic potency in knockout mice than in wild type mice. Analgesic effects of [d-Pen2,d-Pen5]enkephalin (DPDPE, a selective delta-opioid receptor agonist) were observed in wild type mice but abolished in knockout mice. SNC80 (a selective delta-opioid receptor agonist) was not antinociceptive even in wild type mice. The present study demonstrated that morphine can produce thermal antinociception via the kappa opioid receptor in the spinal cord in the absence of the mu opioid receptor. Lower potency of U50,488H in mu opioid receptor knockout mice suggests interaction between kappa and mu opioid receptors at the spinal level.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Drug Tolerance/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Morphine/pharmacology
- Narcotic Antagonists/pharmacology
- Pain/drug therapy
- Pain/genetics
- Pain/metabolism
- Pain Measurement/drug effects
- Pain Threshold/drug effects
- Pain Threshold/physiology
- Reaction Time/drug effects
- Reaction Time/physiology
- Receptor Cross-Talk/physiology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/genetics
- Spinal Cord/drug effects
- Spinal Cord/metabolism
Collapse
Affiliation(s)
- Hiroaki Yamada
- Department of Autonomic Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba-ken 260-8670, Japan
| | | | | | | | | | | | | |
Collapse
|
17
|
Daniels DJ, Lenard NR, Etienne CL, Law PY, Roerig SC, Portoghese PS. Opioid-induced tolerance and dependence in mice is modulated by the distance between pharmacophores in a bivalent ligand series. Proc Natl Acad Sci U S A 2005; 102:19208-13. [PMID: 16365317 PMCID: PMC1323165 DOI: 10.1073/pnas.0506627102] [Citation(s) in RCA: 247] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Accepted: 11/03/2005] [Indexed: 11/18/2022] Open
Abstract
Given the mounting evidence for involvement of delta opioid receptors in the tolerance and physical dependence of mu opioid receptor agonists, we have investigated the possible physical interaction between mu and delta opioid receptors by using bivalent ligands. Based on reports of suppression of antinociceptive tolerance by the delta antagonist naltrindole (NTI), bivalent ligands [mu-delta agonist-antagonist (MDAN) series] that contain different length spacers, and pharmacophores derived from NTI and the mu agonist oxymorphone, have been synthesized and evaluated by intracerebroventricular (i.c.v.) administration in the tail-flick test in mice. In acute i.c.v. studies, the bivalent ligands functioned as agonists with potencies ranging from 1.6- to 45-fold greater than morphine. In contrast, the monovalent mu agonist analogues were substantially more potent than the MDAN congeners and were essentially equipotent with one another and oxymorphone. Pretreatment with NTI decreased the ED(50) values for MDAN-19 to a greater degree than for MDAN-16 but had no effect on MDAN-21. Chronic i.c.v. studies revealed that MDAN ligands whose spacer was 16 atoms or longer produced less dependence than either morphine or mu monovalent control MA-19. On the other hand, both physical dependence and tolerance were suppressed at MDAN spacer lengths of 19 atoms or greater. These data suggest that physical interaction between the mu and delta opioid receptors modulates mu-mediated tolerance and dependence. Because MDAN-21 was found to be 50-fold more potent than morphine by the i.v. route (i.v.), it offers a previously uncharacterized approach for the development of analgesics devoid of tolerance and dependence.
Collapse
MESH Headings
- Analgesics/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Chemistry, Pharmaceutical/methods
- Dose-Response Relationship, Drug
- Ligands
- Male
- Mice
- Mice, Inbred ICR
- Models, Chemical
- Models, Statistical
- Morphine/pharmacology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Narcotics/metabolism
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, mu/chemistry
- Time Factors
Collapse
Affiliation(s)
- David J Daniels
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Cannabinoids and opioids both produce analgesia through a G-protein-coupled mechanism that blocks the release of pain-propagating neurotransmitters in the brain and spinal cord. However, high doses of these drugs, which may be required to treat chronic, severe pain, are accompanied by undesirable side effects. Thus, a search for a better analgesic strategy led to the discovery that delta 9-tetrahydrocannabinol (THC), the major psychoactive constituent of marijuana, enhances the potency of opioids such as morphine in animal models. In addition, studies have determined that the analgesic effect of THC is, at least in part, mediated through delta and kappa opioid receptors, indicating an intimate connection between cannabinoid and opioid signaling pathways in the modulation of pain perception. A host of behavioral and molecular experiments have been performed to elucidate the role of opioid receptors in cannabinoid-induced analgesia, and some of these findings are presented below. The aim of such studies is to develop a novel analgesic regimen using low dose combinations of cannabinoids and opioids to effectively treat acute and chronic pain, especially pain that may be resistant to opioids alone.
Collapse
Affiliation(s)
- Diana L Cichewicz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
19
|
Liu ZH, Jin WQ, Dai QY, Chen XJ, Zhang HP, Chi ZQ. Opioid activity of C8813, a novel and potent opioid analgesic. Life Sci 2003; 73:233-41. [PMID: 12738037 DOI: 10.1016/s0024-3205(03)00263-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Compound trans-4-(p-bromophenyl)-4-(dimethylamino)-1-(2-thiophen-2-yl-ethyl)-cyclohexanol (C8813), structurally unrelated to morphine, is a novel analgesic. The present study examined the antinociception, opioid receptor selectivity and in vitro activity of C8813. The antinociceptive activity was evaluated using mouse hot plate and acetic acid writhing tests. In mouse hot plate test, the antinociceptive ED(50) of C8813 was 11.5 microg/kg, being 591 times and 3.4 times more potent than morphine and fentanyl respectively. In mouse writhing test, the antinociceptive ED(50) of C8813 was 16.9 microg/kg, being 55 times and 2.3 times more active than morphine and fentanyl respectively. In the opioid receptor binding assay, C8813 showed high affinity for mu-opioid receptor (K(i) = 1.37 nM) and delta-opioid receptor (K(i) = 3.24 nM) but almost no affinity for kappa-opioid receptor (at 1 microM). In the bioassay, the inhibitory effect of C8813 in the guinea-pig ileum (GPI) was 16.5 times more potent than in the mouse vas deferens (MVD). The inhibitory effects of C8813 in the GPI and MVD could be antagonized by mu-opioid receptor antagonist naloxone and delta-opioid receptor antagonist ICI174,864 respectively. However, the inhibitory effect of C8813 in the rabbit vas deferens was very weak. These results indicated that C8813 was a potent analgesic and a high affinity agonist for the mu- and delta-opioid receptors.
Collapse
Affiliation(s)
- Zhong-Hua Liu
- 2nd Department of Pharmacology, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China
| | | | | | | | | | | |
Collapse
|
20
|
Oakley SM, Toth G, Borsodi A, Kieffer BL, Kitchen I. G-protein coupling of delta-opioid receptors in brains of mu-opioid receptor knockout mice. Eur J Pharmacol 2003; 466:91-8. [PMID: 12679145 DOI: 10.1016/s0014-2999(03)01531-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mu-opioid receptor knockout mice have been reported to show loss of some delta-opioid receptor function. We hypothesised that this is due to some delta-opioid receptors being uncoupled from G-proteins in the absence of the mu-opioid receptor. To address this possibility, we have used quantitative autoradiography to determine the binding of three delta-opioid receptor agonist ligands ([3H]deltorphin I, [3H] [R-Atc(3), Ile(5,6)]deltorphin II, [3H] 4-[(alpaR)-alpha-((2S,5R)-4-propyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC-121)) and the delta-opioid receptor antagonist, [3H]naltrindole in the presence and absence of a GTP analogue, guanylylimidodiphosphate (GMPPNP) in the brains of mice lacking the mu-opioid receptor gene. Guanylylimidodiphosphate caused a decrease in the binding of the agonist ligands (to differing extents) and an increase in binding for the antagonist in wild-type controls. Overall, there were no major differences in the effects of guanylylimidodiphosphate for the agonist ligands in mu-knockout mice although a few structures showed a smaller sensitivity to the effects of this GTP analogue most notably for [3H]naltrindole. These findings suggest that in the majority of brain regions, G-protein coupling is unaltered in mu-opioid receptor knockout mice.
Collapse
MESH Headings
- Animals
- Autoradiography
- Binding Sites
- Brain/drug effects
- Brain/metabolism
- Female
- GTP-Binding Proteins/metabolism
- Guanylyl Imidodiphosphate/pharmacology
- Male
- Mice
- Mice, Knockout
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- Sarah M Oakley
- Pharmacology Group, School of Biomedical and Life Sciences, University of Surrey, Surrey GU2 7XH, Guildford, UK
| | | | | | | | | |
Collapse
|
21
|
Cichewicz DL, McCarthy EA. Antinociceptive synergy between delta(9)-tetrahydrocannabinol and opioids after oral administration. J Pharmacol Exp Ther 2003; 304:1010-5. [PMID: 12604676 DOI: 10.1124/jpet.102.045575] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The analgesic effects of opioids, such as morphine and codeine, in mice are enhanced by oral administration of the cannabinoid delta(9)-tetrahydrocannabinol (delta(9)-THC). However, isobolographic analysis has never been done to confirm a synergy between delta(9)-THC and morphine or codeine via oral routes of administration. To determine the nature of the interaction between these drugs for pain relief and extend previous experimental results, we performed an isobolographic analysis to evaluate for additivity or synergy in the tail-flick test. Fixed-ratio combinations of delta(9)-THC with either morphine or codeine were tested for antinociceptive effects. The experimentally derived ED(50) for each combination was compared with the theoretical additive ED(50), using an isobolographic analysis. All of the fixed-ratio combinations tested produced greater antinociception (synergy) than predicted from simple additivity. These findings suggest that the use of a low-dose combination of analgesics is a valid and effective approach for the treatment of pain and necessitates further study.
Collapse
Affiliation(s)
- Diana L Cichewicz
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia, USA.
| | | |
Collapse
|
22
|
Goody RJ, Oakley SM, Filliol D, Kieffer BL, Kitchen I. Quantitative autoradiographic mapping of opioid receptors in the brain of delta-opioid receptor gene knockout mice. Brain Res 2002; 945:9-19. [PMID: 12113946 DOI: 10.1016/s0006-8993(02)02452-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Using quantitative receptor autoradiography we have determined if deletion of the delta-opioid receptor gene (Oprd1) results in compensatory changes in the expression of other opioid receptors. Gene targeting was used to delete exon 1 of the mouse delta-opioid receptor gene and autoradiography was carried out on brains from wild-type, heterozygous and homozygous knockout mice. Delta-opioid receptors were labeled with [(3)H]deltorphin I (7 nM), mu- with [(3)H]DAMGO (4 nM), and kappa- with [(3)H]CI-977 (2.5 nM) or [(3)H]bremazocine (2 nM in the presence of DPDPE and DAMGO) and non-specific binding determined with naloxone. [(3)H]Deltorphin I binding was reduced by approximately 50% in heterozygous animals. In homozygous animals specific binding could only be detected after long-term film exposure (12 weeks). Regions exhibiting this residual [(3)H]deltorphin I binding correlated significantly with those demonstrating high levels of the mu-receptor and were abolished in the presence of the mu-agonist DAMGO. Autoradiographic mapping showed significant overall reductions in [(3)H]DAMGO and [(3)H]CI-977 binding throughout the brain following loss of both copies of the Oprd1 gene. In contrast, overall levels of [(3)H]bremazocine binding were higher in brains from -/- than +/+ mice. Our findings suggest that residual [(3)H]deltorphin I binding in the brain of delta-receptor gene knockout mice is the result of cross-reactivity with mu-sites and that there are no delta-receptor subtypes derived from a different gene. Changes in mu- and kappa-receptor labeling suggest compensatory changes in these subtypes in response to the absence of the delta-receptor. The differences in [(3)H]CI-977 and [(3)H]bremazocine binding indicate these ligands show differential recognition of the kappa-receptor.
Collapse
MESH Headings
- Animals
- Autoradiography
- Benzofurans/metabolism
- Benzomorphans/metabolism
- Brain/metabolism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Female
- Heterozygote
- Homozygote
- Male
- Mice
- Mice, Knockout/genetics
- Mice, Knockout/metabolism
- Oligopeptides/metabolism
- Pyrrolidines/metabolism
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Robin J Goody
- Pharmacology Group, School of Biomedical and Life Sciences, University of Surrey, Guilford, Surrey GU2 7XH, UK
| | | | | | | | | |
Collapse
|
23
|
Rady JJ, Portoghese PS, Fujimo JM. Methadone and heroin antinociception: predominant delta-opioid-receptor responses in methadone-tolerant mice. JAPANESE JOURNAL OF PHARMACOLOGY 2002; 88:319-31. [PMID: 11949888 DOI: 10.1254/jjp.88.319] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Antinociceptive tail flick responses to heroin and 6-monoacetylmorphine mediated in the brain by mu-opioid receptor are switched by morphine pellet implantation to delta1- and delta2-opioid-receptors mediation, respectively. Present results showed that the mu-receptor response (inhibited by beta-funaltrexamine) to methadone was changed by morphine pellet implantation to delta1 (inhibited by 7-benzylidenenaltrexone)- and delta2 (inhibited by naltriben)-opioid-receptor responses. Methadone pellet implantation likewise changed mediation from mu- to delta-opioid receptors for heroin and methadone but not for morphine (beta-funaltrexamine continued to inhibit). Methadone mu action in the brain was linked through a descending system to activate spinal serotonin receptors (inhibited by methysergide), but this link was gone in the methadone-pellet-implanted group. In the latter group, the new delta1- and delta2-receptor responses were mediated by spinal GABAA (inhibited by bicuculline) and GABAB (inhibited by 2-hydroxysaclofen) receptors. These shifts in neuronal systems meant that mu receptors on a given neuron were not changed into delta receptors. Preliminary results showed that delta-agonist action for methadone was prevented from appearing by MK801, a NMDA-receptor antagonist, and did not occur in 129S6/SvEv mice which lack NMDA responsiveness. Could methadone maintenance treatment in humans uncover delta-agonist actions?
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Drug Implants
- Drug Tolerance
- Heroin/pharmacology
- Injections, Intraventricular
- Methadone/administration & dosage
- Methadone/pharmacology
- Mice
- Receptors, N-Methyl-D-Aspartate/drug effects
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, mu/antagonists & inhibitors
- Species Specificity
Collapse
Affiliation(s)
- Jodie J Rady
- Research Service, VA Medical Center, Milwaukee, WI 53295, USA.
| | | | | |
Collapse
|
24
|
Alt A, Clark MJ, Woods JH, Traynor JR. Mu and Delta opioid receptors activate the same G proteins in human neuroblastoma SH-SY5Y cells. Br J Pharmacol 2002; 135:217-25. [PMID: 11786497 PMCID: PMC1573101 DOI: 10.1038/sj.bjp.0704430] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2001] [Accepted: 10/09/2001] [Indexed: 11/09/2022] Open
Abstract
1. There is evidence for interactions between mu and delta opioid systems both in vitro and in vivo. This work examines the hypothesis that interaction between these two receptors can occur intracellularly at the level of G protein in human neuroblastoma SH-SY5Y cells. 2. The [(35)S]GTP gamma S binding assay was used to measure G protein activation following agonist occupation of opioid receptors. The agonists DAMGO (EC(50), 45 nM) and SNC80 (EC(50), 32 nM) were found to be completely selective for stimulation of [(35)S]-GTP gamma S binding through mu and delta opioid receptors respectively. Maximal stimulation of [(35)S]-GTP gamma S binding produced by SNC80 was 57% of that seen with DAMGO. When combined with a maximally effective concentration of DAMGO, SNC80 caused no additional [(35)S]-GTP gamma S binding. This effect was also seen when measured at the level of adenylyl cyclase. 3. Receptor activation increased the dissociation of pre-bound [(35)S]-GTP gamma S. In addition, the delta agonist SNC80 promoted the dissociation of [(35)S]-GTP gamma S from G proteins initially labelled using the mu agonist DAMGO. Conversely, DAMGO promoted the dissociation of [(35)S]-GTP gamma S from G proteins initially labelled using SNC80. 4. Tolerance to DAMGO and SNC80 in membranes from cells exposed to agonist for 18 h was homologous and there was no evidence for alteration in G protein activity. 5. The findings support the hypothesis that mu- and delta-opioid receptors share a common G protein pool, possibly through a close organization of the two receptors and G protein at the plasma membrane.
Collapse
MESH Headings
- Adenylyl Cyclases/metabolism
- Analgesics, Opioid/pharmacology
- Benzamides/pharmacology
- Cyclic AMP/biosynthesis
- Dose-Response Relationship, Drug
- Drug Interactions
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, D-Penicillamine (2,5)-/metabolism
- GTP-Binding Proteins/drug effects
- GTP-Binding Proteins/metabolism
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- Humans
- Ligands
- Neuroblastoma
- Piperazines/pharmacology
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
- Sulfur Radioisotopes
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- A Alt
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, U.S.A
| | - M J Clark
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, U.S.A
| | - J H Woods
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, U.S.A
- Department of Psychology, University of Michigan, Ann Arbor, Michigan, U.S.A
| | - J R Traynor
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, U.S.A
| |
Collapse
|
25
|
Maldonado R, Severini C, Matthes HW, Kieffer BL, Melchiorri P, Negri L. Activity of mu- and delta-opioid agonists in vas deferens from mice deficient in MOR gene. Br J Pharmacol 2001. [PMID: 11264242 DOI: 10.1038/sj.bjp.0703966].] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Mice lacking the mu-opioid receptor have been recently generated. Centrally mediated responses of mu-opioid agonists are suppressed whereas some of the delta-opioid responses are preserved in these mutant mice. 2. The vas deferens bioassay has been used in this study to investigate the functional activity at a peripheral level of mu- and delta-opioid agonists in mice lacking mu-opioid receptors. 3. The different mu-opioid agonists evaluated, morphine, DAMGO, dermorphin and [Lys(7)]-dermorphin produced an inhibitory response in vas deferens from wild-type mice but had no relevant activity on vas deferens from mutant mice. 4. The selective delta-opioid agonists DPDPE, BUBU, deltorphin I, deltorphin II and [D-Met(2)]-deltorphin induced inhibitory effects in vas deferens from both wild-type and mutant mice. However, the biological activities of these ligands were slightly reduced in preparations from mutant mice. The inhibitory responses of all these delta-opioid agonists were prevented by the administration of the selective delta-opioid antagonist naltrindole. 5. These data indicate that delta-opioid agonists, but not mu-opioid agonists, are biologically active in vas deferens from mice lacking mu-opioid receptors. The decreased response of delta-agonists in mutant mice suggests that some cooperativity may exist between mu- and delta-opioid receptors in these vas deferens preparations.
Collapse
Affiliation(s)
- R Maldonado
- Laboratori de Neurofarmacologia, Facultat de Ciences de la Salut i de la Vida, Universitat Pompeu Fabra, c/Dr Aiguader 80, 08003 Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
26
|
Maldonado R, Severini C, Matthes HW, Kieffer BL, Melchiorri P, Negri L. Activity of mu- and delta-opioid agonists in vas deferens from mice deficient in MOR gene. Br J Pharmacol 2001; 132:1485-92. [PMID: 11264242 PMCID: PMC1572701 DOI: 10.1038/sj.bjp.0703966] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
1. Mice lacking the mu-opioid receptor have been recently generated. Centrally mediated responses of mu-opioid agonists are suppressed whereas some of the delta-opioid responses are preserved in these mutant mice. 2. The vas deferens bioassay has been used in this study to investigate the functional activity at a peripheral level of mu- and delta-opioid agonists in mice lacking mu-opioid receptors. 3. The different mu-opioid agonists evaluated, morphine, DAMGO, dermorphin and [Lys(7)]-dermorphin produced an inhibitory response in vas deferens from wild-type mice but had no relevant activity on vas deferens from mutant mice. 4. The selective delta-opioid agonists DPDPE, BUBU, deltorphin I, deltorphin II and [D-Met(2)]-deltorphin induced inhibitory effects in vas deferens from both wild-type and mutant mice. However, the biological activities of these ligands were slightly reduced in preparations from mutant mice. The inhibitory responses of all these delta-opioid agonists were prevented by the administration of the selective delta-opioid antagonist naltrindole. 5. These data indicate that delta-opioid agonists, but not mu-opioid agonists, are biologically active in vas deferens from mice lacking mu-opioid receptors. The decreased response of delta-agonists in mutant mice suggests that some cooperativity may exist between mu- and delta-opioid receptors in these vas deferens preparations.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Electric Stimulation
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Female
- Genotype
- In Vitro Techniques
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Mice, Mutant Strains
- Morphine/pharmacology
- Muscle Contraction/drug effects
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Oligopeptides/pharmacology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/genetics
- Vas Deferens/drug effects
- Vas Deferens/physiology
Collapse
Affiliation(s)
- R Maldonado
- Laboratori de Neurofarmacologia, Facultat de Ciences de la Salut i de la Vida, Universitat Pompeu Fabra, c/Dr Aiguader 80, 08003 Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
27
|
Hutcheson DM, Matthes HWD, Valjent E, Sanchez-Blazquez P, Rodriguez-Diaz M, Garzon J, Kieffer BL, Maldonado R. Lack of dependence and rewarding effects of deltorphin II in mu-opioid receptor-deficient mice. Eur J Neurosci 2001. [DOI: 10.1046/j.1460-9568.2001.01363.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
Hutcheson DM, Matthes HWD, Valjent E, Sánchez-Blázquez P, Rodríguez-Díaz M, Garzón J, Kieffer BL, Maldonado R. Lack of dependence and rewarding effects of deltorphin II in mu-opioid receptor-deficient mice. Eur J Neurosci 2001. [DOI: 10.1111/j.1460-9568.2001.01363.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Park Y, Ma T, Tanaka S, Jang C, Loh HH, Ko KH, Ho IK. Comparison of G-protein activation in the brain by mu-, delta-, and kappa-opioid receptor agonists in mu-opioid receptor knockout mice. Brain Res Bull 2000; 52:297-302. [PMID: 10856828 DOI: 10.1016/s0361-9230(00)00265-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Mice lacking the mu-opioid receptor gene have been developed by a gene knockout procedure. In this study, the activity of opioid receptor coupled G-proteins was examined to investigate whether there is a change in the extent of coupling for mu, delta-, and kappa-opioid receptors in mu-opioid receptor knockout mice. Selective agonists of mu- (DAMGO), delta- (DPDPE), and kappa- (U-69,593) opioid receptors stimulated [(35)S]GTPgammaS binding in the caudate putamen and cortex of wild-type mice. In contrast, only U-69,593 stimulated [(35)S]GTPgammaS binding in these regions of mu-opioid receptor knockout mice. These results confirmed the absence of G-protein activation by a mu-opioid receptor agonist in mu-opioid receptor knockout mice, and demonstrated that coupling of the kappa-opioid receptor to G-proteins is preserved in these mice. However, G-protein activation by the delta-opioid receptor agonist, DPDPE, was reduced in the mu-opioid receptor knockout mice, at least in the brain regions studied using autoradiography.
Collapse
MESH Headings
- Alleles
- Analgesics, Opioid/pharmacology
- Animals
- Autoradiography
- Benzeneacetamides
- Binding, Competitive/drug effects
- Blotting, Southern
- Brain/drug effects
- Brain/metabolism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- GTP-Binding Proteins/metabolism
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Pyrrolidines/pharmacology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/genetics
- Sulfur Radioisotopes
Collapse
Affiliation(s)
- Y Park
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 32916-4505, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Hosohata Y, Vanderah TW, Burkey TH, Ossipov MH, Kovelowski CJ, Sora I, Uhl GR, Zhang X, Rice KC, Roeske WR, Hruby VJ, Yamamura HI, Lai J, Porreca F. delta-Opioid receptor agonists produce antinociception and [35S]GTPgammaS binding in mu receptor knockout mice. Eur J Pharmacol 2000; 388:241-8. [PMID: 10675732 DOI: 10.1016/s0014-2999(99)00897-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We examined the effects of [D-Pen(2),D-Pen(5)]enkephalin (DPDPE), [D-Ala(2),Glu(4)]deltorphin (DELT), and (+)-4-[(alphaR)-alpha((2S, 5R)-4-Allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N, N-diethylbenzamide (SNC80) on [35S]GTPgammaS binding in brain membranes prepared from micro-opioid receptor knockout (-/-) mice. The potency and maximal response (E(max)) of these agonists were unchanged compared to control mice. In contrast, while the potency of [D-Pen(2),pCl-Phe(4),D-Pen(5)]enkephalin (pCl-DPDPE) was not significantly different, the E(max) was reduced as compared to controls. In the tail-flick test, intracerebroventricular (i.c.v.) or intrathecal (i.th.) DELT produced antinociceptive effects in -/- mice with potency that did not differ significantly from controls. In contrast, the antinociceptive potency of i.c.v. and i.th. DPDPE was displaced to the right by 4- and 9-fold in -/- compared to control mice, respectively. Reduced DPDPE antinociceptive potency in -/- mice, taken together with reduced DPDPE- and pCl-DPDPE- stimulated G protein activity in membranes prepared from -/- mice, demonstrate that these agonists require mu-opioid receptors for full activity. However, because DELT mediated G protein activation and antinociception were both comparable between -/- and wild type mice, we conclude that the mu-opioid receptor is not a critical component of delta-opioid receptor function.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Benzamides/pharmacology
- Brain/metabolism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Enkephalins/pharmacology
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- In Vitro Techniques
- Membranes
- Mice
- Mice, Knockout
- Oligopeptides/pharmacology
- Pain Measurement
- Piperazines/pharmacology
- Protein Binding
- Radioligand Assay
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/genetics
- Spinal Cord/metabolism
Collapse
Affiliation(s)
- Y Hosohata
- Department of Pharmacology, University of Arizona Heatth Sciences Center, Tucson, AZ, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Capasso A. Further studies on the involvement of the arachidonic acid cascade in the acute dependence produced by mu, kappa and delta opioid agonists in isolated tissues. Neuropharmacology 1999; 38:871-7. [PMID: 10465690 DOI: 10.1016/s0028-3908(99)00004-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The effects of phospholipase A2, cyclooxygenase-1, cyclooxygenase-2, and 5-lipoxygenase inhibitors on acute opiate withdrawal induced by selective mu, kappa and delta receptor agonists was investigated in vitro. After a 4 min in vitro exposure to D-Ala2-N-methyl-Phe-Gly5-ol)enkephalin (DAMGO; a highly selective mu agonist) and trans(+/-)-3,4-dichloro-N-methyl-N-(2(1pyrrolidynyl)-cyclohexyl)-+ ++benzeneacetamid (U50-488H; a highly selective K agonist) a strong contraction of the guinea pig isolated ileum was observed after the addition of naloxone. This effect was also observed when rabbit isolated jejunum was pretreated with deltorphin (a highly selective delta agonist). Mepacrine (a phospholipase A2 inhibitor), tolmetin (a selective cyclooxygenase-1 inhibitor) and meloxicam (a selective cyclooxygenase-2 inhibitor) treatment before or after DAMGO or U50-488H were able to both prevent and reverse the naloxone-induced contraction after exposure to the opioid agonists, in a concentration-dependent fashion. In addition, nordihydroguaiaretic acid (a 5-lipooxygenase inhibitor) was able to block the naloxone-induced contraction following exposure to DAMGO or U50-488H if injected either before or after the opioid agonist. In contrast, mepacrine, tolmetin, meloxicam and nordihydroguaiaretic acid did not affect the naloxone-induced contraction after exposure to deltorphin. The results of the present study confirm and extend a previous study performed with morphine indicating that arachidonic acid and its metabolites (prostaglandins and leukotrienes) are involved in the development of opioid withdrawal induced by selective mu and kappa opioid agonists whereas no effects were observed on withdrawal induced by the selective delta opioid agonist deltorphin.
Collapse
Affiliation(s)
- A Capasso
- Department of Pharmaceutical Sciences, University of Salerno, Italy.
| |
Collapse
|
32
|
Rady JJ, Gorny JM, Fujimoto JM. Heroin antinociception changed from mu to delta receptor in streptozotocin-treated mice. JAPANESE JOURNAL OF PHARMACOLOGY 1998; 78:443-54. [PMID: 9920201 DOI: 10.1254/jjp.78.443] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
CD-1 mice were treated intravenously with streptozotocin, 200 mg/kg, and tested 2 weeks later or treated with 60 mg/kg and tested 3 days later. Both treatments changed the tail flick response of heroin and 6-monoacetylmorphine (6 MAM) given intracerebroventricularly from a mu- to delta-opioid receptor-mediated action as determined by differential effects of opioid receptor antagonists. The response to morphine remained mu. Heroin and 6 MAM responses involved delta1 (inhibited by 7-benzylidenenaltrexone) and delta2 (inhibited by naltriben) receptors, respectively. These delta-agonist actions did not synergize with the mu-agonist action of morphine in the diabetic mice. The expected synergism between the delta agonist, [D-Pen2-D-Pen5]enkephalin (DPDPE), and morphine was not obtained in diabetic mice. Thus, diabetes disrupted the purported mu/delta-coupled response. In nondiabetic CD-1 mice, heroin and 6 MAM produced a different mu-receptor response (not inhibited by naloxonazine) from that of morphine (inhibited by naloxonazine). Also, these mu actions, unlike that of morphine, did not synergize with DPDPE. The unique receptor actions and changes produced by streptozotocin suggest that extrinsic in addition to genetic factors influence the opioid receptor selectivity of heroin and 6 MAM.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Anti-Bacterial Agents/pharmacology
- Benzylidene Compounds/pharmacology
- Diabetes Mellitus, Experimental/physiopathology
- Dose-Response Relationship, Drug
- Drug Interactions
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Heroin/pharmacology
- Injections, Intraventricular
- Male
- Mice
- Morphine/pharmacology
- Morphine Derivatives/pharmacology
- Naloxone/analogs & derivatives
- Naloxone/pharmacology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Nociceptors/drug effects
- Pain/drug therapy
- Pain/physiopathology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, mu/drug effects
- Streptozocin/pharmacology
- Time Factors
Collapse
Affiliation(s)
- J J Rady
- Research Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, USA
| | | | | |
Collapse
|
33
|
Activity of the delta-opioid receptor is partially reduced, whereas activity of the kappa-receptor is maintained in mice lacking the mu-receptor. J Neurosci 1998. [PMID: 9736649 DOI: 10.1523/jneurosci.18-18-07285.1998] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Previous pharmacological studies have indicated the possible existence of functional interactions between mu-, delta- and kappa-opioid receptors in the CNS. We have investigated this issue using a genetic approach. Here we describe in vitro and in vivo functional activity of delta- and kappa-opioid receptors in mice lacking the mu-opioid receptor (MOR). Measurements of agonist-induced [35S]GTPgammaS binding and adenylyl cyclase inhibition showed that functional coupling of delta- and kappa-receptors to G-proteins is preserved in the brain of mutant mice. In the mouse vas deferens bioassay, deltorphin II and cyclic[D-penicillamine2, D-penicillamine5] enkephalin exhibited similar potency to inhibit smooth muscle contraction in both wild-type and MOR -/- mice. delta-Analgesia induced by deltorphin II was slightly diminished in mutant mice, when the tail flick test was used. Deltorphin II strongly reduced the respiratory frequency in wild-type mice but not in MOR -/- mice. Analgesic and respiratory responses produced by the selective kappa-agonist U-50,488H were unchanged in MOR-deficient mice. In conclusion, the preservation of delta- and kappa-receptor signaling properties in mice lacking mu-receptors provides no evidence for opioid receptor cross-talk at the cellular level. Intact antinociceptive and respiratory responses to the kappa-agonist further suggest that the kappa-receptor mainly acts independently from the mu-receptor in vivo. Reduced delta-analgesia and the absence of delta-respiratory depression in MOR-deficient mice together indicate that functional interactions may take place between mu-receptors and central delta-receptors in specific neuronal pathways.
Collapse
|
34
|
Romita VV, Henry JL. Spinal mu-, delta- and kappa-opioid receptors mediate intense stimulation-elicited inhibition of a nociceptive reflex in the rat. Eur J Pharmacol 1998; 357:127-38. [PMID: 9797028 DOI: 10.1016/s0014-2999(98)00463-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Intense electrical stimulation of meridian points in the rat inhibits the nociceptive tail withdrawal reflex. The objective of the present study was to determine whether spinal opioid receptors mediate this inhibition. Electrical stimulation was applied with 2 ms square pulses, at 4 Hz for 20 min at 20 times the threshold, to previously defined meridian points in the hindlimb. Threshold was the minimum current required to elicit muscle twitch. In lightly anaesthetized intact rats (n = 8) stimulation inhibited tail withdrawal during and for greater than one hour after the end of stimulation. In unanaesthetized spinal rats (n = 12) this inhibition was less and the post-stimulation effect lasted for 15 min. In control anaesthetized intact (n = 28) and unanaesthetized spinal rats (n = 14) placement of electrodes without stimulation had no effect. In spinal rats, preadministration of naloxone (25 mg/kg, i.p.) blocked the evoked inhibition (n = 11). In intact animals both naloxone (n = 8) and the mu-opioid receptor antagonist, beta-funaltrexamine (10 nmol; n = 9), given via a chronic intrathecal catheter, attenuated inhibitions during and after the end of stimulation by 50-60%. The delta-opioid receptor antagonist H-Tyr-tic psi[CH2NH]Phe-Phe-OH (TIPP[psi]; 10 nmol; n = 7) and the kappa-opioid receptor antagonist nor-binaltorphimine (10 nmol; n = 13) given by lumbar puncture attenuated the inhibition during the stimulation by 30% and 56%, respectively; both antagonists blocked the post-stimulation effect and even facilitated the withdrawal. The data suggest that spinal mu-, delta- and kappa-opioid receptors each contribute to the evoked inhibition.
Collapse
Affiliation(s)
- V V Romita
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
35
|
Ilyina AD, Zaitsev SV, Karasev AA, Kurochkin IN, Grishina IA, Varfolomeyev SD. Kinetic behavior of a receptor-enzyme system: a model for drug addiction. Biosystems 1998; 45:67-76. [PMID: 9492956 DOI: 10.1016/s0303-2647(97)00063-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A theoretical kinetic model describing the behavior of a receptor-enzyme system during formation of drug addiction is considered in this article. The model assumes concomitant action of narcotic on at least two targets with opposite effects. Theoretical kinetic principles that the system must satisfy for the development of drug addiction are formulated. These kinetic principles are the slow inactivation of receptor-enzyme system and the divergence of characteristic times of dynamic concentration of product and enzyme.
Collapse
Affiliation(s)
- A D Ilyina
- Chemistry Department, Autonomous University of Coahuila, México
| | | | | | | | | | | |
Collapse
|
36
|
Stevens CW, Rothe KS. Supraspinal administration of opioids with selectivity for mu-, delta- and kappa-opioid receptors produces analgesia in amphibians. Eur J Pharmacol 1997; 331:15-21. [PMID: 9274924 PMCID: PMC3100726 DOI: 10.1016/s0014-2999(97)01026-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Previous results using an amphibian model showed that systemic and spinal administration of opioids selective for mu, delta and kappa-opioid receptors produce analgesia. It is not known whether non-mammalian vertebrates also contain supraspinal sites mediating opioid analgesia. Thus, opioid agonists selective for mu (morphine; fentanyl), delta (DADLE, [D-Ala2, D-Leu5]-enkephalin; DPDPE, [D-Pen2, D-Pen5]-enkephalin) and kappa (U50488, trans-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl] benzeneacetamide methanesulfonate; CI977, (5R)-(544alpha,744alpha,845beta)-N-methyl-N-[7-(1-p yrr olidinyl)-1-oxaspiro[4,5]dec-8yl]-4-benzofuranaceta mide++ + monohydrochloride) opioid receptors were tested for analgesia following i.c.v. administration in the Northern grass frog, Rana pipiens. Morphine, administered at 0.3, 1, 3 and 10 nmol/frog, produced a dose-dependent and long-lasting analgesic effect. Concurrent naltrexone (10 nmol) significantly blocked analgesia produced by i.c.v. morphine (10 nmol). ED50 values for the six opioids ranged from 2.0 for morphine to 63.9 nmol for U50488. The rank order of analgesic potency was morphine > DADLE > DPDPE > CI977 > fentanyl > U50488. These results show that supraspinal sites mediate opioid analgesia in amphibians and suggest that mechanisms of supraspinal opioid analgesia may be common to all vertebrates.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer
- Analgesics, Opioid/administration & dosage
- Animals
- Benzofurans/administration & dosage
- Dose-Response Relationship, Drug
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalin, Leucine-2-Alanine/administration & dosage
- Enkephalins/administration & dosage
- Female
- Fentanyl/administration & dosage
- Injections, Intraventricular
- Male
- Morphine/administration & dosage
- Morphine/antagonists & inhibitors
- Naltrexone/pharmacology
- Narcotics/administration & dosage
- Pyrrolidines/administration & dosage
- Rana pipiens
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, mu/drug effects
Collapse
Affiliation(s)
- C W Stevens
- Department of Pharmacology and Physiology, Oklahoma State University, College of Osteopathic Medicine, Tulsa 74107, USA.
| | | |
Collapse
|
37
|
Noble F, Fournie-Zaluski MC, Roques BP. Opposite role of delta 1- and delta 2-opioid receptors activated by endogenous or exogenous opioid agonists on the endogenous cholecystokinin system: further evidence for delta-opioid receptor heterogeneity. Neuroscience 1996; 75:917-26. [PMID: 8951884 DOI: 10.1016/0306-4522(96)00323-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Using the mouse caudate-putamen, where delta-opioid receptor subtypes have been shown to regulate adenylyl cyclase activity, we show in this study that endogenous enkephalins inhibit enzyme activity through activation of delta 1- and delta 2-opioid receptors. Thus, naltriben or 7-benzylidenenaltrexone as well as the delta-selective antagonist naltrindole (mixed delta 1 and delta 2 antagonist) antagonized inhibition of adenylyl cyclase activity induced by methionine- or leucine-enkephalin, while the micro-antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP) was without effect. Furthermore, we have previously shown that activation of delta-opioid receptors increases cholecystokinin release in the central nervous system, resulting in a potentiation of micro-opioid antinociceptive responses, and the respective role of delta 1- and delta 2-opioid receptors in this facilitatory effect has now been evaluated. Activation of delta 2-opioid receptors, either by endogenous enkephalins protected from catabolism by the complete enkephalin-degrading enzyme inhibitor N-((R,S)-2-benzyl-3((S)(2-amino-4-methyl-thio) butyldithio)-1-oxopropyl)-L-phenyl-alanine benzyl ester (RB 101), or by the delta 2-selective agonist Tyr-D-Ser(O-tert-butyl)-Gly-Phe-Leu-Thr(O-tert-butyl) (BUBU), potentiated micro-opioid antinociceptive responses in the hot-plate test in mice. This effect was antagonized by a selective cholecystokinin-A antagonist. Activation of delta 1-opioid receptors by endogenous opioid peptides decreased the micro-opioid responses. These results suggest that stimulation of delta 2-opioid receptors potentiates micro-opioid analgesia in the hot-plate test in mice through an increase in endogenous cholecystokinin release, while activation of delta 1-opioid receptors could decrease it. Thus, the pre-existing physiological balance between opioid and cholecystokinin systems seems to be modulated in opposite directions depending on whether delta 1- or delta 2-opioid receptors are selectively activated. This is the first demonstration that endogenous enkephalins, methionine- and leucine-enkephalin, are the natural ligands of delta-opioid receptor subtypes, and that delta 2-opioid receptor activation may facilitate the endogenous cholecystokinin-related modulation of micro-opioid analgesia, while the delta 1-opioid receptors may have an inhibitory role. These results could have important applications for the characterization of opioid delta 1 and delta 2 as subtypes or subsites and in pain alleviation.
Collapse
Affiliation(s)
- F Noble
- Department de Pharmacochimie Moléculaire et Structurale, INSERM U266, CNRS URA D 1500, Université René Descartes, UFR des Sciences Pharmaceutiques et Biologiques, Paris, France
| | | | | |
Collapse
|
38
|
Negus SS, Morgan D, Cook CD, Picker MJ. Effects of the delta opioid against BW373U86 in pigeons trained to discriminate fentanyl, bremazocine and water in a three-choice drug discrimination procedure. Psychopharmacology (Berl) 1996; 126:199-205. [PMID: 8876019 DOI: 10.1007/bf02246449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The delta opioid agonist BW373U86 was examined alone and in combination with mu agonists in pigeons trained to discriminate the mu agonist fentanyl (0.056 mg/kg), the kappa agonist bremazocine (0.017 mg/kg), and distilled water in a three-choice drug discrimination procedure. BW373U86 (0.01-10 mg/kg) produced a dose-dependent increase in fentanyl-appropriate responding and complete generalization to fentanyl in four of five subjects. BW373U86 did not elicit bremazocine-appropriate responding in any of the subjects. Fentanyl-appropriate responding elicited by BW373U86 was antagonized by the delta selective antagonist naltrindole (0.1-10 mg/kg) but not by the mu selective antagonist naloxone (0.1-30.0 mg/kg). When BW373U86 was administered in combination with the mu agonists fentanyl, morphine and nalbuphine, a low dose of BW373U86 (0.01 mg/kg) that elicited primarily water-appropriate responding when administered alone did not produce a significant change in the ED50 values for fentanyl, morphine or nalbuphine. Higher doses of BW373U86 (0.1-1.0 mg/kg) increased levels of fentanyl-appropriate responding elicited by low doses of fentanyl, morphine and nalbuphine to levels similar to those produced by BW373U86 alone. These results indicate that BW373U86 shares discriminative stimulus properties with the mu agonist fentanyl in pigeons, possibly by acting at delta opioid receptors. However, BW373U86 does not potentiate the discriminative stimulus effects of mu agonists or share discriminative stimulus effects with the kappa agonist bremazocine.
Collapse
Affiliation(s)
- S S Negus
- Alcohol and Drug Abuse Research Center, McLean Hospital-Harvard Medical School, Belmont, MA 01278, USA
| | | | | | | |
Collapse
|
39
|
Abstract
Opioids form the cornerstone of the pharmacologic armamentarium for the treatment of pain. Despite their long history of use, much confusion and misperception still surrounds their use. This short review will focus on pharmacodynamic and physiologic considerations in the clinical use of oral and parenteral opioids.
Collapse
Affiliation(s)
- F M Ferrante
- Cancer Pain and Symptom Management Program, Hospital of the University of Pennsylvania, Philadelphia 19104, USA
| |
Collapse
|
40
|
Meert TF. Pharmacotherapy of opioids: present and future developments. PHARMACY WORLD & SCIENCE : PWS 1996; 18:1-15. [PMID: 8861825 DOI: 10.1007/bf00449683] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The clinically available opioids have different physicochemical properties, resulting in differences in clinical profile with regard to potency, onset, and duration of activity. However, they all have comparable side-effects after acute systemic application. Several approaches can be used to overcome these side-effects. The following approaches, with special emphasis on the perioperative use of the opioids, are discussed: (1) the use of alternative routes of administration, such as via the spine (epidurally and intrathecally); (2) optimization of opioid delivery by means of slow-release preparations, chronic infusions with indwelling catheters, and transdermal delivery systems; (3) use of additional agents to potentiate the analgesic properties of the opioids so that the dose of opioid can be reduced; and (4) searching for new analgesics on the basis of knowledge of the pain-transmission system and the different opioid receptors with their functional interactions.
Collapse
Affiliation(s)
- T F Meert
- Department of Neuropsychopharmacology, Janssen Research Foundation, Beerse, Belgium
| |
Collapse
|
41
|
Mosser HC, Boucher RR, MacCreadie TM, Newman JR, Beckman AL. Morphine antinociception in the non-hibernating and hibernating states of the ground squirrel (Citellus lateralis). Life Sci 1995; 57:1441-9. [PMID: 7674835 DOI: 10.1016/0024-3205(95)02107-t] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Previous work in our laboratory demonstrated a significant reduction in the development of morphine physical dependence during hibernation, suggesting a major change in the ability of morphine to act on the central nervous system (CNS) during this naturally altered state. To further investigate the pharmacological actions of morphine during the hibernating (H) state, the present study recorded skin-twitch response (STR) latency as a measure of morphine antinociception in the golden-mantled ground squirrel (Citellus lateralis) during the non-hibernating (NH) and H states. Our results revealed that morphine antinociception continued to develop in hibernation. Moreover, the magnitude of antinociception displayed was greater during the H state than in the NH state. Tolerance to morphine's antinociceptive effects developed in both states as well. The results of the present study indicate that the hibernation-related reduction in the development of morphine dependence represents a selective, rather than a general, suppression of the CNS pharmacological actions of morphine during the H state.
Collapse
Affiliation(s)
- H C Mosser
- Department of Psychology, California State University, Long Beach 90840-0901, USA
| | | | | | | | | |
Collapse
|
42
|
Suzuki T, Tsuji M, Mori T, Misawa M, Endoh T, Nagase H. Effects of a highly selective nonpeptide delta opioid receptor agonist, TAN-67, on morphine-induced antinociception in mice. Life Sci 1995; 57:155-68. [PMID: 7603296 DOI: 10.1016/0024-3205(95)00256-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The effects of a potent and highly selective nonpeptide delta opioid receptor agonist, 2- methyl-4a alpha-(3-hydroxyphenyl)-1,2,3,4,4a,5,12,12a alpha- octahydroquinolino [2,3,3,-g] isoquinoline (TAN-67), on morphine-induced antinociception were examined using the warm-plate (51 degrees C) method. When a peptide delta 1 opioid receptor agonist, [D-Pen2, Pen5]enkephalin (DPDPE), was co-administered with i.c.v. morphine, low-dose morphine-induced antinociception was significantly increased. In contrast, i.c.v. co-administration of a peptide delta 2 opioid receptor agonist, [D-Ala2]deltorphin II (DELT), with morphine did not affect the morphine-induced antinociception. When morphine and TAN-67 were co-administered i.c.v., low-dose morphine-induced antinociception was significantly increased. Moreover, when TAN-67 and morphine were co-administered s.c., the morphine dose-response curve shifted to the left and the ED50 value of morphine decreased. These effects DPDPE and TAN-67 were antagonized by the delta opioid receptor antagonist naltrindole (NTI) and the delta 1 opioid receptor antagonist 7-benzylidenenaltrexone (BNTX) not by the delta 2 opioid receptor antagonist naltriben (NTB). Moreover, the mu opioid receptor antagonist beta-FNA also antagonized the effects of DPDPE and TAN-67. These results suggest that the effect of TAN-67 may result from the activation of central delta 1 opioid receptors, since the effect of TAN-67 was antagonized by NTI and BNTX, but not NTB. Furthermore, since pretreatment with beta-FNA also antagonized the effects of both DPDPE and TAN-67, a beta-FNA-sensitive site, i.e. a mu-delta complex site, may play an important role in the modulation of morphine-induced antinociception.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Drug Interactions
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Injections, Subcutaneous
- Male
- Mice
- Mice, Inbred Strains
- Morphine/pharmacology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Nociceptors/drug effects
- Oligopeptides/pharmacology
- Quinolines/pharmacology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/classification
Collapse
Affiliation(s)
- T Suzuki
- Department of Pharmacology, School of Pharmacy, Hoshi University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Shen KF, Crain SM. Specific N- or C-terminus modified dynorphin and beta-endorphin peptides can selectively block excitatory opioid receptor functions in sensory neurons and unmask potent inhibitory effects of opioid agonists. Brain Res 1995; 673:30-8. [PMID: 7757476 DOI: 10.1016/0006-8993(94)01380-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We recently showed that the opioid alkaloids, etorphine, dihydroetorphine and diprenorphine, have remarkably potent antagonist actions on excitatory opioid receptor functions in mouse sensory dorsal root ganglion (DRG) neurons. Pretreatment of naive nociceptive types of neurons with pM concentrations of these antagonists blocks excitatory prolongation of the calcium-dependent component of the action potential duration (APD) elicited by pM-nM morphine or other bimodally acting mu, delta and kappa opioid agonists and unmasks inhibitory APD shortening which usually requires much higher (ca. microM) concentrations. The present study demonstrates that pM concentrations of [des-Tyr1] fragments of dynorphin and beta-endorphin, as well as beta-endorphin-(1-27), can also selectively block excitatory opioid receptor functions in DRG neurons and unmask potent inhibitory effects of low concentrations of bimodally acting mu, delta and kappa opioid peptides and alkaloid agonists. These N- or C-terminus modified dynorphin or beta-endorphin peptides can be readily formed in neurons by specific peptidase activities. Since sustained activation of excitatory opioid receptor functions is essential for the development of tolerance/dependence in chronic morphine-treated DRG neurons in culture, the present in vitro study may help to account for the unexplained efficacy of [des-Tyr1] dynorphin fragments, as well as the endogenous opioids dynorphin A and beta-endorphin, in suppressing development and expression of naloxone-precipitated withdrawal and morphine tolerance in vivo.
Collapse
MESH Headings
- Action Potentials/drug effects
- Animals
- Cells, Cultured
- Dose-Response Relationship, Drug
- Dynorphins/chemistry
- Dynorphins/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Ganglia, Spinal/cytology
- Ganglia, Spinal/drug effects
- Mice
- Morphine/pharmacology
- Neurons/drug effects
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/drug effects
- beta-Endorphin/chemistry
- beta-Endorphin/pharmacology
Collapse
Affiliation(s)
- K F Shen
- Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA
| | | |
Collapse
|
44
|
Kontinen VK, Kalso EA. Differential modulation of alpha 2-adrenergic and mu-opioid spinal antinociception by neuropeptide FF. Peptides 1995; 16:973-7. [PMID: 7479344 DOI: 10.1016/0196-9781(95)00068-u] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Neuropeptide FF (NPFF) has been found to act as an antiopioid peptide. However, IT NPFF has recently been shown to potentiate the antinociceptive effects of IT morphine and to produce antinociception on its own. The aim of this study was to find out whether pretreatment with NPFF causes a comparable potentiation of dexmedetomidine-induced antinociception. NPFF (0.05-10.0 nmol) produced no antinociceptive effects in the rat tail flick test. NPFF potentiated the antinociceptive effect of IT morphine (7.8 nmol). This potentiation was prevented by IT naltrindole (28 nmol), which did not attenuate the antinociceptive effect of morphine. Dexmedetomidine (1.6-6.4 nmol) produced a dose-dependent antinociceptive effect, which was not potentiated by NPFF. Activation of the endogenous delta-opioid system due to the antiopioid effect of IT NPFF is proposed as an explanation to the reported differential action of NPFF on the mu-opioid and the alpha 2-adrenergic systems.
Collapse
MESH Headings
- Adrenergic alpha-Agonists/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Drug Synergism
- Imidazoles/pharmacology
- Male
- Medetomidine
- Morphine/pharmacology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Neuropeptides/pharmacology
- Oligopeptides/pharmacology
- Pain/physiopathology
- Rats
- Rats, Sprague-Dawley
- Receptors, Adrenergic, alpha-2/drug effects
- Receptors, Adrenergic, alpha-2/physiology
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/physiology
- Spinal Cord/drug effects
- Spinal Cord/physiology
- Time Factors
Collapse
Affiliation(s)
- V K Kontinen
- Department of Anesthesia, University of Helsinki, Finland
| | | |
Collapse
|
45
|
Capasso A, Di Giannuario A, Loizzo A, Pieretti S, Sorrentino L. Dexamethasone influence on morphine-induced analgesia in outbred Swiss and inbred DBA/2J and C57BL/6 mice. Prog Neuropsychopharmacol Biol Psychiatry 1994; 18:779-92. [PMID: 7938566 DOI: 10.1016/0278-5846(94)90084-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
1. The influence of dexamethasone on morphine analgesia in three different strains of mice (Swiss, DBA/2J and C57BL/6) was studied by using the tail flick test. 2. I.c.v. as well as i.p. injections of dexamethasone did not modify nociceptive response in all strains. 3. I.c.v. injection of dexamethasone significantly reduced morphine analgesia in Swiss mice whereas no effects were observed in DBA/2J and C57BL/6 mice. 4. In addition, i.p. injection of dexamethasone significantly reduced morphine analgesia in all three strains. 5. These results suggest that the use of different genetic strains may provide an useful approach for studying dexamethasone-morphine analgesia interaction.
Collapse
Affiliation(s)
- A Capasso
- Department of Experimental Pharmacology, University of Naples Federico II, Italy
| | | | | | | | | |
Collapse
|
46
|
Coderre TJ, Katz J, Vaccarino AL, Melzack R. Contribution of central neuroplasticity to pathological pain: review of clinical and experimental evidence. Pain 1993; 52:259-285. [PMID: 7681556 DOI: 10.1016/0304-3959(93)90161-h] [Citation(s) in RCA: 1296] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Peripheral tissue damage or nerve injury often leads to pathological pain processes, such as spontaneous pain, hyperalgesia and allodynia, that persist for years or decades after all possible tissue healing has occurred. Although peripheral neural mechanisms, such as nociceptor sensitization and neuroma formation, contribute to these pathological pain processes, recent evidence indicates that changes in central neural function may also play a significant role. In this review, we examine the clinical and experimental evidence which points to a contribution of central neural plasticity to the development of pathological pain. We also assess the physiological, biochemical, cellular and molecular mechanisms that underlie plasticity induced in the central nervous system (CNS) in response to noxious peripheral stimulation. Finally, we examine theories which have been proposed to explain how injury or noxious stimulation lead to alterations in CNS function which influence subsequent pain experience.
Collapse
Affiliation(s)
- Terence J Coderre
- Pain Mechanisms Laboratory, Clinical Research Institute of Montreal, MontrealCanada Département de Médecine, Université de Montréal, MontrealCanada Department of Psychology, McGill University, MontrealCanada Department of Psychology, Toronto General Hospital, TorontoCanada Departments of Behavioral Science and Anaesthesia, University of Toronto, TorontoCanada
| | | | | | | |
Collapse
|
47
|
Walker MJ, Le AD, Poulos CX. Effects of naltrindole and nor-binaltorphimine treatment on antinociception induced by sub-acute selective mu opioid receptor blockade. Brain Res 1992; 599:204-8. [PMID: 1337857 DOI: 10.1016/0006-8993(92)90392-m] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
When administered repeatedly, in conjunction with hot plate testing, naloxone and naltrexone have the paradoxical effect of producing antinociception in rats and mice. Recently, we have found that the sub-acute selective blockade of mu opioid receptors leads to the development of antinociception and an augmentation of kappa receptor-mediated antinociception. In this study, acute delta/kappa antagonist treatment produced a significant decrease in paw lick latency in rats displaying antinociception induced by sub-acute mu blockade, however, the response level of these animals was still significantly above the baseline. In addition, rats receiving sub-acute combined mu and delta antagonist treatment took longer to develop an antinociceptive response than those treated with a mu antagonist alone. Sub-acute selective blockade of kappa or delta opioid receptors had no overall effect on paw lick latency during the course of 5 days of hot plate testing. The results indicate that delta receptor activity may play a role in the antinociception induced by sub-acute mu blockade. However, while delta antagonist treatment effected the expression, it did not completely attenuate the antinociception induced by sub-acute mu blockade suggesting that there is still a significant non-opioid component to this analgesic response. The results of a final experiment, in which acute delta antagonist treatment had no effect on antinociception induced by repeated systemic injections of naloxone, supported this hypothesis.
Collapse
Affiliation(s)
- M J Walker
- Addiction Research Foundation, Toronto, Ont., Canada
| | | | | |
Collapse
|
48
|
Kamei J, Kawashima N, Kasuya Y. Paradoxical analgesia produced by naloxone in diabetic mice is attributable to supersensitivity of delta-opioid receptors. Brain Res 1992; 592:101-5. [PMID: 1450902 DOI: 10.1016/0006-8993(92)91663-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The effects of naloxone on the analgesic response were examined using the tail-flick test, in mice with streptozotocin-induced diabetes. Subcutaneous injection of naloxone (5 mg/kg, s.c.) produced a marked analgesia in diabetic mice but not in age-matched non-diabetic mice. Naloxone-induced analgesia in diabetic mice was significantly reduced by pretreatment with naltrindole (0.1 mg/kg, s.c.), a selective antagonist of delta-opioid receptors. By contrast, no significant naloxone-induced increase in tail-flick latency in diabetic mice was observed after chronic treatment with naloxone (5 mg/kg, s.c.) for 5 days. However, the tail-flick latency was significantly increased by chronic treatment with naloxone in non-diabetic mice. Furthermore, the significant naloxone-induced increase in tail-flick latency in non-diabetic mice that had been chronically treated with naloxone was also antagonized by pretreatment with naltrindole. Chronic pretreatment with 5 mg/kg of naloxone for 5 days markedly attenuated the analgesic effect of the delta-agonist DPDPE in diabetic mice, whereas this pretreatment significantly enhanced the effect of DPDPE in non-diabetic mice. These results suggest that naloxone-induced 'paradoxical' analgesia in mice may be mediated predominantly by delta-opioid receptors.
Collapse
Affiliation(s)
- J Kamei
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | | | | |
Collapse
|
49
|
Bakalkin Gya, Demuth HU, Nyberg F. Relationship between primary structure and activity in exorphins and endogenous opioid peptides. FEBS Lett 1992; 310:13-6. [PMID: 1526277 DOI: 10.1016/0014-5793(92)81135-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We have found a correlation between the certain characteristics of primary structure and biologic activity in exorphins and endogenous opioid peptide family. The characteristics of primary structure are the content of certain segment pairs as well as the density of their arrangement in a peptide. These segment pairs represent basic elements of the regulatory peptide primary structure pattern, which was found recently [Dokl. Akad. Nauk USSR 289 (1986) 721-724; Int. J. Peptide Prot. Res. 38 (1991) 505-510].
Collapse
Affiliation(s)
- Bakalkin Gya
- Department of Drug Dependence Research, Karolinska Institute, Stockholm, Sweden
| | | | | |
Collapse
|
50
|
Beckman AL, Llados-Eckman C, Stanton TL. Reduction of hibernation bout duration by intraventricular infusion of met-enkephalin. Brain Res 1992; 588:159-63. [PMID: 1393565 DOI: 10.1016/0006-8993(92)91357-k] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The potential of brain met-enkephalin (met-enk) systems to modulate central nervous system (CNS) activity during periods of general depression (modeled by the mammalian hibernation state) was studied in the ground squirrel (Citellus lateralis). Following entrance into hibernation, continuous met-enk infusion into the lateral ventricle (1 microliter/h; 0.2, 1 and 5 micrograms/microliter) produced a dose-dependent reduction in bout duration ranging from 1.2 to 3.9 days (13.8-44.6% of baseline bout duration). We suggest that the activity of met-enk-releasing neurons may serve to increase the excitability of the depressed CNS, thus accelerating the termination of the hibernation bout.
Collapse
Affiliation(s)
- A L Beckman
- Department of Psychology, California State University, Long Beach 90840-0901
| | | | | |
Collapse
|