1
|
Cox BM, Toll L. Contributions of the International Narcotics Research Conference to Opioid Research Over the Past 50 years. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10115. [PMID: 38390618 PMCID: PMC10880772 DOI: 10.3389/adar.2022.10115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/14/2022] [Indexed: 02/24/2024]
Abstract
The International Narcotics Research Conference (INRC), founded in 1969, has been a successful forum for research into the actions of opiates, with an annual conference since 1971. Every year, scientists from around the world have congregated to present the latest data on novel opiates, opiate receptors and endogenous ligands, mechanisms of analgesic activity and unwanted side effects, etc. All the important discoveries in the opiate field were discussed, often first, at the annual INRC meeting. With an apology to important events and participants not discussed, this review presents a short history of INRC with a discussion of groundbreaking discoveries in the opiate field and the researchers who presented from the first meeting up to the present.
Collapse
Affiliation(s)
- Brian M. Cox
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Lawrence Toll
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
2
|
Tang W, Luo L, Hu B, Zheng M. Butorphanol alleviates lipopolysaccharide-induced inflammation and apoptosis of cardiomyocytes via activation of the κ-opioid receptor. Exp Ther Med 2021; 22:1248. [PMID: 34539844 PMCID: PMC8438658 DOI: 10.3892/etm.2021.10683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/16/2021] [Indexed: 12/25/2022] Open
Abstract
Sepsis-induced myocardial dysfunction is a leading cause of the high mortality rates associated with sepsis. The aim of the present study was to investigate the effect of butorphanol on sepsis-induced cardiomyocyte dysfunction. Lipopolysaccharide (LPS) was used to induce H9C2 cardiomyocytes to establish an in vitro sepsis model. The effect of butorphanol on the viability of LPS-induced H9C2 cells was analyzed using a Cell Counting Kit-8 assay. The levels of tumor necrosis factor-α, interleukin (IL)-1β and IL-6 were detected using ELISA. Western blotting was used to analyze the expression levels of inflammation-and apoptosis-related proteins. Cell apoptosis was measured using a TUNEL assay. The expression levels of κ-opioid receptor (KOR) were analyzed using reverse transcription-quantitative PCR analysis and western blotting. Following LPS induction, the levels of inflammatory cytokines and proapoptotic proteins were found to be upregulated in H9C2 cells, while butorphanol treatment downregulated these levels. The expression levels of KOR were also upregulated following butorphanol treatment in LPS-induced H9C2 cells. Addition of the KOR inhibitor, nor-binaltorphimine, alleviated the inhibitory effects of butorphanol on inflammation and apoptosis in LPS-induced H9C2 cells. In conclusion, the findings of the present study provided evidence indicating that butorphanol may alleviate LPS-induced inflammation and apoptosis in cardiomyocytes by upregulating KOR expression, which may provide a novel insight into the potential therapeutic effects of butorphanol and its underlying mechanism of action.
Collapse
Affiliation(s)
- Weiqing Tang
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Liu Luo
- Department of Anesthesiology, The Affiliated Zhuzhou Hospital of Xiangya School of Medicine, Central South University, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Baoji Hu
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Mingzhi Zheng
- Department of Anesthesiology, The Affiliated Zhuzhou Hospital of Xiangya School of Medicine, Central South University, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| |
Collapse
|
3
|
Abstract
Oxycodone, a semisynthetic opioid analgesic, is widely used in clinical practice. Oxycodone and morphine seem to be equally effective and equipotent; however, morphine is 10 times more potent than oxycodone when given epidurally. This article provides an updated review of the basic pharmacology of oxycodone with a special focus on pharmacokinetic/pharmacodynamics properties. The controversy regarding oxycodone-mediated effects for visceral pain via agonism and the possible role of peripheral opioid analgesia are discussed in the present investigation in an attempt to propose a plausible explanation to the perplexing question of oxycodone analgesia.
Collapse
Affiliation(s)
- Xiulu Ruan
- Department of Anesthesiology, Louisiana State University Health Science Center, 1542 Tulane Avenue, New Orleans, LA 70112, USA.
| | - Ken F Mancuso
- Department of Anesthesiology, Louisiana State University Health Science Center, 1542 Tulane Avenue, New Orleans, LA 70112, USA
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Science Center, 1542 Tulane Avenue, New Orleans, LA 70112, USA
| |
Collapse
|
4
|
Characterization of [ 3 H] oxymorphone binding sites in mouse brain: Quantitative autoradiography in opioid receptor knockout mice. Neurosci Lett 2017; 643:16-21. [DOI: 10.1016/j.neulet.2017.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/01/2017] [Accepted: 02/01/2017] [Indexed: 11/19/2022]
|
5
|
Befort K. Interactions of the opioid and cannabinoid systems in reward: Insights from knockout studies. Front Pharmacol 2015; 6:6. [PMID: 25698968 PMCID: PMC4318341 DOI: 10.3389/fphar.2015.00006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/08/2015] [Indexed: 12/14/2022] Open
Abstract
The opioid system consists of three receptors, mu, delta, and kappa, which are activated by endogenous opioid peptides (enkephalins, endorphins, and dynorphins). The endogenous cannabinoid system comprises lipid neuromodulators (endocannabinoids), enzymes for their synthesis and their degradation and two well-characterized receptors, cannabinoid receptors CB1 and CB2. These systems play a major role in the control of pain as well as in mood regulation, reward processing and the development of addiction. Both opioid and cannabinoid receptors are coupled to G proteins and are expressed throughout the brain reinforcement circuitry. Extending classical pharmacology, research using genetically modified mice has provided important progress in the identification of the specific contribution of each component of these endogenous systems in vivo on reward process. This review will summarize available genetic tools and our present knowledge on the consequences of gene knockout on reinforced behaviors in both systems, with a focus on their potential interactions. A better understanding of opioid-cannabinoid interactions may provide novel strategies for therapies in addicted individuals.
Collapse
Affiliation(s)
- Katia Befort
- CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives - UMR7364, Faculté de Psychologie, Neuropôle de Strasbourg - Université de Strasbourg, Strasbourg France
| |
Collapse
|
6
|
Pennock RL, Hentges ST. Direct inhibition of hypothalamic proopiomelanocortin neurons by dynorphin A is mediated by the μ-opioid receptor. J Physiol 2014; 592:4247-56. [PMID: 25085890 DOI: 10.1113/jphysiol.2014.275339] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
It has recently been shown that dynorphin A (Dyn A), an endogenous agonist of the κ-opioid receptor (KOR), directly inhibits proopiomelanocortin (POMC) neurons in the hypothalamus through activation of G-protein-coupled inwardly rectifying K(+) channels (GIRKs). This effect has been proposed to be mediated by the putative κ2-opioid receptor (KOR-2), and has been suggested as a possible mechanism for the orexigenic actions of KOR agonists. Using whole-cell voltage clamp recordings in brain slice preparations, the present study demonstrates that Dyn A (1 or 5 μm) induces an outward current in POMC neurons that is reversed by the highly selective μ-opioid receptor (MOR) antagonist CTAP and is absent in mice lacking MORs. Additionally, the KOR-2-selective agonist GR89696 binds MORs on POMC neurons but fails to induce an outward current. Similar to Dyn A, the KOR-selective antagonist nor-binaltorphimine (nor-BNI) lacked specificity when used at sufficiently high concentrations. Maximal concentrations of the MOR-selective agonist DAMGO induced outward currents in POMC neurons that were completely reversed by a relatively high concentration of nor-BNI. Experiments using a half-maximal concentration of DAMGO demonstrate that nor-BNI must be used at concentrations <100 nm to avoid non-specific actions of the antagonist at MORs located on POMC neurons. These data suggest that direct inhibition of POMC neurons by Dyn A is mediated through the MOR, not the KOR-2, which is consistent with previous studies demonstrating that Dyn A can act at the μ-opioid receptor (MOR) when present in high concentrations.
Collapse
Affiliation(s)
- Reagan L Pennock
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
7
|
Yoo JH, Bailey A, Borsodi A, Tóth G, Matifas A, Kieffer BL, Kitchen I. Knockout subtraction autoradiography: a novel ex vivo method to detect heteromers finds sparse KOP receptor/DOP receptor heterodimerization in the brain. Eur J Pharmacol 2014; 731:1-7. [PMID: 24657279 DOI: 10.1016/j.ejphar.2014.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/06/2014] [Accepted: 03/10/2014] [Indexed: 10/25/2022]
Abstract
Several methodological approaches suggest that receptor heteromers exist in cell systems, but their presence in physiological tissue is widely contentious. We describe a novel method to determine if heterodimers exist in brain tissue sections using autoradiographic binding comparisons from single and double gene knockout mice, where tissues either have a full receptor complement and can form heterodimers, or are incapable of making heterodimers. We have tested this model, which we have named Knockout Subtraction Autoradiography, to determine if heterodimerisation of the kappa (KOP) and delta opioid (DOP) receptors occurs, as evidence from binding studies in cell systems suggest they are present in the brain. Using labeling of putative KOP receptor/DOP receptor heterodimers with either [(3)H]bremazocine or with [(3)H]naltrindole, two ligands which were used to provide evidence suggesting that these opioid receptor subtypes heterodimerize, we have applied a subtraction equation model based on the principle that receptor gene double knockout of either MOP receptor/KOP receptor (DOP receptor expression only) or MOP receptor/DOP receptor (KOP receptor expression only) produces tissue incapable of making the KOP receptor/DOP receptor heterodimer. We have shown in most brain regions that the labeling fits a simple additive model of monomer labeling, but that in a few brain regions opioid receptor heterodimerization does occur. The data does not support the conclusion that KOP receptor/DOP receptor heterodimerisation is widespread in the central nervous system, but does indicate that this novel methodology can detect heterodimerisation, when ligands with distinct binding affinities for monomer and heterodimer forms exist.
Collapse
Affiliation(s)
- Ji-Hoon Yoo
- Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | - Alexis Bailey
- Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | - Anna Borsodi
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvari krt 62, Szeged H-6726, Hungary
| | - Géza Tóth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvari krt 62, Szeged H-6726, Hungary
| | - Audrey Matifas
- IGBMC, CNRS/INSERM/ULP, 1 rue Laurent Fries, BP 10142, Illkirch, Cedex, France
| | - Brigitte L Kieffer
- IGBMC, CNRS/INSERM/ULP, 1 rue Laurent Fries, BP 10142, Illkirch, Cedex, France
| | - Ian Kitchen
- Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, UK.
| |
Collapse
|
8
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol 2013; 170:1459-581. [PMID: 24517644 PMCID: PMC3892287 DOI: 10.1111/bph.12445] [Citation(s) in RCA: 509] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. G protein-coupled receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
9
|
Involvement of the opioid and cannabinoid systems in pain control: new insights from knockout studies. Eur J Pharmacol 2013; 716:142-57. [PMID: 23523475 DOI: 10.1016/j.ejphar.2013.01.077] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/24/2013] [Accepted: 01/29/2013] [Indexed: 12/20/2022]
Abstract
The endogenous opioid and cannabinoid systems are involved in the physiological inhibitory control of pain and are of particular interest for the development of therapeutic approaches for pain management. The involvement of these endogenous systems in pain control has been studied from decades by the use of compounds with different affinities for each cannabinoid and opioid receptor or for the different enzymes involved in endocannabinoid and endogenous opioid metabolism. However, the selectivity of these pharmacological tools in vivo has represented an important limitation for these studies. The generation of genetically modified mice with selective mutations in specific components of the endocannabinoid and endogenous opioid system has provided important advances in the identification of the specific contribution of each component of these endogenous systems in the perception of noxious stimuli and the development of pathological pain states. Different lines of constitutive and conditional knockout mice deficient in specific cannabinoid and opioid receptors, specific precursors of the endogenous opioid peptides and the main enzymes involved in endocannabinoid and endogenous opioid degradation are now available. These knockout mice have also been used to evaluate the contribution of each component of the endocannabinoid and opioid system in the antinociceptive effects of cannabinoid and opioid agonists, including those currently used to treat pain in humans. This review summarizes the main advances provided in the last 15 years by the use of these genetic tools in the knowledge of the physiological control of pain and the pharmacology of cannabinoid and opioid compounds for pain management.
Collapse
|
10
|
Yoo JH, Kitchen I, Bailey A. The endogenous opioid system in cocaine addiction: what lessons have opioid peptide and receptor knockout mice taught us? Br J Pharmacol 2012; 166:1993-2014. [PMID: 22428846 DOI: 10.1111/j.1476-5381.2012.01952.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cocaine addiction has become a major concern in the UK as Britain tops the European 'league table' for cocaine abuse. Despite its devastating health and socio-economic consequences, no effective pharmacotherapy for treating cocaine addiction is available. Identifying neurochemical changes induced by repeated drug exposure is critical not only for understanding the transition from recreational drug use towards compulsive drug abuse but also for the development of novel targets for the treatment of the disease and especially for relapse prevention. This article focuses on the effects of chronic cocaine exposure and withdrawal on each of the endogenous opioid peptides and receptors in rodent models. In addition, we review the studies that utilized opioid peptide or receptor knockout mice in order to identify and/or clarify the role of different components of the opioid system in cocaine-addictive behaviours and in cocaine-induced alterations of brain neurochemistry. The review of these studies indicates a region-specific activation of the µ-opioid receptor system following chronic cocaine exposure, which may contribute towards the rewarding effect of the drug and possibly towards cocaine craving during withdrawal followed by relapse. Cocaine also causes a region-specific activation of the κ-opioid receptor/dynorphin system, which may antagonize the rewarding effect of the drug, and at the same time, contribute to the stress-inducing properties of the drug and the triggering of relapse. These conclusions have important implications for the development of effective pharmacotherapy for the treatment of cocaine addiction and the prevention of relapse.
Collapse
Affiliation(s)
- Ji Hoon Yoo
- Division of Biochemistry, Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | | | | |
Collapse
|
11
|
Karaji AG, Reiss D, Matifas A, Kieffer BL, Gavériaux-Ruff C. Influence of Endogenous Opioid Systems on T Lymphocytes as Assessed by the Knockout of Mu, Delta and Kappa Opioid Receptors. J Neuroimmune Pharmacol 2011; 6:608-16. [DOI: 10.1007/s11481-011-9314-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 09/08/2011] [Indexed: 01/24/2023]
|
12
|
Hutchinson MR, Shavit Y, Grace PM, Rice KC, Maier SF, Watkins LR. Exploring the neuroimmunopharmacology of opioids: an integrative review of mechanisms of central immune signaling and their implications for opioid analgesia. Pharmacol Rev 2011; 63:772-810. [PMID: 21752874 PMCID: PMC3141878 DOI: 10.1124/pr.110.004135] [Citation(s) in RCA: 291] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vastly stimulated by the discovery of opioid receptors in the early 1970s, preclinical and clinical research was directed at the study of stereoselective neuronal actions of opioids, especially those played in their crucial analgesic role. However, during the past decade, a new appreciation of the non-neuronal actions of opioids has emerged from preclinical research, with specific appreciation for the nonclassic and nonstereoselective sites of action. Opioid activity at Toll-like receptors, newly recognized innate immune pattern recognition receptors, adds substantially to this unfolding story. It is now apparent from molecular and rodent data that these newly identified signaling events significantly modify the pharmacodynamics of opioids by eliciting proinflammatory reactivity from glia, the immunocompetent cells of the central nervous system. These central immune signaling events, including the release of cytokines and chemokines and the associated disruption of glutamate homeostasis, cause elevated neuronal excitability, which subsequently decreases opioid analgesic efficacy and leads to heightened pain states. This review will examine the current preclinical literature of opioid-induced central immune signaling mediated by classic and nonclassic opioid receptors. A unification of the preclinical pharmacology, neuroscience, and immunology of opioids now provides new insights into common mechanisms of chronic pain, naive tolerance, analgesic tolerance, opioid-induced hyperalgesia, and allodynia. Novel pharmacological targets for future drug development are discussed in the hope that disease-modifying chronic pain treatments arising from the appreciation of opioid-induced central immune signaling may become practical.
Collapse
Affiliation(s)
- Mark R Hutchinson
- Discipline of Pharmacology, School of Medical Science, University of Adelaide, South Australia, Australia, 5005.
| | | | | | | | | | | |
Collapse
|
13
|
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
|
14
|
Dietis N, Rowbotham DJ, Lambert DG. Opioid receptor subtypes: fact or artifact? Br J Anaesth 2011; 107:8-18. [PMID: 21613279 DOI: 10.1093/bja/aer115] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
There is a vast amount of pharmacological evidence favouring the existence of multiple subtypes of opioid receptors. In addition to the primary classification of µ (mu: MOP), δ (delta: DOP), κ (kappa: KOP) receptors, and the nociceptin/orphanin FQ peptide receptor (NOP), various groups have further classified the pharmacological µ into µ(1-3), the δ into δ(1-2)/δ(complexed/non-complexed), and the κ into κ(1-3). From an anaesthetic perspective, the suggestions that µ(1) produced analgesia and µ(2) produced respiratory depression are particularly important. However, subsequent to the formal identification of the primary opioid receptors (MOP/DOP/KOP/NOP) by cloning and the use of this information to produce knockout animals, evidence for these additional subtypes is lacking. Indeed, knockout of a single gene (and hence receptor) results in a loss of all function associated with that receptor. In the case of MOP knockout, analgesia and respiratory depression is lost. This suggests that further sub-classification of the primary types is unwise. So how can the wealth of pharmacological data be reconciled with new molecular information? In addition to some simple misclassification (κ(3) is probably NOP), there are several possibilities which include: (i) alternate splicing of a common gene product, (ii) receptor dimerization, (iii) interaction of a common gene product with other receptors/signalling molecules, or (iv) a combination of (i)-(iii). Assigning variations in ligand activity (pharmacological subtypes) to one or more of these molecular suggestions represents an interesting challenge for future opioid research.
Collapse
Affiliation(s)
- N Dietis
- Department of Cardiovascular Sciences (Pharmacology and Therapeutics Group), Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Leicester Royal Infirmary, Leicester LE1 5WW, UK
| | | | | |
Collapse
|
15
|
Yoo JH, Bailey A, Ansonoff M, Pintar JE, Matifas A, Kieffer BL, Kitchen I. Lack of genotype effect on D1, D2 receptors and dopamine transporter binding in triple MOP-, DOP-, and KOP-opioid receptor knockout mice of three different genetic backgrounds. Synapse 2010; 64:520-7. [PMID: 20196137 DOI: 10.1002/syn.20757] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We investigated D1, D2 receptors and dopamine transporter (DAT) binding levels in mice lacking all three opioid receptors and wild-type (WT) mice on three different genetic backgrounds. Quantitative autoradiography was used to determine the level of radioligand binding to the D1 and D2 receptors and DAT labeled with [(3)H]SCH23390, [(3)H]raclopride, and [(3)H]mazindol, respectively in triple-opioid receptor knockout (KO) and WT maintained on C57BL/6 (B6) and 129/SvEvTac (129) as well as C57BL/6 x 129/SvPas (B6 x 129) strains. No significant genotype effect was observed in D1, D2 receptors and DAT binding in any regions analyzed in any of the strains studied, suggesting that a lack of all three opioid receptors does not influence D1, D2 receptors and DAT expression, irrespective of their genetic strain background. However, strain differences were observed in D1 binding between the three strains of mice studied. Lower levels of D1 binding were observed in the substantia nigra of B6 x 129 WT mice compared with the 129 WT mice and in the olfactory tubercle of B6 x 129 WT compared with B6 WT and 129 WT mice. Lower levels of D1 binding were observed in the caudate putamen of B6 x 129 KO mice compared with 129 KO mice. In contrast, no significant strain differences were observed in D2 and DAT binding between the three strains of mice in any regions analyzed. Overall, these results indicate a lack of modulation of the dopaminergic system by the deletion of all three opioid receptors regardless of different background strains.
Collapse
Affiliation(s)
- Ji-Hoon Yoo
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
16
|
Ansonoff MA, Portoghese PS, Pintar JE. Consequences of opioid receptor mutation on actions of univalent and bivalent kappa and delta ligands. Psychopharmacology (Berl) 2010; 210:161-8. [PMID: 20333506 DOI: 10.1007/s00213-010-1826-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 03/04/2010] [Indexed: 10/19/2022]
Abstract
INTRODUCTION During the past decade, substantial evidence has documented that opioid receptor heterodimers form in cell lines expressing one or more opioid receptors. More recent studies have begun to investigate whether heterodimer formation also occurs in vivo. OBJECTIVES We have used opioid receptor knockout mice to determine whether the in vivo intrathecal (i.t.) pharmacological potency of delta, kappa, and bivalent kappa/delta ligands is altered in the absence of the KOR-1 and/or DOR-1 genes. RESULTS We observe that both NorBNI (a kappa antagonist) and KDN-21 (a kappa/delta bivalent antagonist) specifically inhibit DPDPE but not deltorphin II i.t potency in wild-type mice but that following mutation of KOR-1, the ability of either compound to reduce DPDPE potency is lost. In contrast, knockout of KOR-1 unexpectedly slightly reduces the potency of deltorphin II (delta2) but not DPDPE (delta1). Finally, two compounds with kappa agonist activity, 6'-GNTI (a putative kappa/delta heterodimer selective agonist) and KDAN-18 (kappa agonist/delta antagonist bivalent ligand) show reduced potency in DOR-1 KO mice. CONCLUSIONS These results show, genetically, that bivalent ligands with kappa agonist activity require delta receptors for maximal potency in vivo, which is consistent with the presence of opioid heterodimer/oligomer complexes in vivo, and also highlight the complexity of delta drug action even when complementary pharmacologic and genetic approaches are used.
Collapse
Affiliation(s)
- Michael A Ansonoff
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | | | | |
Collapse
|
17
|
Yekkirala AS, Kalyuzhny AE, Portoghese PS. Standard opioid agonists activate heteromeric opioid receptors: evidence for morphine and [d-Ala(2)-MePhe(4)-Glyol(5)]enkephalin as selective μ-δ agonists. ACS Chem Neurosci 2010; 1:146-54. [PMID: 22816017 DOI: 10.1021/cn9000236] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 11/16/2009] [Indexed: 12/15/2022] Open
Abstract
Research in the opioid field has relied heavily on the use of standard agonist ligands such as morphine, [d-Ala(2)-MePhe(4)-Glyol(5)]enkephalin (DAMGO), U69593, bremazocine, [d-Pen(2)d-Pen(5)]enkephalin (DPDPE), and deltorphin-II as tools for investigating the three major types of opioid receptors, MOP (μ), KOP (κ), and DOP (δ), that mediate antinociception. The functional selectivity of these ligands has been based on the assumption that opioid receptors exist as homomers. As numerous studies in cultured cells have suggested that opioid receptors can associate both as homomers and heteromers, we have investigated the selectivity of these standard ligands using intracellular calcium release and [(35)S]GTPγS assays in HEK-293 cells that contain singly and coexpressed opioid receptors. The present study reveals that morphine and DAMGO, traditionally classified as μ selective agonists, selectively activate μ-δ heteromeric opioid receptors with greater efficacy than homomeric opioid receptors. Moreover, standard ligands that have been widely employed as κ- and δ-selective agonists display little or no differences in the activation of homomeric and heteromeric opioid receptors. The far-reaching implications of these results are discussed.
Collapse
Affiliation(s)
| | | | - Philip S. Portoghese
- Department of Pharmacology
- Department of Neuroscience, School of Medicine
- Department of Medicinal Chemistry, College of Pharmacy
| |
Collapse
|
18
|
Zhang S, Yekkirala A, Tang Y, Portoghese PS. A bivalent ligand (KMN-21) antagonist for mu/kappa heterodimeric opioid receptors. Bioorg Med Chem Lett 2009; 19:6978-80. [PMID: 19892550 PMCID: PMC5060004 DOI: 10.1016/j.bmcl.2009.10.045] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 10/08/2009] [Accepted: 10/09/2009] [Indexed: 11/15/2022]
Abstract
In an effort to develop antagonists for kappa-mu opioid receptor heterodimers, a series of bivalent ligands 3-6 containing kappa- and mu-antagonist pharmacophores were designed and synthesized. Evaluation of the series in HEK-293 cells revealed 4 (KMN-21) to selectively antagonize the activation of kappa-mu heterodimers, suggesting possible bridging of receptors when the bivalent ligand spacer contains 21 atoms.
Collapse
Affiliation(s)
- Shijun Zhang
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ajay Yekkirala
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ye Tang
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Philip S. Portoghese
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
19
|
|
20
|
Opioid and opioid-like. Br J Pharmacol 2009. [DOI: 10.1111/j.1476-5381.2009.00501_49.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
21
|
Abstract
BACKGROUND Opioid therapy is the standard treatment for moderate-to-severe cancer pain and is becoming a more frequent treatment for moderate-to-severe chronic noncancer pain. Response to opioids varies significantly between patients and even within the individual patient at different stages of treatment. Finding an opioid at a dose that provides adequate long-term analgesia with minimal adverse effects can be difficult. Opioid switching and opioid rotation, at different stages of therapy, represent two clinical strategies used to optimize opioid response for patients with moderate-to-severe pain. OBJECTIVES Review the theoretical and clinical evidence supporting the concepts of opioid switching and rotation, outline the conditions under which these practices should be considered, and briefly suggest practical steps for their implementation. SCOPE Clinical literature, clinical practice and guideline databases, and professional society websites were searched for articles or reports describing opioid switching or opioid rotation in chronic pain therapy; variability in patient response to opioid therapy; physiologic, pharmacologic, and genetic factors that affect clinical response to opioids; and practical approaches to maximizing analgesia and minimizing adverse effects in opioid therapy. It is outside the scope of this review to evaluate the pharmacoeconomic aspects that affect changes in opioid therapy. FINDINGS The variability in de novo clinical response to opioids likely represents the interaction of the varying properties of the individual opioids with the variability in individual patient biology. This interaction forms the rationale for opioid switching and explains its clinical utility. As with opioid switching, success with opioid rotation is related to the myriad of factors determining an individual patient's response to a specific opioid. However, the benefits of opioid rotation also derive from a partial reversal of tolerance at the mu-opioid receptor and the response of different micro-opioid receptor subtypes to the different opioids.
Collapse
Affiliation(s)
- Neal E Slatkin
- California Cancer Specialists Medical Group, Pasadena, CA 91105, USA.
| |
Collapse
|
22
|
Chefer VI, Shippenberg TS. Augmentation of morphine-induced sensitization but reduction in morphine tolerance and reward in delta-opioid receptor knockout mice. Neuropsychopharmacology 2009; 34:887-98. [PMID: 18704097 PMCID: PMC2639630 DOI: 10.1038/npp.2008.128] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Studies in experimental animals have shown that individuals exhibiting enhanced sensitivity to the locomotor-activating and rewarding properties of drugs of abuse are at increased risk for the development of compulsive drug-seeking behavior. The purpose of the present study was to assess the effect of constitutive deletion of delta-opioid receptors (DOPr) on the rewarding properties of morphine as well as on the development of sensitization and tolerance to the locomotor-activating effects of morphine. Locomotor activity testing revealed that mice lacking DOPr exhibit an augmentation of context-dependent sensitization following repeated, alternate injections of morphine (20 mg/kg; s.c.; 5 days). In contrast, the development of tolerance to the locomotor-activating effects of morphine following chronic morphine administration (morphine pellet: 25 mg: 3 days) is reduced relative to WT mice. The conditioned rewarding effects of morphine were reduced significantly in DOPrKO mice as compared to WT controls. Similar findings were obtained in response to pharmacological inactivation of DOPr in WT mice, indicating that observed effects are not due to developmental adaptations that occur as a consequence of constitutive deletion of DOPr. Together, these findings indicate that the endogenous DOPr system is recruited in response to both repeated and chronic morphine administration and that this recruitment serves an essential function in the development of tolerance, behavioral sensitization, and the conditioning of opiate reward. Importantly, they demonstrate that DOPr has a distinct role in the development of each of these drug-induced adaptations. The anti-rewarding and tolerance-reducing properties of DOPr antagonists may offer new opportunities for the treatment and prevention of opioid dependence as well as for the development of effective analgesics with reduced abuse liability.
Collapse
Affiliation(s)
- V I Chefer
- Integrative Neuroscience Section, Behavioral Neuroscience Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA.
| | | |
Collapse
|
23
|
Gonzalez-Nuñez V, Toth G, Rodríguez RE. Endogenous heptapeptide Met-enkephalin-Gly-Tyr binds differentially to duplicate delta opioid receptors from zebrafish. Peptides 2007; 28:2340-7. [PMID: 18022288 DOI: 10.1016/j.peptides.2007.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Revised: 10/04/2007] [Accepted: 10/04/2007] [Indexed: 11/28/2022]
Abstract
Met-enkephalin-Gly-Tyr (MEGY) is an endogenous peptide that binds to opioid sites in zebrafish and in rat brain homogenates. The aim of this work is to characterize the binding profile of this opioid ligand on two duplicate delta receptors from zebrafish, ZFOR1 and ZFOR4. Our results show that, while ZFOR1 presents one single binding site for [(3)H]-MEGY (K(D)=4.0+/-0.4 nM), the experimental data from ZFOR4 fit better to the two-site binding model (K(D1)=0.8+/-0.2 nM and K(D2)=30.2+/-10.2 nM). Two other MEGY synthetic analogues, (D-Ala(2))-MEGY and (D-Ala(2), Val(5))-MEGY were also prepared and tested, together with the original peptide MEGY and other opioid ligands, in competition binding assays. While these peptides presented K(i) values on the nanomolar range when using [(3)H]-MEGY as radioligand, these parameters were two orders higher in competition binding assays with the antagonist [(3)H]-diprenorphine. Functional [(35)S]GTPgammaS stimulation analysis has revealed that these two receptors can be activated by several opioid agonists. Our results prove that although the MEGY peptide acts as an agonist on ZFOR1 and ZFOR4, there are subtle pharmacological differences between these two delta opioid receptors from zebrafish.
Collapse
Affiliation(s)
- Veronica Gonzalez-Nuñez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Salamanca, Spain
| | | | | |
Collapse
|
24
|
Nielsen CK, Ross FB, Lotfipour S, Saini KS, Edwards SR, Smith MT. Oxycodone and morphine have distinctly different pharmacological profiles: Radioligand binding and behavioural studies in two rat models of neuropathic pain. Pain 2007; 132:289-300. [PMID: 17467904 DOI: 10.1016/j.pain.2007.03.022] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 02/18/2007] [Accepted: 03/19/2007] [Indexed: 11/23/2022]
Abstract
Previously, we reported that oxycodone is a putative kappa-opioid agonist based on studies where intracerebroventricular (i.c.v.) pre-treatment of rats with the kappa-selective opioid antagonist, nor-binaltorphimine (nor-BNI), abolished i.c.v. oxycodone but not morphine antinociception, whereas pretreatment with i.c.v. naloxonazine (mu-selective antagonist) produced the opposite effects. In the present study, we used behavioural experiments in rat models of mechanical and biochemical nerve injury together with radioligand binding to further examine the pharmacology of oxycodone. Following chronic constriction injury (CCI) of the sciatic nerve in rats, the antinociceptive effects of intrathecal (i.t.) oxycodone, but not i.t. morphine, were abolished by nor-BNI. Marked differences were found in the antinociceptive properties of oxycodone and morphine in streptozotocin (STZ)-diabetic rats. While the antinociceptive efficacy of morphine was abolished at 12 and 24 weeks post-STZ administration, the antinociceptive efficacy of s.c. oxycodone was maintained over 24 weeks, albeit with an approximately 3- to 4-fold decrease in potency. In rat brain membranes irreversibly depleted of mu- and delta-opioid binding sites, oxycodone displaced [(3)H]bremazocine (kappa(2)-selective in depleted membranes) binding with relatively high affinity whereas the selective mu- and delta-opioid ligands, CTOP (D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2)) and DPDPE ([D-Pen(2,5)]-enkephalin), respectively, did not. In depleted brain membranes, the kappa(2b)-ligand, leu-enkephalin, prevented oxycodone's displacement of high-affinity [(3)H]bremazocine binding, suggesting the notion that oxycodone is a kappa(2b)-opioid ligand. Collectively, the present findings provide further support for the notion that oxycodone and morphine produce antinociception through distinctly different opioid receptor populations. Oxycodone appears to act as a kappa(2b)-opioid agonist with a relatively low affinity for mu-opioid receptors.
Collapse
Affiliation(s)
- Carsten K Nielsen
- School of Pharmacy, The University of Queensland, St. Lucia Campus, St. Lucia, Brisbane, Qld 4072, Australia
| | | | | | | | | | | |
Collapse
|
25
|
Lunzer MM, Portoghese PS. Selectivity of delta- and kappa-opioid ligands depends on the route of central administration in mice. J Pharmacol Exp Ther 2007; 322:166-71. [PMID: 17400888 DOI: 10.1124/jpet.107.120279] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The existence of heterodimeric opioid receptors has introduced greater complexity to the in vivo characterization of pharmacological selectivity of agonists by antagonists. Because of the possibility of cooperativity between receptors organized as heterodimers, it is conceivable that selective antagonists may antagonize an agonist bound to a neighboring, allosterically coupled receptor. As a consequence, the in vivo selectivity of an opioid antagonist may depend on the organizational state of receptors that mediate analgesia. In this regard, phenotypic delta- and kappa-opioid receptors have been proposed to arise from different organizational states that include oligomeric delta-kappa heterodimers and homomeric delta and kappa receptors. In view of the evidence for analgesia mediated by delta-kappa heterodimers in the spinal cord, but not the brain, we have investigated the selectivity of pharmacologically selective delta and kappa antagonists in mice by both i.t. and i.c.v. routes of administration to evaluate changes in selectivity. Using pharmacologically selective delta (benzylidenenaltrexone, naltrindole, and naltriben) and kappa (norbinaltorphimine) antagonists versus delta ([D-Pen(2),D-Pen(5)]-enkephalin and deltorphin II) and kappa [3,4-dichloro-N-methyl-N-[(1R,2R)-2-(1-pyrrolidinyl)cyclohexyl]-benzeneacetamide (U50488) and bremazocine] agonists, the delta-1/delta-2 selectivity ratios were found to be dependent on the route of administration (i.t. versus i.c.v.). The data from different routes of administration suggest that differences in molecular recognition between spinal delta-kappa heterodimers and supraspinal homomeric delta and kappa receptors may contribute to the divergent selectivity ratios of selective antagonists. In view of the observed tissue-dependent selectivity, we suggest that multiple opioid antagonists be employed routinely in establishing agonist selectivity in vivo.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage
- Animals
- Benzylidene Compounds/administration & dosage
- Enkephalin, D-Penicillamine (2,5)-/administration & dosage
- Injections, Intraventricular
- Injections, Spinal
- Ligands
- Male
- Mice
- Mice, Inbred ICR
- Naltrexone/administration & dosage
- Naltrexone/analogs & derivatives
- Narcotic Antagonists/administration & dosage
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, kappa/antagonists & inhibitors
Collapse
Affiliation(s)
- Mary M Lunzer
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard St. S.E., Minneapolis, MN 55455, USA
| | | |
Collapse
|
26
|
Abstract
Opioids are the most effective and widely used drugs in the treatment of severe pain. They act through G protein-coupled receptors. Four families of endogenous ligands (opioid peptides) are known. The standard exogenous opioid analgesic is morphine. Opioid agonists can activate central and peripheral opioid receptors. Three classes of opioid receptors (mu, delta, kappa) have been identified. Multiple pathways ofopioid receptor signaling (e.g., G(i/o) coupling, cAMP inhibition, Ca++ channel inhibition) have been described. The differential regulation of effectors, preclinical pharmacology, clinical applications, and side effects will be reviewed in this chapter.
Collapse
Affiliation(s)
- C Zöllner
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | | |
Collapse
|
27
|
Bailey A, Gianotti R, Ho A, Kreek MJ. Downregulation of κ-opioid receptors in basolateral amygdala and septum of rats withdrawn for 14 days from an escalating dose “binge” cocaine administration paradigm. Synapse 2007; 61:820-6. [PMID: 17621646 DOI: 10.1002/syn.20436] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
There is evidence showing that the opioid systems play an important role in cocaine addiction; fewer studies have examined their roles in cocaine withdrawal. This study was conducted to determine whether cocaine or chronic withdrawal from cocaine alters the receptor component of the kappa-opioid system. Male Fischer rats were injected with saline or cocaine (3x15 mg/kg/day for 4 days, 3x20 mg/kg/day for 4 days, 3x25 mg/kg/day for 4 days, and 3x30 mg/kg/day for 2 days), three times daily at 1-h intervals in an escalating dose paradigm for 14 days. Identically treated rats were withdrawn from cocaine or saline for 14 days. We performed quantitative autoradiographic mapping of kappa-opioid receptors (KOP-r) in the brains of rats treated with this escalating dose "binge" cocaine administration paradigm and of rats withdrawn from cocaine for 14 days. A significant condition (chronic/withdrawal) effect was shown across all regions analyzed. A significant increase in [3H]CI-977 binding to KOP-r was detected in the septum of rats treated with an escalating dose binge cocaine administration paradigm and killed 30 min after the last cocaine injection. In contrast, there was a decrease in KOP-r binding in the septum and the basolateral amygdala of rats withdrawn for 14 days from chronic escalating dose binge cocaine administration, compared to rats at the end of 14 days chronic escalating dose cocaine administration. These results reconfirm and extend that KOP-r undergoes upregulation in response to chronic binge cocaine administration here, with an escalating dose. The observed lowering in KOP-r binding, which was shown in two brain regions of cocaine withdrawn animals, might contribute to the persistent dysphoria reported a long time after the discontinuation of the drug.
Collapse
Affiliation(s)
- Alexis Bailey
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York 10021, USA.
| | | | | | | |
Collapse
|
28
|
|
29
|
González-Núñez V, Barrallo A, Traynor JR, Rodríguez RE. Characterization of opioid-binding sites in zebrafish brain. J Pharmacol Exp Ther 2006; 316:900-4. [PMID: 16207834 DOI: 10.1124/jpet.105.093492] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The pharmacological profile of opioid-binding sites in zebrafish brain homogenates has been studied using radiolabeled binding techniques. The nonselective antagonist [(3)H]diprenorphine binds with high affinity (K(D) = 0.27 +/- 0.08 nM and a B(max) = 212 +/- 14.3 fmol/mg protein), displaying two different binding sites with affinities of K(D1) = 0.08 +/- 0.02 nM and K(D2) = 17.8 +/- 9.18 nM. The nonselective agonist [(3)H]bremazocine also binds with high affinity to zebrafish brain membranes but only displays one single binding site with a K(D) = 1.1 +/- 0.09 nM and a B(max) = 705 +/- 19.3 fmol/mg protein. Competition binding assays using [(3)H]diprenorphine and several unlabeled ligands were performed. The synthetic selective agonists for mammalian opioid receptors DPDPE ([DPen(2),D-Pen(5)]-enkephalin), DAMGO ([D-Ala(2),NMe-Phe(4),Gly(5)-ol]-enkephalin), and U69,593 [(5alpha,7alpha,8beta)-(+)-N-methyl-N-[7-(1-pyrrolidinyl)-1-oxaspiro[4.5]dec-8-yl]-benzeneacetamide] failed to effectively displace [(3)H]diprenorphine binding, whereas nonselective ligands and the endogenous opioid peptides such as dynorphin A showed good affinities in the nanomolar range, although several of the endogenous peptides only displaced approximately 50% of the specifically bound [(3)H]diprenorphine. Our results provide evidence that, although the selective synthetic compounds for mammalian receptors do not fully recognize the opioid-binding sites in zebrafish brain, the activity of the endogenous zebrafish opioid system might not significantly differ from that displayed by the mammalian opioid system. Hence, the study of zebrafish opioid activity may contribute to an understanding of endogenous opioid systems in higher vertebrates.
Collapse
Affiliation(s)
- Verónica González-Núñez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Salamanca, Spain
| | | | | | | |
Collapse
|
30
|
Gackenheimer SL, Suter TM, Pintar JE, Quimby SJ, Wheeler WJ, Mitch CH, Gehlert DR, Statnick MA. Localization of opioid receptor antagonist [3H]-LY255582 binding sites in mouse brain: comparison with the distribution of mu, delta and kappa binding sites. Neuropeptides 2005; 39:559-67. [PMID: 16289278 DOI: 10.1016/j.npep.2005.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Accepted: 09/24/2005] [Indexed: 11/16/2022]
Abstract
Agonist stimulation of opioid receptors increases feeding in rodents, while opioid antagonists inhibit food intake. The pan-opioid antagonist, LY255582, produces a sustained reduction in food intake and body weight in rodent models of obesity. However, the specific receptor subtype(s) responsible for this activity is unknown. To better characterize the pharmacology of LY255582, we examined the binding of a radiolabeled version of the molecule, [(3)H]-LY255582, in mouse brain using autoradiography. In mouse brain homogenates, the K(d) and B(max) for [(3)H]-LY255582 were 0.156 +/- 0.07 nM and 249 +/- 14 fmol/mg protein, respectively. [(3)H]-LY255582 bound to slide mounted sections of mouse brain with high affinity and low non-specific binding. High levels of binding were seen in areas consistent with the known localization of opioid receptors. These areas included the caudate putamen, nucleus accumbens, claustrum, medial habenula, dorsal endopiriform nucleus, basolateral nucleus of the amygdala, hypothalamus, thalamus and ventral tegmental area. We compared the binding distribution of [(3)H]-LY255582 to the opioid receptor antagonist radioligands [(3)H]-naloxone (mu), [(3)H]-naltrindole (delta) and [(3)H]-norBNI (kappa). The overall distribution of [(3)H]-LY255582 binding sites was similar to that of the other ligands. No specific [(3)H]-LY255582 binding was noted in sections of mu-, delta- and kappa-receptor combinatorial knockout mice. Therefore, it is likely that LY255582 produces its effects on feeding and body weight gain through a combination of mu-, delta- and kappa-receptor activity.
Collapse
MESH Headings
- Animals
- Autoradiography
- Binding Sites
- Brain/anatomy & histology
- Brain/metabolism
- Cyclohexanes/chemistry
- Cyclohexanes/metabolism
- Mice
- Mice, Knockout
- Molecular Structure
- Naloxone/metabolism
- Naltrexone/analogs & derivatives
- Naltrexone/metabolism
- Narcotic Antagonists/metabolism
- Piperidines/chemistry
- Piperidines/metabolism
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Tritium/chemistry
- Tritium/metabolism
Collapse
Affiliation(s)
- S L Gackenheimer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Commiskey S, Fan LW, Ho IK, Rockhold RW. Butorphanol: effects of a prototypical agonist-antagonist analgesic on kappa-opioid receptors. J Pharmacol Sci 2005; 98:109-16. [PMID: 15942128 DOI: 10.1254/jphs.crj05001x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The opioid analgesic, butorphanol (17-cyclobutylmethyl-3,14-dihydroxymorphinan) tartrate is a prototypical agonist-antagonist opioid analgesic agent whose potential for abuse has been the cause of litigation in the United States. With a published affinity for opioid receptors in vitro of 1:4:25 (mu:delta:kappa), the relative contribution of actions at each of these receptors to the in vivo actions of the drug are an issue of active investigation. A body of evidence has been developed which indicates that a substantial selective action of butorphanol on the kappa-opioid receptor mediates the development of tolerance to butorphanol and cross-tolerance to other opioid agonists; to the production of dependence upon butorphanol, particularly in the rodent; and to compensatory alterations in brain opioid receptor-effector systems. This perspective will identify the current state of understanding of the effects produced by butorphanol on brain opioid receptors, particularly on the kappa-opioid receptor subtype, and on the expression of phosphotyrosyl proteins following chronic treatment with butorphanol.
Collapse
Affiliation(s)
- Stephen Commiskey
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, USA
| | | | | | | |
Collapse
|
32
|
Wang D, Sun X, Bohn LM, Sadée W. Opioid receptor homo- and heterodimerization in living cells by quantitative bioluminescence resonance energy transfer. Mol Pharmacol 2005; 67:2173-84. [PMID: 15778451 DOI: 10.1124/mol.104.010272] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Opioid receptors have been shown to dimerize or oligomerize among themselves and each other, affecting their functional properties. This study used bioluminescence resonance energy transfer (BRET) between the mu, delta, and kappa opioid receptors to study opioid receptor aggregation in transfected human embryonic kidney 293 cells. Titration of receptor levels indicated that all three opioid receptors have a similar affinity to form homo- or hetero-oligomers in combination with any other opioid receptor type. In contrast, none of the opioid receptors formed detectable oligomers with the muscarinic M2 receptor, indicating that interactions among opioid receptors are selective. The formation of opioid receptor dimers, rather than higher order oligomers, is supported by binding kinetics in competition experiments between labeled and unlabeled receptors. Opioid receptor dimerization occurred at physiological temperatures upon receptor biosynthesis, before trafficking to the plasma membrane. Moreover, using BRET, coimmunoprecipitation, receptor binding, and G protein coupling, we demonstrate for the first time functional mu opioid receptor-kappa opioid receptor heterodimerization. These combined results demonstrate that opioid receptors can undergo homo- and heterodimerization, a process with potential implications for opioid physiology and pharmacology.
Collapse
Affiliation(s)
- Danxin Wang
- Department of Pharmacology, School of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
33
|
Daniels DJ, Kulkarni A, Xie Z, Bhushan RG, Portoghese PS. A bivalent ligand (KDAN-18) containing delta-antagonist and kappa-agonist pharmacophores bridges delta2 and kappa1 opioid receptor phenotypes. J Med Chem 2005; 48:1713-6. [PMID: 15771416 DOI: 10.1021/jm034234f] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To characterize delta- and kappa-opioid receptor phenotypes, bivalent ligands (KDAN series) containing delta-antagonist (naltrindole) and kappa(1)-agonist (ICI-199,441) pharmacophores were synthesized and evaluated by the intrathecal route using the mouse tail-flick assay and binding studies. The data have suggested that KDAN-18 (2) bridges phenotypic delta(2)- and kappa(1)-receptors. A conceptual model is presented to explain the organizational differences between the opioid receptors that give rise to the phenotypes (delta(1), delta(2), kappa(1), kappa(2)).
Collapse
Affiliation(s)
- David J Daniels
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
34
|
Abstract
Natural and synthetic opioid compounds, either alone or in combination with other drugs, are widely used analgesics for patients with both acute and chronic pain. Decades of extensive pharmacologic investigations have characterized three high-affinity cell-surface neuronal receptors, the activation of which is responsible for both the desirable properties (antinociception) and undesirable properties (respiratory depression, nausea and vomiting, dependence, etc.) of opioid drugs. Recent research in molecular biology and pharmacogenetics in relation to opioids and their receptors has helped clarify previous pharmacologic observations and has laid the groundwork for new analgesic therapies with improved therapeutic outcomes.
Collapse
Affiliation(s)
- Geoffrey K Gourlay
- Pain Management Unit, Department of Anaesthesia and Pain Management, Flinders Medical Centre, The Flinders University of South Australia, 5042, Bedford Park, South Australia, Australia.
| |
Collapse
|
35
|
Bhushan RG, Sharma SK, Xie Z, Daniels DJ, Portoghese PS. A bivalent ligand (KDN-21) reveals spinal delta and kappa opioid receptors are organized as heterodimers that give rise to delta(1) and kappa(2) phenotypes. Selective targeting of delta-kappa heterodimers. J Med Chem 2004; 47:2969-72. [PMID: 15163177 DOI: 10.1021/jm0342358] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In view of recent pharmacological studies suggesting the existence of delta-kappa opioid receptor heterodimers/oligomers in the spinal cord, we have synthesized and evaluated (intrathecally in mice) a series of bivalent ligands (KDN series) containing kappa and delta antagonist pharmacophores. Pharmacological and binding data have provided evidence for the bridging of spinal delta-kappa receptor heterodimers by KDN-21 and for their identification as delta(1) and kappa(2). The selectivity profile of KDN-21 and the apparent absence of coupled delta(1)-kappa(2) phenotypes in the brain suggest a new approach for targeting receptors.
Collapse
Affiliation(s)
- Rashmi G Bhushan
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
36
|
Scherrer G, Befort K, Contet C, Becker J, Matifas A, Kieffer BL. The delta agonists DPDPE and deltorphin II recruit predominantly mu receptors to produce thermal analgesia: a parallel study of mu, delta and combinatorial opioid receptor knockout mice. Eur J Neurosci 2004; 19:2239-48. [PMID: 15090050 DOI: 10.1111/j.0953-816x.2004.03339.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Delta-selective agonists have been developed to produce potent analgesic compounds with limited side-effects. DPDPE and deltorphin II are considered prototypes, but their delta-selectivity in vivo and the true ability of delta receptors to produce analgesia remain to be demonstrated. Here we have performed a parallel analysis of mu, delta and combinatorial opioid receptor knockout mice, in which we found no obvious alteration of G-protein coupling for remaining opioid receptors. We compared behavioural responses in two models of acute thermal pain following DPDPE and deltorphin II administration by intracerebroventricular route. In the tail-immersion test, both compounds were fully analgesic in delta knockout mice and totally inactive in mu knockout mice. In the hotplate test, the two compounds again produced full analgesia in delta knockout mice. In mu knockout mice, there was significant, although much lower, analgesia. Furthermore, DPDPE analgesia in the delta knockout mice was fully reversed by the mu selective antagonist CTOP in both tests. Together, this suggests that mu rather than delta receptors are recruited by the two agonists for the tail withdrawal and the hotplate responses. Finally, deltorphin II slightly prolonged jump latencies in double mu/kappa knockout mice (delta receptors only) and this response was abolished in the triple knockout mice, demonstrating that the activation of delta receptors alone can produce weak but significant mu-independent thermal antinociception.
Collapse
MESH Headings
- Analgesia/methods
- Animals
- Dose-Response Relationship, Drug
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Female
- Hot Temperature
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oligopeptides/pharmacology
- Pain Measurement/drug effects
- Pain Measurement/methods
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/deficiency
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/deficiency
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- Grégory Scherrer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, UMR7104, Parc d'Innovation, 1 rue Laurent Fries BP 10142, 67404 Illkirch Cedex, Strasbourg, France
| | | | | | | | | | | |
Collapse
|
37
|
Gouardères C, Kieffer BL, Zajac JM. Opposite alterations of NPFF1 and NPFF2 neuropeptide FF receptor density in the triple MOR/DOR/KOR-opioid receptor knockout mouse brains. J Chem Neuroanat 2004; 27:119-28. [PMID: 15121216 DOI: 10.1016/j.jchemneu.2004.01.002] [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] [Received: 08/27/2003] [Revised: 12/01/2003] [Accepted: 01/21/2004] [Indexed: 11/23/2022]
Abstract
Mice lacking the mu-delta-kappa-opioid receptor (MOR/DOR/KOR) genes and their corresponding wild-type littermates have been used to quantify NPFF(1) and NPFF(2) (neuropeptide FF) receptors by in vitro autoradiography in the central nervous tissues. Adjacent coronal sections were labelled with [125I]YVP ([125I]YVPNLPQRF-NH(2)) and [125I]EYF ([125I]EYWSLAAPQRF-NH(2)) as specific radioligands for NPFF(1) and NPFF(2) receptors, respectively. NPFF(2) receptors are predominantly expressed in both genotypes, but their density increases significantly in non cortical regions of mutant mice: 64% in the amygdaloid area, 89, 308, 1214 and 49% in the nucleus of the vertical limb of the diagonal band, substantia nigra, the vestibular nucleus and the spinal cord, respectively. In contrast, the density of the NPFF(1) subtype is lower than NPFF(2) in both genotypes and significantly decreased in some brain areas of mutant mice: -99, -90 and -90% in the nucleus of the vertical limb of the diagonal band, substantia nigra and the spinal cord, respectively. This study shows that mice lacking opioid receptors have brain region-dependent increases (NPFF(2)) and decreases (NPFF(1)) in NPFF receptors densities and suggests a different functional participation of each NPFF receptor subtype in the actions of opioids.
Collapse
MESH Headings
- Animals
- Autoradiography
- Brain/anatomy & histology
- Brain/metabolism
- Mice
- Mice, Knockout
- Receptors, Neuropeptide/metabolism
- Receptors, Opioid/deficiency
- Receptors, Opioid/genetics
- Receptors, Opioid, delta/deficiency
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, kappa/deficiency
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- Christine Gouardères
- Institut de Pharmacologie et de Biologie Structurale, C.N.R.S. UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | | | | |
Collapse
|
38
|
Contet C, Matifas A, Kieffer BL. No evidence for G-protein-coupled epsilon receptor in the brain of triple opioid receptor knockout mouse. Eur J Pharmacol 2004; 492:131-6. [PMID: 15178356 DOI: 10.1016/j.ejphar.2004.03.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2003] [Revised: 03/17/2004] [Accepted: 03/30/2004] [Indexed: 11/21/2022]
Abstract
Pharmacological approaches have defined the epsilon receptor as a beta-endorphin-preferring opioid receptor, described in rat vas deferens and in brain of several species. Only three opioid receptors-mu, delta and kappa-have been cloned and the existence of this additional subtype as a distinct protein remains controversial. Recently, the mouse brain epsilon receptor was detected in a G protein activation assay, as mediating residual beta-endorphin activity following pharmacological blockade of mu, delta and kappa receptors. To clarify whether this site is independent from mu, delta and kappa receptors, we performed beta-endorphin-induced [(35)S]GTPgammaS binding using mice lacking these three receptors (triple knockout mice). We tested both pons-medulla and whole brain preparations. beta-Endorphin strongly stimulated [(35)S]GTPgammaS binding in wild-type membranes but had no detectable effect in membranes from triple knockout mice. We conclude that the brain epsilon site involves mu, delta and/or kappa receptors, possibly coupled to nonclassical G proteins.
Collapse
MESH Headings
- Animals
- Binding, Competitive/physiology
- Brain/metabolism
- Female
- GTP-Binding Proteins/agonists
- Humans
- In Vitro Techniques
- Male
- Medulla Oblongata/metabolism
- Mice
- Mice, Knockout
- Pons/metabolism
- Receptors, Opioid/metabolism
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/genetics
- beta-Endorphin/antagonists & inhibitors
- beta-Endorphin/metabolism
Collapse
Affiliation(s)
- Candice Contet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP Parc d'innovation, 1 rue Laurent Fries BP 10142, C.U. de Strasbourg, 67404 Illkirch Cedex, France
| | | | | |
Collapse
|
39
|
Todtenkopf MS, Marcus JF, Portoghese PS, Carlezon WA. Effects of kappa-opioid receptor ligands on intracranial self-stimulation in rats. Psychopharmacology (Berl) 2004; 172:463-70. [PMID: 14727002 DOI: 10.1007/s00213-003-1680-y] [Citation(s) in RCA: 224] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2003] [Accepted: 11/20/2003] [Indexed: 11/27/2022]
Abstract
RATIONALE Elevations in cAMP response element binding protein (CREB) function within the mesolimbic system of rats reduce cocaine reward in place conditioning studies and increase immobility in the forced swim test. Each of these behavioral adaptations can be interpreted as a depressive-like effect (i.e., anhedonia, despair) that may reflect reduced activity of brain reward systems. Furthermore, each effect appears due to increases in CREB-mediated expression of dynorphin, since each is attenuated by intracranial injections of the kappa-opioid receptor antagonist norBNI. OBJECTIVES Intracranial self-stimulation (ICSS) studies were conducted in rats to determine whether administration of a kappa-agonist would have depressive-like effects on brain stimulation reward, and whether pretreatment with a kappa-antagonist would attenuate any such effects. Conditions that have depressive effects in people (e.g., drug withdrawal) increase the threshold amounts of stimulation required to sustain ICSS in rats. METHODS Sprague-Dawley rats with lateral hypothalamic stimulating electrodes were tested in a "curve-shift" variant of the ICSS procedure after systemic administration of the kappa-agonist U-69593 alone, the novel kappa-antagonist 5'-acetamidinoethylnaltrindole (ANTI) alone, or co-administration of both drugs. RESULTS U-69593 dose dependently increased ICSS thresholds, suggesting that activation of kappa-receptors reduced the rewarding impact of the brain stimulation. ANTI had no effects on its own, but it attenuated increases in ICSS thresholds caused by the agonist. CONCLUSIONS These data provide further evidence that stimulation of brain kappa-receptors may trigger certain depressive-like signs, and that kappa antagonists may have efficacy as antidepressants without having reward-related actions of their own.
Collapse
Affiliation(s)
- Mark S Todtenkopf
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, 115 Mill Street, MRC 217, Belmont, MA 02478, USA
| | | | | | | |
Collapse
|
40
|
Butelman ER, Ball JW, Kreek MJ. Peripheral selectivity and apparent efficacy of dynorphins: comparison to non-peptidic kappa-opioid agonists in rhesus monkeys. Psychoneuroendocrinology 2004; 29:307-26. [PMID: 14644063 DOI: 10.1016/s0306-4530(03)00030-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The potency and effectiveness of dynorphin A(1-17), E-2078 (a synthetic dynorphin A(1-8) analog) and non-peptidic kappa-opioid agonists were studied in rhesus monkeys in two assays: 1) a drug discrimination assay with the centrally-penetrating kappa-agonist U69,593 as the training stimulus (n=3) and 2) a prolactin release assay; a neuroendocrine effect thought to be mediated by kappa-receptors located in hypothalamic nuclei outside the blood-brain barrier. The non-peptidic kappa-agonists, U69,593 and bremazocine (0.00032-0.01 mg/kg, s.c.) were dose-dependently generalized by all the subjects trained to discriminate U69,593. U69,593 and bremazocine also caused dose-dependent prolactin release (n=4). By contrast, dynorphin A(1-17) and E-2078 (0.1-1 mg/kg, i.v.) were only generalized by one of the U69,593 discriminating subjects. However, both these dynorphins produced potent and robust prolactin release (0.0032-0.032 mg/kg, i.v.), when tested under an identical time course design as above. Naltrexone (0.1 or 0.32 mg/kg), caused a parallel rightward shift in the dose-effect curves for all the above ligands, consistent with kappa-receptor mediation of this neuroendocrine effect. The peripherally selective antagonist, quaternary naltrexone (0.32 mg/kg, s.c.) partially blocked the neuroendocrine effects of U69,593 and E-2078 (0.0032 mg/kg, s.c. and i.v., respectively). Overall, these findings are consistent with the hypothesis that the dynorphins act as high efficacy, peripherally selective kappa-agonists following systemic administration in primates.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, Rockefeller University (Box 171), 1230 York Avenue, New York, NY 10021, USA.
| | | | | |
Collapse
|
41
|
Minowa S, Ishihara S, Tsuchiya S, Horie S, Watanabe K, Murayama T. Involvement of glutamate and gamma-amino-butyric acid receptor systems on gastric acid secretion induced by activation of kappa-opioid receptors in the central nervous system in rats. Br J Pharmacol 2003; 138:1049-58. [PMID: 12684260 PMCID: PMC1573744 DOI: 10.1038/sj.bjp.0705082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
1. Various neurotransmitters in the brain regulate gastric acid secretion. Previously, we reported that the central injection of kappa-opioid receptor agonists stimulated this secretion in rats. Although the existence of kappa(1)-kappa(3)-opioid receptor subtypes has been proposed, the character is not defined. We investigated the interactions between kappa-opioid receptor subtypes and glutamate, gamma-amino-butyric acid (GABA) or 5-hydroxy tryptamine (5-HT) receptors in the rat brain. 2. Gastric acid secretion induced by the injection of U69593 (8.41 nmol, a putative kappa(1)-opioid receptor agonist) into the lateral cerebroventricle was completely inhibited by the central injection of 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX, 10.9 nmol, an antagonist for non-N-methyl-D-aspartate (non-NMDA) receptors) and by bicuculline infusion (222 micro g kg(-1) per 10 min, i.v., GABA(A) receptor antagonist). The secretion induced by bremazocine (8.52 nmol, a putative kappa(2)-opioid receptor agonist) was inhibited by bicuculline infusion, but not by CNQX. The secretion induced by naloxone benzoylhydrazone (224 nmol, a putative kappa(3)-opioid receptor agonist) was slightly and partially inhibited by CNQX and bicuculline. 3. Treatment with CNQX and bicuculline inhibited gastric acid secretion induced by the injection of dynorphin A-(1-17) into the lateral, but not the fourth, cerebroventricle. Antagonists for NMDA, GABA(B) and 5-HT(2/1C) receptors did not inhibit the secretions by kappa-opioid receptor agonists. 4. In rat brain regions close to the lateral cerebroventricle, kappa-opioid receptor systems (kappa(1)>kappa(3)>>kappa(2)) are regulated by the non-NMDA type of glutamate receptor system, and kappa(1)- and kappa(2)-opioid receptor systems are regulated by the GABA(A) receptor system. The present findings show pharmacological evidence for kappa-opioid receptor subtypes that regulate gastric acid secretion in the rat brain.
Collapse
MESH Headings
- 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology
- Animals
- Baclofen/analogs & derivatives
- Baclofen/pharmacology
- Benzeneacetamides/pharmacology
- Benzomorphans/pharmacology
- Bicuculline/pharmacology
- Brain/physiology
- Dynorphins/pharmacology
- Gastric Acid/metabolism
- Gastric Acid/physiology
- Injections, Intraventricular
- Ketanserin/pharmacology
- Male
- Perfusion/methods
- Piperazines/pharmacology
- Pyrrolidines/pharmacology
- Rats
- Rats, Wistar
- Receptors, GABA-A
- Receptors, Glutamate/drug effects
- Receptors, Kainic Acid/drug effects
- Receptors, N-Methyl-D-Aspartate
- Receptors, Opioid, kappa/administration & dosage
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/physiology
- Receptors, Serotonin
- Stomach/drug effects
- Stomach/physiopathology
- gamma-Aminobutyric Acid
Collapse
Affiliation(s)
- Sachie Minowa
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan
| | - Satomi Ishihara
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan
| | - Shizuko Tsuchiya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan
| | - Syunji Horie
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan
| | - Kazuo Watanabe
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan
- Author for correspondence:
| |
Collapse
|
42
|
Martin M, Matifas A, Maldonado R, Kieffer BL. Acute antinociceptive responses in single and combinatorial opioid receptor knockout mice: distinct mu, delta and kappa tones. Eur J Neurosci 2003; 17:701-8. [PMID: 12603260 DOI: 10.1046/j.1460-9568.2003.02482.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have examined responses of mice lacking mu, delta and kappa opioid receptor (MOR, DOR and KOR, respectively) genes, as well as combinatorial mutants, in several pain models. This is the first truly comparative study of all three opioid receptor-deficient mice, with genotypes and gender analysis using mice on the hybrid 50% 129/SV : 50% C57BL/6 genetic background. In the tail-immersion test, only KOR-/- females showed decreased withdrawal latencies. This modification was also found in MOR/KOR and MOR/DOR/KOR, but not MOR/DOR mutants. The hotplate test revealed increased nociceptive sensitivity for MOR-/-, a phenotype which was also observed in double mutants involving the MOR deletion, and in the triple mutants. The tail-pressure test showed increased response for both MOR-/- and DOR-/- mutants, a modification which was enhanced in the triple-mutant mice. In the formalin test, MOR-/- and DOR-/- mice showed increased responses in the early and late phases, respectively, while the triple mutant tended to show enhanced nociception in both phases. Finally, the enhanced response of KOR-/- mice in the writhing test, which we have demonstrated previously, was confirmed in double MOR/KOR- and triple-mutant mice. Together, the data support the existence of an antinociceptive opioid tone. Each receptor presents a distinct pattern of activities, with mu receptors influencing responses to mechanical, chemical and thermal nociception at a supraspinal level, kappa receptors involved in spinally mediated thermal nociception and chemical visceral pain, and delta receptors modulating mechanical nociception and inflammatory pain. Phenotypes of mutant mice were subtle, suggesting a low endogenous opioid tone in the regulation of physiological pain.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Female
- Formaldehyde
- Gene Deletion
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Pain/chemically induced
- Pain/genetics
- Pain Measurement/methods
- Reaction Time
- Receptors, Opioid/genetics
- Receptors, Opioid/physiology
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/physiology
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/physiology
- Sex Factors
Collapse
Affiliation(s)
- Miquel Martin
- Laboratori de Neurofarmacologia, Facultat de Ciéncies de la Salut i de la Vida, Universitat Pompeu Fabra, C/Dr Aiguader 80, 08003 Barcelona, Spain
| | | | | | | |
Collapse
|
43
|
Clarke S, Czyzyk T, Ansonoff M, Nitsche JF, Hsu MS, Nilsson L, Larsson K, Borsodi A, Toth G, Hill R, Kitchen I, Pintar JE. Autoradiography of opioid and ORL1 ligands in opioid receptor triple knockout mice. Eur J Neurosci 2002; 16:1705-12. [PMID: 12431223 DOI: 10.1046/j.1460-9568.2002.02239.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Three genes for the opioid receptors ( micro, delta and kappa) and a gene coding for a related receptor (ORL1) have been cloned but pharmacological studies suggest that further subtypes exist that remain poorly understood. To determine if there are other classically defined opioid binding sites we have carried out homogenate binding and section autoradiography with [3H]naloxone in mice that lack all three opioid genes and are hyperalgesic in a thermal nociceptive test. We have also examined [3H]bremazocine labelling in triple knockout brain and spinal cord as this ligand has been proposed to label novel kappa-receptors. No receptor labelling for either ligand was detected in the brains or spinal cord of knockout mice demonstrating that all binding is the product of the three known receptors and that there is no cross-labelling of the ORL1 receptor. Nociceptin (1 micro m) caused marked displacement of [3H]bremazocine in wild-type brains indicating that nociceptin at high concentrations can displace classical opioid binding. As a number of studies have proposed a close association between the classical opioid receptors and the ORL1 system we also hypothesized that loss of all of the classical opioid receptors might lead to compensatory changes in ORL1 receptors. Labelling of the ORL1 receptor with [3H]nociceptin showed region-dependent quantitative increases in triple knockout brains indicating a close relationship between the two systems in specific brain areas.
Collapse
Affiliation(s)
- Siân Clarke
- Pharmacology Group, School of Biomedical and Life Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mitolo-Chieppa D, Natale L, Marasciulo FL, De Salvatore G, Mitolo CI, Siro-Brigiani G, Renna G, De Salvia MA. Involvement of kappa-opioid receptors in peripheral response to nerve stimulation in kappa-opioid receptor knockout mice. AUTONOMIC & AUTACOID PHARMACOLOGY 2002; 22:233-9. [PMID: 12656949 DOI: 10.1046/j.1474-8673.2002.00263.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
1 The present study aimed to evaluate the role of kappa-opioid receptors at two peripheral sites, the vas deferens and the proximal colon, in kappa-opioid receptor knockout mice. We investigated the role of the kappa-opioid receptor in the vas deferens twitch response and in the colonic "off-contraction", a rebound contractile response which follows the inhibitory response to low frequencies stimulation (10, 20, 30 Hz) and which has been suggested to "locally" reproduce the contractile component of the peristaltic reflex. 2 Transmural stimulation of the vas deferens at lower frequencies (10 Hz, 10 V, 1 ms pulse trains lasting 0.5 s) evoked a contractile response that was significantly higher in the preparations from knockout mice because of lack of kappa-opioid receptors than in wild type mice. A selective kappa-opioid receptor agonist, U-50,488H, induced a dose-dependent inhibition of the electrically stimulated contraction in vas deferens. The percentages of reduction of the twitch response were significantly lower in knockout mice than in wild type mice after treatment with U-50,488H. The reduction of twitch response caused by U-50,488H was not reversed by administration of nor-binaltorphimine (nor-BNI) (5 x 10-6 m), a selective kappa-opioid receptor antagonist, in preparations from both knockout mice and wild type mice. U-50,488H has no effect on postsynaptic adrenergic receptors, as its administration did not affect the direct contractile response to noradrenaline. 3 Transmural stimulation (5 Hz, 20 V, 2 ms pulse trains lasting 30 s) induced inhibition of spontaneous activity of colonic strips during the period of stimulation, followed by an "off-contraction" after the cessation of stimulation. The statistical evaluation of the "off-contraction" responses between the two strains showed no significant difference. The off-contraction, measured in specimens from knockout mice, was inhibited concentration-dependently by U-50,488H (P < 0.01) and significantly less than from wild type mice. 4 The effect of U-50,488H was not reversed by administration of nor-BNI (5 x 10-6 m), either in preparations from knockout mice or from wild type mice. 5 Our data may suggest that kappa-opioid receptors are involved in some peripheral responses to the nerve stimulation, as indicated by the effect of U-50,488H, a selective kappa-opioid receptor agonist. However, the involvement of kappa-opioid receptor was also present, although less apparent, in kappa -opioid receptor knockout mice, suggesting either that this drug acts not only on kappa-opioid receptors but also on other receptor sites, such as kappa-like receptors. An alternative interpretation can be related to a sodium channel blocking action of U-50,488H, which could explain the inhibitory effects of twitch response still present but less evident in knockout strain and the lack of effect of the antagonist nor-BNI.
Collapse
Affiliation(s)
- D Mitolo-Chieppa
- Department of Pharmacology and Human Physiology, Medical School, University of Bari, Piazza G. Cesare, 70124 Bari, Italy
| | | | | | | | | | | | | | | |
Collapse
|
45
|
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: 45] [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
|
46
|
Abstract
The endogenous opioid system consists of three opioid peptide precursor genes encoding enkephalins (preproenkephalin, Penk), dynorphins (preprodynorphin, Pdyn) and beta-endorphin (betaend), proopiomelanocortin (POMC) and three receptor genes encoding mu-opiod receptor (MOR), delta-opiod receptor (DOR) and kappa-opiod receptor (KOR). In the past years, all six genes have been inactivated in mice by homologous recombination. The analysis of spontaneous behavior in mutant mice has demonstrated significant and distinct roles of each gene in modulating locomotion, pain perception and emotional behaviors. The observation of opposing phenotypes of MOR- and DOR-deficient mice in several behaviors highlights unexpected roles for DOR to be further explored genetically and using more specific delta compounds. The analysis of responses of mutant mice to exogenous opiates has definitely clarified the essential role of MOR in both morphine analgesia and addiction, and demonstrated that DOR and KOR remain promising targets for pain treatment. These studies also show that prototypic DOR agonists partially require MOR for their biological activity and provide some support for the postulated mu-delta interactions in vivo. Finally, data confirm and define a role for several genes of the opioid system in responses to other drugs of abuse, and the triple opioid receptor knockout mutant allows exploring non-classical opioid pharmacology. In summary, the study of null mutant mice has extended our previous knowledge of the opioid system by identifying the molecular players in opioid pharmacology and physiology. Future studies should involve parallel behavioral analysis of mice lacking receptors and peptides and will benefit from more sophisticated gene targeting approaches, including site-directed and anatomically-restricted mutations.
Collapse
Affiliation(s)
- Brigitte L Kieffer
- IGBMC UMR 7104, Parc d'innovation 1, rue Laurent Fries, B.P.163, 67404 Illkirch Cedex, France.
| | | |
Collapse
|
47
|
Abstract
The opioid system controls nociception, stress responses, and addictive behaviors. Exogenous alkaloid opiates and endogenous opioid peptides stimulate mu-, delta- and kappa-opioid receptors, whose activities have long been analyzed by pharmacological tools. Mice lacking opioid receptor and opioid peptide precursor genes have now been produced by gene targeting. Behavioral analysis of mutant animals in the absence of drug has highlighted a distinct role of opioid receptors or peptides in nociception and revealed an important role for delta receptors in emotional behaviors. The examination of responses to drugs has clarified involvement of each receptor as molecular targets for exogenous opiates in vivo. Those data have also demonstrated the critical role of mu-receptor in cannabinoid and alcohol reinforcement and confirmed the involvement of kappa receptor in several dysphoric responses. Ongoing studies therefore help in understanding the molecular basis of opioid-controlled behaviors and will contribute to the development of novel therapeutics for pain, anxiety, and drug abuse.
Collapse
MESH Headings
- Affect/physiology
- Analgesia
- Animals
- Humans
- Mice
- Mice, Knockout
- Pain/genetics
- Pain/physiopathology
- Receptors, Opioid/drug effects
- Receptors, Opioid/genetics
- Receptors, Opioid/physiology
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/physiology
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/physiology
- Substance-Related Disorders/genetics
Collapse
Affiliation(s)
- C Gavériaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, BP 163, 67404, Illkirch Cedex, C.U. de Strasbourg, France
| | | |
Collapse
|
48
|
Shannon HE, Lutz EA. Comparison of the peripheral and central effects of the opioid agonists loperamide and morphine in the formalin test in rats. Neuropharmacology 2002; 42:253-61. [PMID: 11804622 DOI: 10.1016/s0028-3908(01)00173-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The effects of the peripherally restricted opioid agonist loperamide were compared to those of morphine in the formalin test in rats. Both loperamide and morphine were efficacious in producing antihyperalgesia after both subcutaneous and intracisternal administration. The antihyperalgesic effects of peripherally administered loperamide and morphine were antagonized by both naloxone and its quaternary derivative naloxone methiodide. The effects of intracisternally administered loperamide and morphine were antagonized by naloxone SC. However, quaternary naloxone SC did not block the effects of intracisternally administered loperamide, and, quaternary naloxone blocked intracisternally morphine only at a dose approximately 10-fold higher than that required to block peripherally administered morphine. In addition, approximately 10-fold higher doses of naloxone administered SC were required to antagonize loperamide compared to doses required to antagonize morphine when the agonists were administered subcutaneously, suggesting that the effects of loperamide might be mediated by opioid receptors different from those which mediated the effects of morphine. However, neither the kappa-receptor selective antagonist nor-binaltorphimine nor the delta-receptor selective antagonist naltrindole blocked the effects of either opioid agonist. The present results are consistent with the interpretation that the antihyperalgesic effects of opioid agonists can have both a peripheral and a central component of action, and that the peripheral component of action is sufficient to produce antihyperalgesia in the formalin test after peripheral administration. The present results provide further evidence that peripherally restricted opioid agonists might provide clinically useful treatment of some pain states, in particular pain states that might involve sensitization of peripheral nociceptors.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/antagonists & inhibitors
- Analgesics, Opioid/pharmacology
- Animals
- Antidiarrheals/administration & dosage
- Antidiarrheals/antagonists & inhibitors
- Antidiarrheals/pharmacology
- Cisterna Magna
- Dose-Response Relationship, Drug
- Formaldehyde
- Injections
- Injections, Subcutaneous
- Loperamide/administration & dosage
- Loperamide/antagonists & inhibitors
- Loperamide/pharmacology
- Male
- Morphine/administration & dosage
- Morphine/antagonists & inhibitors
- Morphine/pharmacology
- Naloxone/pharmacology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Pain Measurement/drug effects
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, mu/drug effects
Collapse
Affiliation(s)
- Harlan E Shannon
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | |
Collapse
|