1
|
Wang J, Hu J, Liao P, Xue S, He S, Chen R, Zhao X, Liu W. The Synthesis of Biphasic Metabolites of Carfentanil. Molecules 2023; 28:7625. [PMID: 38005347 PMCID: PMC10674982 DOI: 10.3390/molecules28227625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Carfentanil is an ultra-potent synthetic opioid. The Russian police force used both carfentanil and remifentanil to resolve a hostage incident in Moscow. This reported use sparked an interest in the pharmacology and toxicology of carfentanil in the human body, and data on its metabolites were later published. However, there have been few studies on the synthesis of carfentanil metabolites, and biological extraction has also put forward large uncertainty in subsequent studies. The aim of the present study is to investigate the synthesis of biphasic metabolites that are unique to carfentanil. The purpose was to produce corresponding metabolites conveniently, quickly, and at low cost that can be used for comparison with published structures and to confirm the administration of carfentanil.
Collapse
Affiliation(s)
- Junchang Wang
- Shanghai Yuansi Standard Science and Technology Co., Ltd., Shanghai 200072, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai 200072, China
| | - Jianwen Hu
- Shanghai Yuansi Standard Science and Technology Co., Ltd., Shanghai 200072, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai 200072, China
| | - Pingyong Liao
- Shanghai Yuansi Standard Science and Technology Co., Ltd., Shanghai 200072, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai 200072, China
| | - Shang Xue
- Shanghai Yuansi Standard Science and Technology Co., Ltd., Shanghai 200072, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai 200072, China
| | - Shan He
- Shanghai Yuansi Standard Science and Technology Co., Ltd., Shanghai 200072, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai 200072, China
| | - Ruijia Chen
- Shanghai Yuansi Standard Science and Technology Co., Ltd., Shanghai 200072, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai 200072, China
| | - Xuejun Zhao
- Shanghai Yuansi Standard Science and Technology Co., Ltd., Shanghai 200072, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai 200072, China
| | - Wenbin Liu
- Shanghai Yuansi Standard Science and Technology Co., Ltd., Shanghai 200072, China
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai 200072, China
| |
Collapse
|
2
|
Rossi GC, Bodnar RJ. Interactive Mechanisms of Supraspinal Sites of Opioid Analgesic Action: A Festschrift to Dr. Gavril W. Pasternak. Cell Mol Neurobiol 2021; 41:863-897. [PMID: 32970288 DOI: 10.1007/s10571-020-00961-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/03/2020] [Indexed: 12/30/2022]
Abstract
Almost a half century of research has elaborated the discoveries of the central mechanisms governing the analgesic responses of opiates, including their receptors, endogenous peptides, genes and their putative spinal and supraspinal sites of action. One of the central tenets of "gate-control theories of pain" was the activation of descending supraspinal sites by opiate drugs and opioid peptides thereby controlling further noxious input. This review in the Special Issue dedicated to the research of Dr. Gavril Pasternak indicates his contributions to the understanding of supraspinal mediation of opioid analgesic action within the context of the large body of work over this period. This review will examine (a) the relevant supraspinal sites mediating opioid analgesia, (b) the opioid receptor subtypes and opioid peptides involved, (c) supraspinal site analgesic interactions and their underlying neurophysiology, (d) molecular (particularly AS) tools identifying opioid receptor actions, and (e) relevant physiological variables affecting site-specific opioid analgesia. This review will build on classic initial studies, specify the contributions that Gavril Pasternak and his colleagues did in this specific area, and follow through with studies up to the present.
Collapse
Affiliation(s)
- Grace C Rossi
- Department of Psychology, C.W. Post College, Long Island University, Post Campus, Brookville, NY, USA.
| | - Richard J Bodnar
- Department of Psychology, Queens College of the City University of New York, Flushing, NY, USA
- CUNY Neuroscience Collaborative, Graduate Center, CUNY, New York, NY, USA
| |
Collapse
|
3
|
Islam A, Rahman MA, Brenner MB, Moore A, Kellmyer A, Buechler HM, DiGiorgio F, Verchio VR, McCracken L, Sumi M, Hartley R, Lizza JR, Moura-Letts G, Fischer BD, Keck TM. Abuse Liability, Anti-Nociceptive, and Discriminative Stimulus Properties of IBNtxA. ACS Pharmacol Transl Sci 2020; 3:907-920. [DOI: 10.1021/acsptsci.0c00066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Bradford D. Fischer
- Cooper Medical School of Rowan University, 401 Broadway, Camden, New Jersey 08103, United States
| | - Thomas M. Keck
- Cooper Medical School of Rowan University, 401 Broadway, Camden, New Jersey 08103, United States
| |
Collapse
|
4
|
Dhummakupt ES, Rizzo GM, Feasel M, Mach PM, Tran BQ, Carmany DO, Demond PS, McBride EM, Maughan M, Sekowski JW, Glaros T. Proteomic and Metabolomic Profiling Identify Plasma Biomarkers for Exposure to Ultra-low Levels of Carfentanil. Toxicol Sci 2020; 167:524-535. [PMID: 30321427 DOI: 10.1093/toxsci/kfy259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Despite the recent epidemic of fentanyl abuse, there are few validated assays capable of rapidly detecting these compounds. In order to improve the ability to detect carfentanil at physiologically relevant concentrations, we developed a systems biology approach to discover host-based markers which are specifically amplified upon exposure in a rabbit model. For this work, two "omics" pipelines utilizing mass spectrometry were developed and leveraged. First, a proteomics pipeline was developed to interrogate the blood plasma for protein-based biomarkers. Due to the incredible dynamic range of the plasma protein content, a multi-dimensional fractionation technique was used to partition and more accurately investigate the circulating plasma proteome. Isobaric tandem mass tags were integrated into the workflow to make quantitative assessments across all animals for an extended time course post-exposure. In addition to the proteomics efforts, blood plasma was also processed through an untargeted metabolomics pipeline. This approach allows for the identification of >800 small molecule features. By processing and analyzing data sets in parallel, we were able to identify a unique fingerprint of protein and metabolite perturbations that manifest following exposure to carfentanil.
Collapse
Affiliation(s)
- Elizabeth S Dhummakupt
- Biosciences Division, BioDefense Branch, US Army Edgewood Chemical Biological Center, Aberdeen Proving Ground, MD 21010, USA
| | | | - Michael Feasel
- Biosciences Division, BioDefense Branch, US Army Edgewood Chemical Biological Center, Aberdeen Proving Ground, MD 21010, USA
| | - Phillip M Mach
- Biosciences Division, BioDefense Branch, US Army Edgewood Chemical Biological Center, Aberdeen Proving Ground, MD 21010, USA
| | | | | | | | - Ethan M McBride
- Biosciences Division, BioDefense Branch, US Army Edgewood Chemical Biological Center, Aberdeen Proving Ground, MD 21010, USA
| | | | - Jennifer W Sekowski
- Biosciences Division, BioDefense Branch, US Army Edgewood Chemical Biological Center, Aberdeen Proving Ground, MD 21010, USA
| | - Trevor Glaros
- Biosciences Division, BioDefense Branch, US Army Edgewood Chemical Biological Center, Aberdeen Proving Ground, MD 21010, USA
| |
Collapse
|
5
|
Smith LC, Bremer PT, Hwang CS, Zhou B, Ellis B, Hixon MS, Janda KD. Monoclonal Antibodies for Combating Synthetic Opioid Intoxication. J Am Chem Soc 2019; 141:10489-10503. [PMID: 31187995 DOI: 10.1021/jacs.9b04872] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Opioid abuse in the United States has been declared a national crisis and is exacerbated by an inexpensive, readily available, and illicit supply of synthetic opioids. Specifically, fentanyl and related analogues such as carfentanil pose a significant danger to opioid users due to their high potency and rapid acting depression of respiration. In recent years these synthetic opioids have become the number one cause of drug-related deaths. In our research efforts to combat the public health threat posed by synthetic opioids, we have developed monoclonal antibodies (mAbs) against the fentanyl class of drugs. The mAbs were generated in hybridomas derived from mice vaccinated with a fentanyl conjugate vaccine. Guided by a surface plasmon resonance (SPR) binding assay, we selected six hybridomas that produced mAbs with 10-11 M binding affinity for fentanyl, yet broad cross-reactivity with related fentanyl analogues. In mouse antinociception models, our lead mAb (6A4) could blunt the effects of both fentanyl and carfentanil in a dose-responsive manner. Additionally, mice pretreated with 6A4 displayed enhanced survival when subjected to fentanyl above LD50 doses. Pharmacokinetic analysis revealed that the antibody sequesters large amounts of these drugs in the blood, thus reducing drug biodistribution to the brain and other tissue. Lastly, the 6A4 mAb could effectively reverse fentanyl/carfentanil-induced antinociception comparable to the opioid antagonist naloxone, the standard of care drug for treating opioid overdose. While naloxone is known for its short half-life, we found the half-life of 6A4 to be approximately 6 days in mice, thus monoclonal antibodies could theoretically be useful in preventing renarcotization events in which opioid intoxication recurs following quick metabolism of naloxone. Our results as a whole demonstrate that monoclonal antibodies could be a desirable treatment modality for synthetic opioid overdose and possibly opioid use disorder.
Collapse
Affiliation(s)
- Lauren C Smith
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 N Torrey Pines Road , La Jolla , California 92037 , United States
| | - Paul T Bremer
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 N Torrey Pines Road , La Jolla , California 92037 , United States.,Cessation Therapeutics LLC , 3031 Tisch Way Ste 505 , San Jose , California 95128 , United States
| | - Candy S Hwang
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 N Torrey Pines Road , La Jolla , California 92037 , United States.,Department of Chemistry , Southern Connecticut State University , New Haven , Connecticut 06515 , United States
| | - Bin Zhou
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 N Torrey Pines Road , La Jolla , California 92037 , United States
| | - Beverly Ellis
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 N Torrey Pines Road , La Jolla , California 92037 , United States
| | - Mark S Hixon
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 N Torrey Pines Road , La Jolla , California 92037 , United States.,Mark S. Hixon Consulting LLC , 11273 Spitfire Road , San Diego , California 92126 , United States
| | - Kim D Janda
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 N Torrey Pines Road , La Jolla , California 92037 , United States
| |
Collapse
|
6
|
Dragovic L, Corsi N. Fatal overdoses involving carfentanil: A case series. JOURNAL OF FORENSIC SCIENCE AND MEDICINE 2019. [DOI: 10.4103/jfsm.jfsm_74_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
7
|
Sader S, Anant K, Wu C. To probe interaction of morphine and IBNtxA with 7TM and 6TM variants of the human μ-opioid receptor using all-atom molecular dynamics simulations with an explicit membrane. Phys Chem Chem Phys 2018; 20:1724-1741. [PMID: 29265141 DOI: 10.1039/c7cp06745c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IBNtxA, a morphine derivative, is 10-fold more potent and has a better safety profile than morphine. Animal studies indicate that the analgesic effect of IBNtxA appears to be mediated by the activation of truncated splice variants (6TM) of the Mu opioid receptor (MOR-1) where transmembrane helix 1 (TM1) is removed. Interestingly, morphine is unable to activate 6TM variants. To date, a high resolution structure of 6TM variants is missing, and the interaction of 6TM variants with IBNtxA and morphine remains elusive. In this study we used homology modeling, docking and molecular dynamics (MD) simulations to study a representative 6TM variant (G1) and a full-length 7TM variant of human MOR-1 in complex with IBNtxA and morphine respectively. The structural models of human G1 and 7TM were obtained by homology modeling using the X-ray solved crystal structure of the active mouse 7TM bound to an agonist BU72 (PDB id: ) as the template. Our 6000 ns MD data show that either TM1 truncation (i.e. from 7TM to 6TM) or ligand modification (i.e. from morphine to IBNtxA) alone causes the loss of key morphine-7TM interactions that are well-known to be required for MOR-1 activation. Receptor disruptions are mainly located at TMs 2, 3, 6 and 7 in comparison with the active crystal complex. However, when both perturbations occur in the 6TM-IBNtxA complex, the key ligand-receptor interactions and the receptor conformation are recovered to resemble those in the active 7TM-morphine complex. Our molecular switch analysis further explains well why morphine is not able to activate 6TM variants. The close resemblance between 6TM-IBTtxA and 7TM in complex with PZM21, a G-protein biased 7TM agonist, suggests the possible biased agonism of IBNtxA on G1, which is consistent with its reduced side effects.
Collapse
Affiliation(s)
- Safaa Sader
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA.
| | | | | |
Collapse
|
8
|
He C, Li H, Zhang J, Kang Y, Jia F, Dong S, Zhou L. Supraspinal inhibitory effects of chimeric peptide MCRT on gastrointestinal motility in mice. J Pharm Pharmacol 2017. [DOI: 10.1111/jphp.12761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Abstract
Objectives
Chimeric peptide MCRT, based on morphiceptin and PFRTic-NH2, was a bifunctional ligand of μ- and δ-opioid receptors (MOR-DOR) and produced potent analgesia in tail-withdrawal test. The study focused on the supraspinal effects of morphiceptin, PFRTic-NH2 and MCRT on gastrointestinal motility. Moreover, opioid receptor antagonists, naloxone (non-selective), cyprodime (MOR selective) and naltrindole (DOR selective) were utilized to explore the mechanisms.
Methods
Intracerebroventricular administration was achieved via the implanted cannula. Gastric emptying and intestinal transit were measured to evaluate gastrointestinal motility.
Key findings
(1) At supraspinal level, morphiceptin, PFRTic-NH2 and MCRT significantly decreased gastric emptying and intestinal transit; (2) MCRT at 1 nmol/mouse, far higher than its analgesic dose (ED50 = 29.8 pmol/mouse), failed to regulate the gastrointestinal motility; (3) MCRT-induced gastrointestinal dysfunction could be completely blocked by naloxone and naltrindole, but not affected by cyprodime.
Conclusions
(1) Morphiceptin and PFRTic-NH2 played important roles in the regulation of gastrointestinal motility; (2) MCRT possessed higher bioactivity of pain relief than gastrointestinal regulation, suggesting its promising analgesic property; (3) MCRT-induced motility disorders were sensitive to DOR but not to MOR blockade, indicating the pain-relieving specificity of speculated MOR subtype or splice variant or MOR-DOR heterodimer.
Collapse
Affiliation(s)
- Chunbo He
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hailan Li
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Jing Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yanping Kang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Fang Jia
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Shouliang Dong
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, China
| | - Lanxia Zhou
- The Core Laboratory of the First Affiliated Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou, China
| |
Collapse
|
9
|
Genetic dissociation of morphine analgesia from hyperalgesia in mice. Psychopharmacology (Berl) 2017; 234:1891-1900. [PMID: 28343361 PMCID: PMC5520541 DOI: 10.1007/s00213-017-4600-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/10/2017] [Indexed: 01/10/2023]
Abstract
RATIONALE Morphine is the prototypic mu opioid, producing its analgesic actions through traditional 7 transmembrane domain (7TM) G-protein-coupled receptors generated by the mu opioid receptor gene (Oprm1). However, the Oprm1 gene undergoes extensive alternative splicing to yield three structurally distinct sets of splice variants. In addition to the full-length 7TM receptors, it produces a set of truncated variants comprised of only 6 transmembrane domains (6TM). OBJECTIVES This study explored the relative contributions of 7TM and 6TM variants in a range of morphine actions. METHODS Groups of male and mixed-gender wild-type and exon 11 Oprm1 knockout mice were examined in a series of behavioral assays measuring analgesia, hyperalgesia, respiration, and reward in conditioned place preference assays. RESULTS Loss of the 6TM variants in an exon 11 knockout (E11 KO) mouse did not affect morphine analgesia, reward, or respiratory depression. However, E11 KO mice lacking 6TM variants failed to show morphine-induced hyperalgesia, developed tolerance more slowly than wild-type mice, and did not display hyperlocomotion. CONCLUSIONS Together, our findings confirm the established role of 7TM mu receptor variants in morphine analgesia, reward, and respiratory depression, but reveal an unexpected obligatory role for 6TM variants in morphine-induced hyperalgesia and a modulatory role in morphine tolerance and dependence.
Collapse
|
10
|
Feasel MG, Wohlfarth A, Nilles JM, Pang S, Kristovich RL, Huestis MA. Metabolism of Carfentanil, an Ultra-Potent Opioid, in Human Liver Microsomes and Human Hepatocytes by High-Resolution Mass Spectrometry. AAPS JOURNAL 2016; 18:1489-1499. [PMID: 27495118 DOI: 10.1208/s12248-016-9963-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/15/2016] [Indexed: 11/30/2022]
Abstract
Carfentanil is an ultra-potent synthetic opioid. No human carfentanil metabolism data are available. Reportedly, Russian police forces used carfentanil and remifentanil to resolve a hostage situation in Moscow in 2002. This alleged use prompted interest in the pharmacology and toxicology of carfentanil in humans. Our study was conducted to identify human carfentanil metabolites and to assess carfentanil's metabolic clearance, which could contribute to its acute toxicity in humans. We used Simulations Plus's ADMET Predictor™ and Molecular Discovery's MetaSite™ to predict possible metabolite formation. Both programs gave similar results that were generally good but did not capture all metabolites seen in vitro. We incubated carfentanil with human hepatocytes for up to 1 h and analyzed samples on a Sciex 3200 QTRAP mass spectrometer to measure parent compound depletion and extrapolated that to represent intrinsic clearance. Pooled primary human hepatocytes were then incubated with carfentanil up to 6 h and analyzed for metabolite identification on a Sciex 5600+ TripleTOF (QTOF) high-resolution mass spectrometer. MS and MS/MS analyses elucidated the structures of the most abundant metabolites. Twelve metabolites were identified in total. N-Dealkylation and monohydroxylation of the piperidine ring were the dominant metabolic pathways. Two N-oxide metabolites and one glucuronide metabolite were observed. Surprisingly, ester hydrolysis was not a major metabolic pathway for carfentanil. While the human liver microsomal system demonstrated rapid clearance by CYP enzymes, the hepatocyte incubations showed much slower clearance, possibly providing some insight into the long duration of carfentanil's effects.
Collapse
Affiliation(s)
- Michael G Feasel
- Edgewood Chemical Biological Center, Research Development and Engineering Command, U.S. Army, 5183 Blackhawk Road, Gunpowder, Maryland, 21010-5424, USA.
| | - Ariane Wohlfarth
- Chemistry and Drug Metabolism, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, 21224, USA.,Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, 58758, Linköping, Sweden.,Division of Drug Research, Department of Medical Health Sciences, Linköping University, 58185, Linköping, Sweden
| | | | | | - Robert L Kristovich
- Edgewood Chemical Biological Center, Research Development and Engineering Command, U.S. Army, 5183 Blackhawk Road, Gunpowder, Maryland, 21010-5424, USA
| | - Marilyn A Huestis
- Chemistry and Drug Metabolism, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, 21224, USA
| |
Collapse
|
11
|
Gretton SK, Droney J. Splice variation of the mu-opioid receptor and its effect on the action of opioids. Br J Pain 2015; 8:133-8. [PMID: 26516547 DOI: 10.1177/2049463714547115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
An individual's response to opioids is influenced by a complex combination of genetic, molecular and phenotypic factors.Intra- and inter-individual variations in response to mu opioids have led to the suggestion that mu-opioid receptor subtypes exist.Scientists have now proven that mu-opioid receptor subtypes exist and that they occur through a mechanism promoting protein diversity, called alternative splicing.The ability of mu opioids to differentially activate splice variants may explain some of the clinical differences observed between mu opioids.This article examines how differential activation of splice variants by mu opioids occurs through alternative mu-opioid receptor binding, through differential receptor activation, and as a result of the distinct distribution of variants located regionally and at the cellular level.
Collapse
|
12
|
Grinnell SG, Majumdar S, Narayan A, Le Rouzic V, Ansonoff M, Pintar JE, Pasternak GW. Pharmacologic characterization in the rat of a potent analgesic lacking respiratory depression, IBNtxA. J Pharmacol Exp Ther 2014; 350:710-8. [PMID: 24970924 DOI: 10.1124/jpet.114.213199] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IBNtxA (3'-iodobenzoyl-6β-naltrexamide) is a potent analgesic in mice lacking many traditional opioid side effects. In mice, it displays no respiratory depression, does not produce physical dependence with chronic administration, and shows no cross-tolerance to morphine. It has limited effects on gastrointestinal transit and shows no reward behavior. Biochemical studies indicate its actions are mediated through a set of μ-opioid receptor clone MOR-1 splice variants associated with exon 11 that lack exon 1 and contain only six transmembrane domains. Like the mouse and human, rats express exon 11-associated splice variants that also contain only six transmembrane domains, raising the question of whether IBNtxA would have a similar pharmacologic profile in rats. When given systemically, IBNtxA is a potent analgesic in rats, with an ED50 value of 0.89 mg/kg s.c., approximately 4-fold more potent than morphine. It shows no analgesic cross-tolerance in morphine-pelleted rats. IBNtxA displays no respiratory depression as measured by blood oxygen saturation. In contrast, oximetry shows that an equianalgesic dose of morphine lowers blood oxygen saturation values by 30%. IBNtxA binding is present in a number of brain regions, with the thalamus standing out with very high levels and the cerebellum with low levels. As in mice, IBNtxA is a potent analgesic in rats with a favorable pharmacologic profile and reduced side effects.
Collapse
Affiliation(s)
- Steven G Grinnell
- Department of Neurology (S.M., G.W.P.) and Molecular Pharmacology and Chemistry Program (V.L.R., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York; Neuroscience (S.G.G., A.N., G.W.P.) and Pharmacology (G.W.P.) Graduate Programs, Weill Cornell Graduate School of Medical Sciences, New York, New York; and Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey (M.A., J.E.P.)
| | - Susruta Majumdar
- Department of Neurology (S.M., G.W.P.) and Molecular Pharmacology and Chemistry Program (V.L.R., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York; Neuroscience (S.G.G., A.N., G.W.P.) and Pharmacology (G.W.P.) Graduate Programs, Weill Cornell Graduate School of Medical Sciences, New York, New York; and Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey (M.A., J.E.P.)
| | - Ankita Narayan
- Department of Neurology (S.M., G.W.P.) and Molecular Pharmacology and Chemistry Program (V.L.R., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York; Neuroscience (S.G.G., A.N., G.W.P.) and Pharmacology (G.W.P.) Graduate Programs, Weill Cornell Graduate School of Medical Sciences, New York, New York; and Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey (M.A., J.E.P.)
| | - Valerie Le Rouzic
- Department of Neurology (S.M., G.W.P.) and Molecular Pharmacology and Chemistry Program (V.L.R., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York; Neuroscience (S.G.G., A.N., G.W.P.) and Pharmacology (G.W.P.) Graduate Programs, Weill Cornell Graduate School of Medical Sciences, New York, New York; and Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey (M.A., J.E.P.)
| | - Michael Ansonoff
- Department of Neurology (S.M., G.W.P.) and Molecular Pharmacology and Chemistry Program (V.L.R., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York; Neuroscience (S.G.G., A.N., G.W.P.) and Pharmacology (G.W.P.) Graduate Programs, Weill Cornell Graduate School of Medical Sciences, New York, New York; and Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey (M.A., J.E.P.)
| | - John E Pintar
- Department of Neurology (S.M., G.W.P.) and Molecular Pharmacology and Chemistry Program (V.L.R., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York; Neuroscience (S.G.G., A.N., G.W.P.) and Pharmacology (G.W.P.) Graduate Programs, Weill Cornell Graduate School of Medical Sciences, New York, New York; and Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey (M.A., J.E.P.)
| | - Gavril W Pasternak
- Department of Neurology (S.M., G.W.P.) and Molecular Pharmacology and Chemistry Program (V.L.R., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York; Neuroscience (S.G.G., A.N., G.W.P.) and Pharmacology (G.W.P.) Graduate Programs, Weill Cornell Graduate School of Medical Sciences, New York, New York; and Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey (M.A., J.E.P.)
| |
Collapse
|
13
|
Abstract
Opioids remain the mainstay of severe pain management in patients with cancer. The hallmark of pain management is individualization of therapy. Although almost all clinically used drugs act through mu opioid receptors, they display subtle but important differences pharmacologically. Furthermore, not all patients respond equally well to all drugs. Evidence suggests that these variable responses among patients have a biologic basis and are likely to involve both biased agonism and the many mu opioid receptor subtypes that have been cloned.
Collapse
Affiliation(s)
- Gavril W Pasternak
- From Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY.
| |
Collapse
|
14
|
Pasternak GW. Opioids and their receptors: Are we there yet? Neuropharmacology 2014; 76 Pt B:198-203. [PMID: 23624289 PMCID: PMC4216569 DOI: 10.1016/j.neuropharm.2013.03.039] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/02/2013] [Accepted: 03/26/2013] [Indexed: 01/06/2023]
Abstract
Opioids have an important place in pharmacology. While their clinical use as analgesics is fundamental in medicine, their use is constrained by their side-effects and abuse potential. Pharmacologists have sought analgesics lacking side-effects and the abuse liability of the current agents. The identification of the opioid receptors in 1973 marked the beginning of our understanding of the molecular mechanisms of these agents. The isolation of the opioid peptides quickly followed, along with the classification of three families of opioid receptors. Clinicians have long been aware of subtle differences among the mu opioids that were not easily reconciled with a single receptor and selective antagonists implied two subdivisions of mu receptors. However, the cloning of the mu opioid receptor MOR-1 has led to the realization of the extensive complexity of the mu opioid receptor gene and its vast array of splice variants. Many of these splice variants are truncated and do not conform to the structure of traditional G-protein coupled receptors. Yet, evidence now shows that they are quite important and may prove valuable targets in the development of potent analgesics lacking the undesirable properties of current opioids. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Gavril W Pasternak
- Molecular Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA.
| |
Collapse
|
15
|
Abstract
Opiates are among the oldest medications available to manage a number of medical problems. Although pain is the current focus, early use initially focused upon the treatment of dysentery. Opium contains high concentrations of both morphine and codeine, along with thebaine, which is used in the synthesis of a number of semisynthetic opioid analgesics. Thus, it is not surprising that new agents were initially based upon the morphine scaffold. The concept of multiple opioid receptors was first suggested almost 50 years ago (Martin, 1967), opening the possibility of new classes of drugs, but the morphine-like agents have remained the mainstay in the medical management of pain. Termed mu, our understanding of these morphine-like agents and their receptors has undergone an evolution in thinking over the past 35 years. Early pharmacological studies identified three major classes of receptors, helped by the discovery of endogenous opioid peptides and receptor subtypes-primarily through the synthesis of novel agents. These chemical biologic approaches were then eclipsed by the molecular biology revolution, which now reveals a complexity of the morphine-like agents and their receptors that had not been previously appreciated.
Collapse
Affiliation(s)
- Gavril W Pasternak
- Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065.
| | | |
Collapse
|
16
|
Majumdar S, Subrath J, Le Rouzic V, Polikar L, Burgman M, Nagakura K, Ocampo J, Haselton N, Pasternak AR, Grinnell S, Pan YX, Pasternak GW. Synthesis and evaluation of aryl-naloxamide opiate analgesics targeting truncated exon 11-associated μ opioid receptor (MOR-1) splice variants. J Med Chem 2012; 55:6352-62. [PMID: 22734622 DOI: 10.1021/jm300305c] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
3-Iodobenzoylnaltrexamide 1 (IBNtxA) is a potent analgesic acting through a novel receptor target that lack many side-effects of traditional opiates composed, in part, of exon 11-associated truncated six transmembrane domain MOR-1 (6TM/E11) splice variants. To better understand the SAR of this drug target, a number of 4,5-epoxymorphinan analogues were synthesized. Results show the importance of a free 3-phenolic group, a phenyl ring at the 6 position, an iodine at the 3'or 4' position of the phenyl ring, and an N-allyl or c-propylmethyl group to maintain high 6TM/E11 affinity and activity. 3-Iodobenzoylnaloxamide 15 (IBNalA) with a N-allyl group displayed lower δ opioid receptor affinity than its naltrexamine analogue, was 10-fold more potent an analgesic than morphine, elicited no respiratory depression or physical dependence, and only limited inhibition of gastrointestinal transit. Thus, the aryl-naloxamide scaffold can generate a potent analgesic acting through the 6TM/E11 sites with advantageous side-effect profile and greater selectivity.
Collapse
Affiliation(s)
- Susruta Majumdar
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Truncated G protein-coupled mu opioid receptor MOR-1 splice variants are targets for highly potent opioid analgesics lacking side effects. Proc Natl Acad Sci U S A 2011; 108:19778-83. [PMID: 22106286 DOI: 10.1073/pnas.1115231108] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pain remains a pervasive problem throughout medicine, transcending all specialty boundaries. Despite the extraordinary insights into pain and its mechanisms over the past few decades, few advances have been made with analgesics. Most pain remains treated by opiates, which have significant side effects that limit their utility. We now describe a potent opiate analgesic lacking the traditional side effects associated with classical opiates, including respiratory depression, significant constipation, physical dependence, and, perhaps most important, reinforcing behavior, demonstrating that it is possible to dissociate side effects from analgesia. Evidence indicates that this agent acts through a truncated, six-transmembrane variant of the G protein-coupled mu opioid receptor MOR-1. Although truncated splice variants have been reported for a number of G protein-coupled receptors, their functional relevance has been unclear. Our evidence now suggests that truncated variants can be physiologically important through heterodimerization, even when inactive alone, and can comprise new therapeutic targets, as illustrated by our unique opioid analgesics with a vastly improved pharmacological profile.
Collapse
|
18
|
Study on the activation of the opioid receptors by a set of morphine derivatives in a well-defined assay system. Neurochem Res 2011; 37:410-6. [PMID: 22002663 DOI: 10.1007/s11064-011-0627-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 09/29/2011] [Accepted: 09/30/2011] [Indexed: 10/16/2022]
Abstract
As a first step in our search for new opiates, we have established cellular assays to monitor opioid receptor activation and study the activities of a set of morphine derivatives. Intracellular calcium changes were monitored in human embryonic kidney-293 T cells expressing individual opioid receptors upon cotransfection with a chimeric G protein. This assay was validated by comparing the potencies of the endogenous peptides to reported values. All of the opiates were found to interact with the three opioid receptor subtypes but with a range of differences in efficacies and potencies. Most of the opiates preferentially acted at the μ receptor. None of the opiates showed a preference for the δ receptor. Only oripavine and its precursor thebaine showed a preference for the κ over the μ receptor. The results indicate that the opiates with a C-3 hydroxyl group or C-6 ketone group but in the presence of a 7, 8-single bond exhibit higher activity. It is noteworthy that the 6-O-methyl group seems to improve the selectivity for κ receptor. This is the first comparative and comprehensive report on the activation of the three different opioid receptors by a set of morphine derivatives in a well-defined assay system. These data can serve as a basis for the characterization of novel opiates.
Collapse
|
19
|
Mu opioid receptors in pain management. ACTA ACUST UNITED AC 2011; 49:21-5. [PMID: 21453899 DOI: 10.1016/j.aat.2010.12.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 11/30/2010] [Accepted: 12/03/2010] [Indexed: 11/22/2022]
Abstract
Most of the potent analgesics currently in use act through the mu opioid receptor. Although they are classified as mu opioids, clinical experience suggests differences among them. The relative potencies of the agents can vary from patient to patient, as well as the side-effect profiles. These observations, coupled with pharmacological approaches in preclinical models, led to the suggestion of multiple subtypes of mu receptors. The explosion in molecular biology has led to the identification of a single gene encoding mu opioid receptors. It now appears that this gene undergoes extensive splicing, in which a single gene can generate multiple proteins. Evidence now suggests that these splice variants may help explain the clinical variability in responses among patients.
Collapse
|
20
|
Rossi GC, Matulonis JE, Richelson E, Barbut D, Pasternak GW. Systemically and topically active antinociceptive neurotensin compounds. J Pharmacol Exp Ther 2010; 334:1075-9. [PMID: 20576795 DOI: 10.1124/jpet.109.165282] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurotensin is a neurotransmitter/modulator with a wide range of actions. Using a series of 10 stable analogs, we have examined neurotensin antinociception in mice. By incorporating (2S)-2-amino-3-(1H-4-indoyl)propanoic acid (l-neoTrp), a series of neurotensin analogs have been synthesized that are stable in serum and are systemically active in vivo. When administered in mice, they all were antinociceptive in the radiant heat tail-flick assay. Time-action curves revealed a peak effect at 30 min and a duration of action ranging from 2 to 4 h. Dose-response curves revealed that two compounds were partial agonists with maximal responses below 75%, whereas all of the remaining compounds displayed a full response. Overall, the compounds were quite potent, with ED(50) values similar to those of opioids. At peak effect, the ED(50) values ranged from 0.91 to 9.7 mg/kg s.c. Two of the analogs were active topically. Together, these studies support the potential of neurotensin analogs as analgesics. They are active systemically and by using them topically, it may be possible to avoid problematic side effects, such as hypothermia and hypotension.
Collapse
Affiliation(s)
- Grace C Rossi
- Department of Psychology, C.W. Post Campus of Long Island University, Brookville, New York, USA
| | | | | | | | | |
Collapse
|
21
|
Meyer PJ, Fossum EN, Ingram SL, Morgan MM. Analgesic tolerance to microinjection of the micro-opioid agonist DAMGO into the ventrolateral periaqueductal gray. Neuropharmacology 2007; 52:1580-5. [PMID: 17445843 PMCID: PMC1971241 DOI: 10.1016/j.neuropharm.2007.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 02/22/2007] [Accepted: 03/01/2007] [Indexed: 11/22/2022]
Abstract
Repeated administration of the relatively low-efficacy micro-opioid receptor agonist morphine induces tolerance to its antinociceptive effects. High-efficacy agonists such as D-Ala2NMePhe4,Gly-ol5 (DAMGO) have been shown to be less effective at producing tolerance, suggesting that different neural mechanisms underlie tolerance to these agonists. However, the correlation between agonist efficacy and tolerance development has not been examined within the ventrolateral periaqueductal gray (vPAG), a brain area known to be crucial for the development of morphine tolerance. The current studies examined whether tolerance to DAMGO occurs within the vPAG, and whether repeated treatment with DAMGO into the vPAG alters the development of morphine tolerance. The results showed that repeated vPAG microinjections of DAMGO induced robust tolerance and cross-tolerance to morphine. Further, co-administration of a low dose of DAMGO with morphine potentiated morphine tolerance. These findings indicate that similar mechanisms underlie tolerance to morphine and DAMGO within the vPAG.
Collapse
Affiliation(s)
- Paul J Meyer
- Washington State University Vancouver, 14204 NE Salmon Creek Ave, Vancouver, WA 98660, USA
| | | | | | | |
Collapse
|
22
|
Mahurter L, Garceau C, Marino J, Schmidhammer H, Tóth G, Pasternak GW. Separation of binding affinity and intrinsic activity of the potent mu-opioid 14-methoxymetopon. J Pharmacol Exp Ther 2006; 319:247-53. [PMID: 16801454 DOI: 10.1124/jpet.106.105395] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Receptor binding studies of 5,14-O-dimethyloxymorphone (14-methoxymetopon) in brain membranes have established its high affinity for mu-binding sites, but its analgesic potency far exceeds the modest increase in binding affinity relative to other opioids. The current study has established the selectivity of [(3)H]14-methoxymetopon for mu sites in calf striatal membranes and for a number of full-length splice variants of the cloned murine mu-opioid receptor 1 (mMOR-1) in transfected cell lines. The binding affinity of [(3)H]14-methoxymetopon for the variants expressed in Chinese hamster ovary cells was quite high, with K(D) values around 0.2 nM for all of the variants with the exception of mMOR-1F (K(D) of 1.2 nM). The affinity for most of the expressed variants was greater than that seen in the brain membranes (K(D) of 0.99 nM). Functionally, in guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) binding assays with the MOR-1 variants, 14-methoxymetopon and the mu-opioid peptide [d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO) showed similar efficacies, as determined by maximal stimulation, but 14-methoxymetopon was up to 65-fold more potent than DAMGO. The greatest difference was seen with mMOR-1E and the least with mMOR-1C, which displayed only a 10-fold difference. These potency differences in the stimulation of [(35)S]GTPgammaS binding far exceeded the differences in binding affinity. The differences between 14-methoxymetopon and DAMGO remained after normalizing the potency shifts based upon receptor binding affinities and varied from 1.2-fold with mMOR-1C to 21-fold for mMOR-1 and 42-fold with mMOR-1F. Thus, 14-methoxymetopon is a potent agonist against all of the mMOR-1 splice variants, but its potency ranged widely despite similar binding affinities for most of the variants and may give insight into its unusual pharmacological profile.
Collapse
Affiliation(s)
- Loriann Mahurter
- Laboratory of Molecular Neuropharmacology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave., New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
23
|
Lewanowitsch T, Miller JH, Irvine RJ. Reversal of morphine, methadone and heroin induced effects in mice by naloxone methiodide. Life Sci 2005; 78:682-8. [PMID: 16102783 DOI: 10.1016/j.lfs.2005.05.062] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Accepted: 05/06/2005] [Indexed: 10/25/2022]
Abstract
Opioid overdose, which is commonly associated with opioid induced respiratory depression, is a problem with both therapeutic and illicit opioid use. While the central mechanisms involved in the effects of opioids are well described, it has also been suggested that a peripheral component may contribute to the effects observed. This study aimed to further characterise the effects of the peripherally acting naloxone methiodide on the respiratory, analgesic and withdrawal effects produced by various opioid agonists. A comparison of the respiratory and analgesic effects of morphine, methadone and heroin in male Swiss-Albino mice was conducted and respiratory depressive ED(80) doses of each opioid determined. These doses (morphine 9 mg/kg i.p., methadone 7 mg/kg i.p., and heroin 17 mg/kg i.p.) were then used to show that both naloxone (3 mg/kg i.p.) and naloxone methiodide (30-100 mg/kg i.p.) could reverse the respiratory and analgesic effects of these opioid agonists, but only naloxone precipitated withdrawal. Further investigation in female C57BL/6J mice using barometric plethysmography found that both opioid antagonists could reverse methadone induced decreases in respiratory rate and increases in tidal volume. Its effects do not appear to be strain or sex dependent. It was concluded that naloxone methiodide can reverse the respiratory and analgesic actions of a variety of opioid agonists, without inducing opioid withdrawal.
Collapse
Affiliation(s)
- Tanya Lewanowitsch
- Department of Clinical and Experimental Pharmacology, 5th Floor Medical School North, University of Adelaide, Adelaide 5005, Australia.
| | | | | |
Collapse
|
24
|
Laferrière A, Colin-Durand J, Moss IR. Ontogeny of respiratory sensitivity and tolerance to the mu-opioid agonist fentanyl in rat. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 156:210-7. [PMID: 16099308 DOI: 10.1016/j.devbrainres.2005.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2004] [Revised: 03/01/2005] [Accepted: 03/03/2005] [Indexed: 11/18/2022]
Abstract
Whereas developmental changes in analgesic sensitivity and tolerance to the mu-opioid agonist fentanyl have been reported, knowledge of respiratory responses to that drug is lacking. Using 7- and 14-day-old (P7, P14) and adult conscious rats, we first established, using whole body plethysmography, the fentanyl dose that decreased minute ventilation by 50% (ED50) at each age. ED50 increased with postnatal age (40, 60 and 120 microg/kg sc, respectively), indicating a high sensitivity to fentanyl in the youngest rats that decreased with maturation. In separate rat groups of the 3 ages, we injected each ED50 dose, once a day, for several consecutive days, until tolerance was established. Tolerance was defined as a reduction in respiratory depression from 50% to 75% of baseline. All age groups reached tolerance in minute ventilation, respiratory frequency, tidal volume and instantaneous flow (equivalent to respiratory drive). The P14 rat pups attained tolerance more rapidly (at 2.6 days) than did either the younger (5.1 days) or the adult rats (4.4 days). These results indicate that respiratory sensitivity and tolerance to fentanyl in rat vary in a distinct manner during maturation.
Collapse
Affiliation(s)
- Andrè Laferrière
- Developmental Respiratory Laboratory, Montreal Children's Hospital Research Institute, Montreal, QC, Canada H3H 1P3
| | | | | |
Collapse
|
25
|
Pasternak GW. Multiple opiate receptors: déjà vu all over again. Neuropharmacology 2004; 47 Suppl 1:312-23. [PMID: 15464147 DOI: 10.1016/j.neuropharm.2004.07.004] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Revised: 06/07/2004] [Accepted: 06/30/2004] [Indexed: 11/23/2022]
Abstract
The concept of multiple opioid receptors has changed dramatically since their initial proposal by Martin nearly 40 years ago. Three major opioid receptor families have now been proposed: mu, kappa and delta. Most of the opioid analgesics used clinically selectively bind to mu opioid receptors. Yet, clinicians have long appreciated subtle, but significant, differences in their pharmacology. These observations suggested more than one mu opioid receptor mechanism of action and led us to propose multiple mu opioid receptors over 20 years ago based upon a range of pharmacological and receptor binding approaches. A mu opioid receptor, MOR-1, was cloned about a decade ago. More recent studies have now identified a number of splice variants of this clone. These splice variants may help explain the pharmacology of the mu opioids and open interesting directions for future opioid research.
Collapse
Affiliation(s)
- Gavril W Pasternak
- Department of Neurology, Laboratory of Molecular Pharmacology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| |
Collapse
|
26
|
|
27
|
King MA, Su W, Nielan CL, Chang AH, Schütz J, Schmidhammer H, Pasternak GW. 14-Methoxymetopon, a very potent mu-opioid receptor-selective analgesic with an unusual pharmacological profile. Eur J Pharmacol 2003; 459:203-9. [PMID: 12524147 DOI: 10.1016/s0014-2999(02)02821-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
14-Methoxymetopon is a potent opioid analgesic. When given systemically, it is approximately 500-fold more active than morphine. However, this enhanced potency is markedly increased with either spinal or supraspinal administration, where its analgesic activity is more than a million-fold greater than morphine. It was mu-opioid receptor selective in binding assays and its analgesia was blocked only by mu-opioid receptor-selective antagonists. Yet, it had a different selectivity profile than either morphine or morphine-6beta-glucuronide. Unlike morphine, 14-methoxymetopon was antagonized by 3-O-methylnaltrexone, it was sensitive to antisense probes targeting exons 1, 2 and 8 of the opioid receptor gene and was inactive both spinally and supraspinally in CXBK mice. Although it retarded gastrointestinal transit, it displayed a ceiling effect with no dose lowering transit by more than 65%, in contrast to the complete inhibition of transit by morphine. These finding demonstrate that 14-methoxymetopon is a highly potent mu-opioid with a pharmacological profile distinct from that of the traditional mu-opioid morphine.
Collapse
Affiliation(s)
- Michael A King
- The Cotzias Laboratory of Neuro-Oncology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Neilan CL, Nguyen TM, Schiller PW, Pasternak GW. Pharmacological characterization of the dermorphin analog [Dmt(1)]DALDA, a highly potent and selective mu-opioid peptide. Eur J Pharmacol 2001; 419:15-23. [PMID: 11348625 DOI: 10.1016/s0014-2999(01)00946-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The dermorphin-derived peptide [Dmt(1)]DALDA (H-Dmt-D-Arg-Phe-Lys-NH(2)), labels mu-opioid receptors with high affinity and selectivity in receptor binding assays. In mouse, radiant heat tail-flick assay [Dmt(1)]DALDA produced profound spinal and supraspinal analgesia, being approximately 5000- and 100-fold more potent than morphine on a molar basis, respectively. When administered systemically, [Dmt(1)]DALDA was over 200-fold more potent than morphine. Pharmacologically, [Dmt(1)]DALDA was distinct from morphine. [Dmt(1)]DALDA displayed no cross-tolerance to morphine in the model used and it retained supraspinal analgesic activity in morphine-insensitive CXBK mice. Supraspinally, it also differed from morphine in its lack of sensitivity towards naloxonazine. Finally, in antisense mapping studies, [Dmt(1)]DALDA was insensitive to MOR-1 exon probes that reduced morphine analgesia, implying a distinct receptor mechanism of action. Thus, [Dmt(1)]DALDA is an interesting and extraordinarily potent, systemically active peptide analgesic, raising the possibility of novel approaches in the design of clinically useful drugs.
Collapse
Affiliation(s)
- C L Neilan
- Department of Molecular Neuropharmacology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | |
Collapse
|
29
|
Abstract
Morphine and most clinical opioids act through mu opioid receptors. Yet, their pharmacological profiles differ. The presence of incomplete cross-tolerance among these drugs clinically was one of the first indications that these mu opioids differed in their receptor mechanisms of action. This was followed by similar studies in preclinical models, which also found genetic differences in sensitivity toward morphine and other mu opioids. This concept of mu receptor multiplicity is now supported by antisense and gene knockout models. Although all the mu opioids are sensitive to antisense probes against the mu opioid receptor gene MOR-1, the sensitivity profiles of the drugs to the antisense probes differ based on the exon being targeted. Knockout mice also reveal striking differences. In one knockout mouse, morphine analgesia is completely lost while the potent mu drugs morphine-6beta-glucuronide and heroin both retain analgesic activity. Finally, cloning studies have identified at least seven different splice variants of the MOR-1 gene, with more likely. These studies illustrate the complexity of mu opioid pharmacology.
Collapse
Affiliation(s)
- G W Pasternak
- Laboratory of Molecular Neuropharmacology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| |
Collapse
|
30
|
King M, Su W, Chang A, Zuckerman A, Pasternak GW. Transport of opioids from the brain to the periphery by P-glycoprotein: peripheral actions of central drugs. Nat Neurosci 2001; 4:268-74. [PMID: 11224543 DOI: 10.1038/85115] [Citation(s) in RCA: 148] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Many peptides and transmitters found within the brain also have peripheral sites of action. We now demonstrate that the brain releases functionally active neurotransmitters/neuromodulators directly from the brain into the blood through a saturable P-glycoprotein (Pgp) transport system. Downregulating Pgp1 expression with antisense reduced the brain-to-blood transport of morphine, beta-endorphin and other opioids. Lowering Pgp expression significantly enhanced systemic morphine analgesia and prevented tolerance, but diminished the analgesic activity of centrally administered morphine, implying that supraspinal analgesia resulted from a combination of central and peripheral mechanisms activated by morphine transported from the brain to the blood. Similarly, mice with a disruption of the Mdr1a gene were more sensitive to systemic morphine and less sensitive to morphine given centrally. This ability of the Pgp transport system to pump functionally active compounds from the brain to periphery defines a potentially important mechanism for the central nervous system to modulate peripheral systems.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/drug effects
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP-Binding Cassette Transporters/metabolism
- Animals
- Biological Transport/drug effects
- Biological Transport/physiology
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/physiology
- Brain/drug effects
- Brain/metabolism
- Down-Regulation/drug effects
- Down-Regulation/physiology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics
- Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics
- Male
- Mice
- Mice, Knockout
- Morphine/pharmacokinetics
- Narcotics/blood
- Narcotics/pharmacokinetics
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- beta-Endorphin/pharmacokinetics
Collapse
Affiliation(s)
- M King
- The Cotzias Laboratory of Neuro-Oncology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| | | | | | | | | |
Collapse
|
31
|
Czapla MA, Gozal D, Alea OA, Beckerman RC, Zadina JE. Differential cardiorespiratory effects of endomorphin 1, endomorphin 2, DAMGO, and morphine. Am J Respir Crit Care Med 2000; 162:994-9. [PMID: 10988119 DOI: 10.1164/ajrccm.162.3.9911102] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The novel endogenous mu-opioid receptor (MOR) agonists endomorphin 1 (EM1) and 2 (EM2) were tested for their cardiorespiratory effects in conscious, freely behaving rats. After systemic (intravenous) administration of EM1, EM2, or the selective MOR agonist DAMGO, analgesia, minute ventilation (V E), heart rate (HR) and mean arterial blood pressure (BP) were measured. The threshold dose for analgesia was similar for all 3 peptides ( approximately 900 nmol/kg). All 3 compounds elicited biphasic V E responses, with marked, short-lived V E depressions (4-6 s) followed by more sustained V E increases (10-12 min). However, compared with responses elicited by EM2 or DAMGO, EM1 decreased V E only at higher doses, and produced greater V E stimulation. Morphine produced a V E decrease, but no subsequent V E increase. EM2 and DAMGO decreased HR and BP, while EM1 decreased HR, but did not decrease BP in conscious rats at doses up to 9,600 nmol/kg. In anesthetized rats, all 3 peptides decreased HR and BP. The decreases in V E, HR, and BP were blocked by the MOR antagonist, naloxone HCI (NIx). Only the HR and BP responses, however, were blocked by naloxone-methiodide (MeNIx), indicating central mediation of V E responses and peripheral mediation of cardiovascular responses. We conclude that MOR-selective compounds vary in their cardiorespiratory response characteristics which could be linked to differential cellular actions. The results support the concept that the analgesic, respiratory, and cardiovascular effects of MOR agonists can be dissociated and that EM1-like compounds could provide the basis for novel, safer analgesics.
Collapse
Affiliation(s)
- M A Czapla
- Constance S. Kaufman Pediatric Pulmonary Research Laboratory and Departments of Pediatrics, Physiology, Neuroscience, and Medicine, Tulane University School of Medicine, and Veterans Affairs Medical Center, New Orleans, Louisiana, USA
| | | | | | | | | |
Collapse
|
32
|
Verborgh C, Meert TF. Antagonistic effects of naloxone and naloxonazine on sufentanil-induced antinociception and respiratory depression in rats. Pain 1999; 83:17-24. [PMID: 10506668 DOI: 10.1016/s0304-3959(99)00068-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Several binding studies in rodent brain homogenates have revealed two distinct micro-opiate binding sites based on differences in binding affinity of several opiate peptides and opiate alkaloids. Naloxonazine (NLZ), which preferentially binds to the high affinity micro(1) sites, is often used to discriminate between pharmacological effects mediated by micro(1) and micro(2) binding sites. The present series of experiments were undertaken to compare the opioid antagonistic properties of naloxonazine and naloxone (NLX) (a non-selective micro(1)-antagonist) on intravenous (i.v.) and intrathecal (i.t.) sufentanil (SUF)-induced antinociception and respiratory depression. The opioid antagonists were given either intravenously at 5 min after SUF, or subcutaneously (s.c.) 24 h prior to the opioid. Intravenous NLX and NLZ reduced the i.v. and i. t. SUF-induced antinociception, hypercapnia and hypoxia when given directly after the opioid. There were no major differences in activity between both antagonists. Pretreatment with 30 mg/kg NLX did not reverse the i.v. or i.t. SUF-induced antinociception and respiratory depression. Subcutaneous pretreatment with doses up to 30 mg/kg NLX only partially antagonized the i.v. SUF-induced antinociception, while a complete reversal was present of the opioid-induced hypercapnia and hypoxia. With regard to i.t. SUF, doses up to 30 mg/kg NLZ were unable to reduce the antinociception. The respiratory depression was partially affected; with 30 mg/kg NLZ, the i.t. SUF-induced hypercapnia returned to baseline levels, whereas the SUF-induced hypoxia was only minimally affected. These results challenge the classical view of the selectivity of NLZ for the high affinity micro(1) binding sites. They further fail to conform an exclusive role for micro(2) receptor sites in the respiratory depression and spinal analgesia induced by a strong lipophilic opioid such as SUF in rats.
Collapse
Affiliation(s)
- C Verborgh
- Departement Anesthesiologie, Akademisch Ziekenhuis Vrije Universiteit Brussel, Laarbeeklaan 101, B-1090, Brussels, Belgium
| | | |
Collapse
|
33
|
Loh HH, Liu HC, Cavalli A, Yang W, Chen YF, Wei LN. mu Opioid receptor knockout in mice: effects on ligand-induced analgesia and morphine lethality. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 54:321-6. [PMID: 9555078 DOI: 10.1016/s0169-328x(97)00353-7] [Citation(s) in RCA: 274] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mu opioid receptor gene (MOR) was mutated in mice by a gene targeting procedure. In these MOR-knockout mice, the analgesic effects of morphine, its major metabolites, morphine-6-glucuronide (M-6-G) and morphine-6-ethereal sulfate (M-6-S), and endomorphin-2, as well as morphine-induced lethality, were drastically reduced, whereas the effects of DPDPE and U50488 remained unchanged. It is concluded that analgesic effects of mu-specific opioid ligands and acute morphine lethality are mediated by the mu receptor.
Collapse
Affiliation(s)
- H H Loh
- Dept. of Pharmacology, University of Minnesota Medical School, 3-249 Millard Hall, 435 Delaware St. S.E., Minneapolis, MN 55455, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Ismail Z, el-Guebaly N. Nicotine and endogenous opioids: toward specific pharmacotherapy. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 1998; 43:37-42. [PMID: 9494745 DOI: 10.1177/070674379804300103] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To address the theoretical framework behind opioid receptor antagonism for the treatment of nicotine abuse. The current literature is reviewed with a focus on opioid-nicotine interactions in animals and humans. Furthermore, previous studies addressing the effect of opioid antagonism on smoking behaviour are reviewed critically with a focus on suggestions and implications for future trials. METHOD Computerized data bases and reference lists of existing articles were searched for prior publications in 3 areas: 1) the association between nicotine and endogenous opioids, 2) nicotine and reward, and 3) opioid antagonism in the treatment of nicotine use. RESULTS Nicotine affects the mesolimbic reward pathway postsynaptically via nicotinic cholinergic receptors and presynaptically via the central nervous system's (CNS) neurohumoral pathways. Thus nicotine results in the release of endogenous opioids that bind to mu receptors, which increases the release of dopamine along this pathway. Studies to date have shown mixed results on the effect of opioid antagonism on smoking behaviour. CONCLUSIONS The role of opioid antagonism on smoking behaviour is unclear, despite the publication of 5 trials on the subject. Further trials of longer duration should be undertaken and use both longer-acting medications and those more specific to the mu receptor to further focus on the rewarding aspects of nicotine ingestion, thus addressing the craving for this drug. The development of adequate compounds has just begun, and psychiatrists can hope to have a more specific pharmacotherapy to address the cravings and short-term rewards of nicotine use.
Collapse
Affiliation(s)
- Z Ismail
- Department of Psychiatry, University of Alberta, Edmonton
| | | |
Collapse
|
35
|
Schmidhammer H. Opioid Receptor Antagonists**This review is dedicated to the memory of the late Drs. Sidney Archer and Hans W. Kosterlitz. PROGRESS IN MEDICINAL CHEMISTRY 1998. [DOI: 10.1016/s0079-6468(08)70035-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
36
|
Chen SW, Maguire PA, Davies MF, Beatty MF, Loew GH. Evidence for mu1-opioid receptor involvement in fentanyl-mediated respiratory depression. Eur J Pharmacol 1996; 312:241-4. [PMID: 8894602 DOI: 10.1016/0014-2999(96)00571-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Several fentanyl analogs (Bagley et al., 1989, J. Med. Chem. 32, 663) were compared to fentanyl and morphine for their effects on respiratory depression as determined by arterial blood gas (pH, pCO2 and pO2) measurements. Fentanyl (0.1 mg/kg), morphine (10 mg/kg), #16 (1-phenethyl-4-[N-(pyridin-2-yl)-N-(methoxymethylcarbonyl)amino] piperidine, 1 mg/kg), #17 (1-phenethyl-4-[N-(pyridin-2-yl) -N-(2-furoyl)amino]piperidine, 0.5 mg/kg) and #29 (1-phenethyl-4-[N- (pyrimidin-2-yl)-N-(methoxy-methylcarbonyl) amino]piperidine, 10 mg/kg) produced significant respiratory depression in rats. Pretreatment with the mu1-opioid receptor selective antagonist, naloxonazine (10 mg/kg), blocked the respiratory effect of fentanyl and its analogs, but not that of morphine. The results suggest that the mu1-opioid receptor plays an important role in the respiratory effects of fentanyl and its analogs. Hence, the mechanism of fentanyl-induced respiratory depression appears to be distinct from that produced by morphine. The most likely explanation for this difference is the possible contribution of muscle rigidity and catalepsy to the observed changes in blood gas parameters caused by the fentanyl analogs, while the respiratory depression of morphine, measured by these same parameters, appears to be independent of its effect on muscle rigidity.
Collapse
Affiliation(s)
- S W Chen
- Molecular Research Institute, Palo Alto, CA 94304, USA
| | | | | | | | | |
Collapse
|
37
|
Tokuyama S, Nakamura F, Nakao K, Takahashi M, Kaneto H. A potent mu-opioid receptor agonist, dihydroetorphine, fails to produce the conditioned place preference in mice. JAPANESE JOURNAL OF PHARMACOLOGY 1996; 71:357-60. [PMID: 8886936 DOI: 10.1254/jjp.71.357] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Reinforcing effects of dihydroetorphine (DHE) and morphine were evaluated by the conditioned place preference paradigm. Both DHE and morphine produced an antinociceptive effect in a dose-dependent manner. On the other hand, DHE (0.1, 1, 3 and 10 micrograms/kg, i.p.) failed to induce the conditioned preference, while morphine (0.1, 1, 3 and 10 mg/kg, i.p.) caused a dose-dependent preference for the drug-paired place. Thus, these characteristic properties of DHE make it attractive for development as a novel potent analgesic compound that has less dependence liability.
Collapse
Affiliation(s)
- S Tokuyama
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Nagasaki University, Japan
| | | | | | | | | |
Collapse
|
38
|
Gatch MB, Liguori A, Negus SS, Mello NK, Bergman J, Liguori T. Naloxonazine antagonism of levorphanol-induced antinociception and respiratory depression in rhesus monkeys. Eur J Pharmacol 1996; 298:31-6. [PMID: 8867916 DOI: 10.1016/0014-2999(95)00769-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The mu-opioid receptor antagonist effects of naloxonazine on levorphanol-induced thermal antinociception and respiratory depression were examined in rhesus monkeys. Levorphanol (0.032-3.2 mg/kg) produced dose-dependent increases in tail-withdrawal latencies from 50 degrees C water in a warm-water tail-withdrawal assay and dose-dependent decreases in ventilation in both air and 5% CO2 mixed in air. Naloxonazine (0.1-3.0 mg/kg) antagonized both the antinociceptive and ventilatory effects of levorphanol to a similar degree, and the antagonist effects of naloxonazine were greater after 1 h than after 24 h. Under all conditions, the antagonist effects of naloxonazine were fully surmountable. Schild analysis of the antagonist effects of naloxonazine after 1 h pretreatment in the antinociception assay yielded a pA2 value of 7.6 and a slope of -0.50; by comparison, quadazocine yielded a pA2 value of 7.5 and a slope of -1.05. These results suggest that naloxonazine acts as a potent and fully reversible mu-opioid receptor antagonist with a moderately long duration of action in rhesus monkeys. In addition, these results suggest that the antinociceptive and ventilatory effects of mu-opioid receptor agonists in rhesus monkeys are mediated by pharmacologically similar populations of mu opioid receptors.
Collapse
Affiliation(s)
- M B Gatch
- Alcohol and Drug Abuse Research Center, McLean Hospital, Belmont, MA 02178, USA
| | | | | | | | | | | |
Collapse
|
39
|
Opioids and the Control of Pain. Neurotherapeutics 1996. [DOI: 10.1007/978-1-59259-466-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
40
|
|
41
|
Noble F, Roques BP. Assessment of endogenous enkephalins efficacy in the hot plate test in mice: comparative study with morphine. Neurosci Lett 1995; 185:75-8. [PMID: 7746508 DOI: 10.1016/0304-3940(94)11228-b] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The in vivo spare receptor population and the relative efficacies of morphine and the endogenous enkephalins to alleviate thermal nociceptive inputs were compared by using the mu irreversible antagonist beta-funaltrexamine (beta-FNA). Twenty-four hours after i.c.v. administration of beta-FNA at increasing concentrations (0.005-2.5 micrograms), parallel rightward shifts of both morphine and RB 101 (mixed enkephalin-degrading-enzyme inhibitor) dose-response curves, were observed, but the concentration of beta-FNA required to reduce the analgesic responses was about 10 times higher for RB 101 (0.1 microgram) than for morphine (0.01 microgram). The preferential involvement of mu receptors in the analgesic responses obtained after beta-FNA pretreatment, was supported by the inability of the delta-selective antagonist naltrindole to block these effects. In conclusion, it seems that to elicit the same antinociceptive responses, enkephalins could occupy a smaller proportion of mu opioid receptors than morphine, suggesting that the endogenous peptides have a higher efficacy.
Collapse
Affiliation(s)
- F Noble
- Département de Pharmacochimie Moléculaire et Structurale, U266 INSERM--URA D1500 CNRS, Université René Descartes, UFR des Sciences Pharmaceutiques et Biologiques, Paris, France
| | | |
Collapse
|
42
|
Press JB, Raffa RB. Patent Update Central & Peripheral Nervous System: Recent advances in opioid and non-opioid analgesia (1992–1993). Expert Opin Ther Pat 1994. [DOI: 10.1517/13543776.4.4.379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Connelly CD, Martinez RP, Schupsky JJ, Porreca F, Raffa RB. Etonitazene-induced antinociception in mu1 opioid receptor deficient CXBK mice: evidence for a role for mu2 receptors in supraspinal antinociception. Life Sci 1994; 54:PL369-74. [PMID: 7910928 DOI: 10.1016/0024-3205(94)90037-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The prevailing view is that supraspinal mu opioid-mediated antinociception in mice is mediated via the mu 1 subtype. The purpose of the present study was to determine if the highly mu-selective compound etonitazene could produce supraspinal (intracerebroventricular; i.c.v.) antinociception in CXBK mice, which are deficient in brain mu1, but not mu2, opioid receptors. CXBK or normal Crl:CD-1 (ICR)BR mice were administered graded doses of etonitazene i.c.v. and 15 min later antinociception was assessed by a standard radiant-heat or 55 degrees C water tail-flick test. Etonitazene produced dose-related antinociception that was blocked by naloxone and by beta-FNA (demonstrating a mu opioid mechanism), but not by either ICI-174,864 or naltrindole (demonstrating the lack of involvement of delta opioid receptors). These findings suggest that mu2 opioid receptors are important contributors to opioid-induced supraspinal antinociception in mice.
Collapse
MESH Headings
- Animals
- Benzimidazoles/administration & dosage
- Benzimidazoles/pharmacology
- Cerebral Ventricles/drug effects
- Cerebral Ventricles/physiology
- Dose-Response Relationship, Drug
- Enkephalin, Leucine/administration & dosage
- Enkephalin, Leucine/analogs & derivatives
- Enkephalin, Leucine/pharmacology
- Hot Temperature
- Injections, Intraventricular
- Injections, Subcutaneous
- Male
- Mice
- Mice, Inbred ICR
- Mice, Mutant Strains
- Naloxone/administration & dosage
- Naloxone/pharmacology
- Naltrexone/administration & dosage
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Pain/physiopathology
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/physiology
- Spinal Cord/drug effects
- Spinal Cord/physiology
Collapse
Affiliation(s)
- C D Connelly
- R.W. Johnson Pharmaceutical Research Institute, Spring House, PA 19477
| | | | | | | | | |
Collapse
|
44
|
Raffa RB, Schupsky JJ. Opioid mu receptor subtypes (possibly mu 1 and mu 2) revealed by morphine-induced antinociception vs endothelin-1 in recombinant inbred CXBK mice. Life Sci 1994; 54:PL57-62. [PMID: 8289581 DOI: 10.1016/0024-3205(94)00822-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Morphine was administered intracerebroventricularly to normal or recombinant inbred CXBK (mu-opioid receptor deficient) mice and antinociception was determined against two different stimuli. Morphine-induced antinociception against acetylcholine was strain-dependent, whereas against endothelin-1 it was not. The antinociception was mediated via opioid mu receptors (blocked by beta-FNA, but not naltrindole, ICI 174,864 or nor-BNI) through separate pathways, one naloxonazine-sensitive and the other naloxonazine-insensitive. Taken together, these results appear to demonstrate supraspinal morphine-induced antinociception through distinct subtypes of the mu opioid receptor, supporting the possibility of novel subtype-selective therapeutic agents with greater separation between analgesia and side-effects or physical dependence. Furthermore, the methodology described herein provides model systems for the in vivo screening of such agents.
Collapse
Affiliation(s)
- R B Raffa
- Drug Discovery, The R. W. Johnson Pharmaceutical Research Institute, Spring House, PA 19477-0776
| | | |
Collapse
|
45
|
Suzuki T, Funada M, Narita M, Misawa M, Nagase H. Morphine-induced place preference in the CXBK mouse: characteristics of mu opioid receptor subtypes. Brain Res 1993; 602:45-52. [PMID: 8383571 DOI: 10.1016/0006-8993(93)90239-j] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The role of mu opioid receptor subtypes, mu 1 and mu 2, in morphine-conditioned place preference was examined using ddY and mu 1 opioid receptor-deficient CXBK mice. In ddY mice, the mu receptor agonist morphine caused a dose-related preference for the drug-associated place, but the kappa agonist U-50,488H produced a dose-related place aversion. These results demonstrated that the mouse is available for place preference conditioning using opioids. Under this condition, the influence of pretreatment with the selective mu 1 opioid receptor antagonist naloxonazine on morphine-induced place preference was investigated in ddY mice. Although pretreatment with the selective mu 1 antagonist naloxonazine (35 mg/kg, s.c.) did not modify the morphine-induced place preference, pretreatment with the selective mu antagonist beta-funaltrexamine (beta-FNA 10 mg/kg, s.c.) eliminated the appetitive effect of morphine. Furthermore, morphine (1-5 mg/kg, s.c.) produced a dose-related preference for the drug-associated place in CXBK mice. These findings suggest that the morphine-induced conditioned place preference may be mediated by naloxonazine-insensitive sites (mu 2 opioid receptors). In addition, chronic infusion of the dopamine D1 antagonist SCH23390 (1.0 mg/kg/day) during the conditioning sessions eliminated the morphine-induced place preference in CXBK mice. Similarly, morphine combined with naloxonazine failed to produce the place preference in ddY mice chronically treated with SCH23390. The blocking effect of SCH23390 on the morphine-conditioned place preference suggests that mu 2 receptors may regulate the dopaminergic system, especially dopamine D1 receptors, and are also involved in the reinforcing effects of morphine.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer
- Animals
- Appetitive Behavior/drug effects
- Benzazepines/pharmacology
- Choice Behavior/drug effects
- Conditioning, Classical/drug effects
- Dose-Response Relationship, Drug
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Morphine/antagonists & inhibitors
- Morphine/pharmacology
- Motivation
- Naloxone/analogs & derivatives
- Naloxone/pharmacology
- Pyrrolidines/pharmacology
- Receptors, Dopamine D1/antagonists & inhibitors
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/physiology
- Sodium Chloride/pharmacology
Collapse
Affiliation(s)
- T Suzuki
- Department of Pharmacology, Hoshi University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
46
|
Cheng PY, Wu D, Decena J, Soong Y, McCabe S, Szeto HH. Opioid-induced stimulation of fetal respiratory activity by [D-Ala2]deltorphin I. Eur J Pharmacol 1993; 230:85-8. [PMID: 8381356 DOI: 10.1016/0014-2999(93)90413-c] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
[D-Ala2]deltorphin I effects on fetal respiratory activity was characterized to determine the role delta-opioid receptors play in modulating fetal respiratory activity. [D-Ala2]deltorphin I, infused at 0.3 or 100 micrograms/h, intracerebroventricularly (i.c.v.), stimulated fetal respiratory activity without changing blood pH, PCO2 or PO2. Stimulation by 0.3 micrograms/h, but not 100 micrograms/h, was blocked by i.c.v. infusion of the delta-opioid receptor antagonist, naltrindole. Stimulation by 100 micrograms/h was blocked by the mu 1-opioid receptor antagonist naloxonazine. These data suggest stimulation of fetal respiratory activity by 0.3 micrograms/h [D-Ala2]deltorphin I are mediated specifically through delta-opioid receptors; while [D-Ala2]deltorphin I at 100 micrograms/h is no longer selective for the delta-opioid receptor, and the stimulation may be mediated through the mu 1-opioid receptor.
Collapse
Affiliation(s)
- P Y Cheng
- Department of Pharmacology, Cornell University Medical College, New York, NY 10021
| | | | | | | | | | | |
Collapse
|
47
|
Suzuki T, Hayashi Y, Misawa M. The role of mu1 receptor in physical dependence on morphine using the mu receptor deficient CXBK mouse. Life Sci 1992; 50:849-56. [PMID: 1312192 DOI: 10.1016/0024-3205(92)90203-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
It is known that the CXBK inbred strain of mouse is deficient in mu1 opioid receptors, whereas the strain has a delta opioid receptor population that is less consistently altered. In the present study, we compared physical dependence on morphine between CXBK and C57BL/6 mice. Both strains of mice were treated with morphine-admixed food for 5 days. During the treatment, the two strains of mice showed no signs of toxicity. There was no significant difference in morphine intake during the treatment between CXBK and C57BL/6 mice. After the treatment, the withdrawal was precipitated by injecting naloxone (0.01-30 mg/kg, s.c.). CXBK mice showed weight loss, diarrhea and ptosis, but not jumping and body shakes after low dose of naloxone. Whereas, C57BL/6 mice showed weight loss, diarrhea, ptosis, body shakes and jumping. These results suggest that naloxone-precipitated weight loss, diarrhea and ptosis may be mediated by mu2 and/or delta opioid receptor, while naloxone-precipitated jumping and body shakes may be mediated by mu1 opioid receptors.
Collapse
Affiliation(s)
- T Suzuki
- Department of Applied Pharmacology, School of Pharmacy, Hoshi University, Japan
| | | | | |
Collapse
|
48
|
Hucks D, Thompson PI, McLoughlin L, Joel SP, Patel N, Grossman A, Rees LH, Slevin ML. Explanation at the opioid receptor level for differing toxicity of morphine and morphine 6-glucuronide. Br J Cancer 1992; 65:122-6. [PMID: 1310249 PMCID: PMC1977344 DOI: 10.1038/bjc.1992.23] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The radiolabelled opioid receptor binding affinities of morphine and its active metabolite morphine 6-glucuronide at the total mu, mu 1, mu 2 and delta receptors were determined. Morphine 6-glucuronide was found to have a 4-fold lower affinity for the mu 2 receptor (IC50 17 nM and 82 nM for morphine and morphine 6-glucuronide respectively, P = 0.01), the receptor postulated to be responsible for mediating the respiratory depression and gastrointestinal effects after morphine. This provides a possible explanation for the reduced respiratory depression and vomiting seen following morphine 6-glucuronide in man. A similar reduction in affinity of morphine 6-glucuronide was seen at the total mu receptor whilst there was no significant difference seen at the mu 1 or delta receptor. Hence the increased analgesic potency of morphine 6-glucuronide over morphine remains unexplained.
Collapse
MESH Headings
- Animals
- Brain/drug effects
- Brain/metabolism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalin, Leucine/analogs & derivatives
- Enkephalin, Leucine/metabolism
- Enkephalins/metabolism
- Kinetics
- Morphine/toxicity
- Morphine Derivatives/toxicity
- Rats
- Rats, Inbred Strains
- Receptors, Opioid/drug effects
- Receptors, Opioid/metabolism
- Receptors, Opioid, delta
- Receptors, Opioid, mu
- Subcellular Fractions/metabolism
Collapse
Affiliation(s)
- D Hucks
- Department of Chemical Endocrinology, St Bartholomew's Hospital, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Baumann MH, Rabii J. Inhibition of suckling-induced prolactin release by mu- and kappa-opioid antagonists. Brain Res 1991; 567:224-30. [PMID: 1667901 DOI: 10.1016/0006-8993(91)90799-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Evidence suggests that endogenous opioid peptides (EOP) are involved in the hyperprolactinemia and suppression of luteinizing hormone (LH) release associated with lactation. To address this hypothesis, we investigated the effects of various opioid receptor antagonists on suckling-induced prolactin (PRL) and LH responses in primiparous, lactating rats. All animals were fitted with indwelling jugular catheters to allow serial blood sampling, and some rats received intracerebroventricular (i.c.v.) cannulae for central drug injection. Naloxone (2.0 mg/kg, i.v.) was employed as a broad spectrum opioid antagonist, whereas beta-funaltrexamine (beta-FNA, 1.0-5.0 micrograms, i.c.v.), naloxonazine (NAZ, 20 mg/kg, i.v.) and nor-binaltorphimine (nor-BNI, 4.0-16.0 micrograms, i.c.v.) were used to block mu, mu 1 and kappa receptor sites, respectively. In vehicle-treated rats, pup suckling evoked a dramatic increase in plasma PRL and a concurrent decrease in circulating LH. Naloxone caused a modest, though significant, attenuation of the PRL surge during nursing. beta-FNA and nor-BNI inhibited suckling-induced PRL release in a dose-related fashion, and at sufficient doses, both antagonists abolished the PRL response. Conversely, the suckling-induced rise in plasma PRL was not affected by NAZ. Naloxone, beta-FNA, and NAZ did not alter the profile of circulating LH in suckled rats, but the highest dose nor-BNI (16 micrograms, i.c.v.) produced a significant elevation in plasma LH. However, even in rats treated with 16.0 micrograms of nor-BNI, plasma LH levels declined in response to the nursing stimulus.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- M H Baumann
- Department of Biological Sciences, Rutgers University, Piscataway, NJ 08855
| | | |
Collapse
|
50
|
Fanselow MS, Kim JJ, Young SL, Calcagnetti DJ, DeCola JP, Helmstetter FJ, Landeira-Fernandez J. Differential effects of selective opioid peptide antagonists on the acquisition of pavlovian fear conditioning. Peptides 1991; 12:1033-7. [PMID: 1686930 DOI: 10.1016/0196-9781(91)90056-u] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pretreatment with opioid antagonists enhances acquisition of Pavlovian fear conditioning. The present experiments attempted to characterize the type of opioid receptor responsible for this effect using a procedure that assessed the fear of rats to a chamber previously associated with electric shock (1 mA, 0.75 s). Freezing, a species-typical immobility, was employed as an index of fear. Two mu opioid antagonists, CTOP (40 ng) and naloxonazine (10 micrograms), enhanced conditioning. On the other hand, the kappa antagonist nor-binaltorphimine reduced conditioning. Two delta antagonist treatments (16-methyl cyprenorphine and naltrindole) had no reliable effect on acquisition. Thus the enhancement of conditioning appears to be mediated by mu receptors. Previous research has shown that the conditional fear produced by these procedures caused an analgesia that is also mediated by mu receptors. It is argued that the enhancement effect occurs because of an antagonism of this analgesia and that the analgesia normally acts to regulate the level of fear conditioning.
Collapse
Affiliation(s)
- M S Fanselow
- Department of Psychology, University of California, Los Angeles 90024
| | | | | | | | | | | | | |
Collapse
|