1
|
Lobe MMM, Verma S, Patil VM, Iyer MR. A review of kappa opioid receptor antagonists and their clinical trial landscape. Eur J Med Chem 2025; 287:117205. [PMID: 39893986 DOI: 10.1016/j.ejmech.2024.117205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 02/04/2025]
Abstract
Myriad signaling pathways are implicated in neuropsychiatric disorders, yet many mechanisms are unknown and current treatment options are limited. The intriguing dynorphin/kappa opioid receptor (KOR) system that is widely distributed throughout the brain appears to be essential in regulating many physiological and pathophysiological processes. This review explores up to date advances on the relationship between the dynorphin/KOR system with a particular focus on the KOR antagonist compounds tested as clinical candidates that could offer potential treatment options for CNS disorders.
Collapse
Affiliation(s)
- Maloba M M Lobe
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Saroj Verma
- Department of Pharmacy, School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram, Haryana, India
| | - Vaishali M Patil
- Charak School of Pharmacy, Chaudhary Charan Singh University, Bharat, Meerut, Uttar Pradesh, 250004, India
| | - Malliga R Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA.
| |
Collapse
|
2
|
Bodnar RJ. A 40-year analysis of central neuroanatomical and neurochemical circuits mediating homeostatic intake and hedonic intake and preferences in rodents. Brain Res 2025; 1857:149604. [PMID: 40180145 DOI: 10.1016/j.brainres.2025.149604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/05/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
This perspective review was written in response to the celebration of the 60th anniversary of the journal, Brain Research, and covers the evolving focus of my laboratory's work over 40 years in the neurobiological substrates of ingestive behavior in rodents. Following our initial work examining the effects of systemic and ventricular administration of general and selective opioid receptor agonists and antagonists on food intake under spontaneous, deprivation, glucoprivic and hedonic conditions, my laboratory in close collaboration with Drs. Gavril Pasternak and Ying-Xian Pan utilized an antisense oligodoxynucleotide knock-down technique affecting MOR-1, DOR-1, KOR-1 and ORL-1 genes as well as against G-protein subunits to study receptor mediation of opioid receptor agonist-induced feeding as well as feeding following regulatory challenges. Our laboratory employed intracerebral microinjection techniques to map limbic nucleus accumbens and ventral tegmental area central brain circuits mediating homeostatic and hedonic feeding responses through the use of selective mu, delta1, delta2 and kappa opioid receptor subtype agonists in combination with general and selective opioid, dopamineric, glutamatergic and GABAergic antagonists administered into the same site or the reciprocal site, allowing for the identification of a distributed brain network mediating these ingestive effects. Our laboratory in close collaboration with Dr. Anthony Sclafani then focused on the pharmacological, neuroanatomical and learning mechanisms related to the development of sugar- (sucrose, glucose and fructose) and fat- (corn oil) conditioned flavor preferences (CFP) in rats, and on murine genetic variance in food intake, preferences and the process of appetition.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology, Queens College, and Psychology Doctoral Program, The Graduate Center, City University of New York, United States.
| |
Collapse
|
3
|
Tran LT, Freeman KT, Lunzer MM, Portoghese PS, Haskell-Luevano C. Recommended Opioid Receptor Tool Compounds: Comparative In Vitro for Receptor Selectivity Profiles and In Vivo for Pharmacological Antinociceptive Profiles. ACS Pharmacol Transl Sci 2025; 8:225-244. [PMID: 39816790 PMCID: PMC11729433 DOI: 10.1021/acsptsci.4c00604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/18/2025]
Abstract
Opioid agonist ligands bind opioid receptors and stimulate downstream signaling cascades for various biological processes including pain and reward. Historically, before cloning the receptors, muscle contraction assays using isolated organ tissues were used followed by radiolabel ligand binding assays on native tissues. Upon cloning of the opioid G protein-coupled receptors (GPCRs), cell assays using transfected opioid receptor DNA plasmids became the standard practice including 35S-GTPγS functional and cAMP based assays. A number of research laboratories have studied key "tool" reference opioid receptor ligands for decades and used them as control reference compounds. Some, but not all, of these commonly used tool compounds have been characterized and compared side by side in parallel assays for selectivity profiles at the different human opioid receptors isoforms. Herein, we performed the standard FLIPR calcium mobilization assay using HEK293 cells engineered to stably express the GαΔ6qi4myr in parallel, at human MOR, KOR, DOR, and NOP opioid receptors. The following tool compounds: morphine, fentanyl, oxycodone, DAMGO, DPDPE, U69593, deltorphin II, and nociceptin, were examined herein. These included the substance use disorder (SUD) compounds morphine, fentanyl, and oxycodone. Additionally, the antagonist tool compounds naloxone, NTI, norBNI, and β-FNA were assayed in parallel at the human MOR, KOR, DOR, and NOP opioid receptors. Furthermore, the agonist tool compounds were tested in the same in vivo tail-flick antinociception assays via intrathecal injection for ED50 potencies. These data provide both in vitro comparative pharmacology as a reference for cellular activities and in vivo antinociception profiles for these tool compounds.
Collapse
Affiliation(s)
- Linh T. Tran
- Department
of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Katie T. Freeman
- Department
of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary M. Lunzer
- Department
of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Philip S. Portoghese
- Department
of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carrie Haskell-Luevano
- Department
of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
Bossert JM, Caldwell KE, Korah H, Batista A, Bonbrest H, Fredriksson I, Jackson SN, Sulima A, Rice KC, Zaveri NT, Shaham Y. Effect of chronic delivery of the NOP/MOR partial agonist AT-201 and NOP antagonist J-113397 on heroin relapse in a rat model of opioid maintenance. Psychopharmacology (Berl) 2024; 241:2497-2511. [PMID: 39269500 PMCID: PMC11569015 DOI: 10.1007/s00213-024-06678-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024]
Abstract
RATIONALE The opioid crisis persists despite availability of effective opioid agonist maintenance treatments (methadone and buprenorphine). Thus, there is a need to advance novel medications for the treatment of opioid use and relapse. OBJECTIVES We recently modeled maintenance treatment in rats and found that chronic delivery of buprenorphine and the mu opioid receptor (MOR) partial agonist TRV130 decreases relapse to oxycodone seeking and taking. In contrast, chronic delivery of the buprenorphine analog BU08028 had mixed effects on different heroin relapse-related measures. Here, we tested the effect of the mixed nociceptin (NOP) receptor/MOR partial agonist AT-201 and the NOP receptor antagonist J-113397 on different heroin relapse-related measures. METHODS We trained male and female rats to self-administer heroin (6-h/d, 14-d) in context A and then implanted osmotic minipumps containing AT-201 (0, 3.8, or 12 mg/kg/d) or J-113397 (0, 12.6, or 40 mg/kg/d). Next, we tested the effect of chronic delivery of the compounds on (1) incubation of heroin seeking in a non-drug context B, (2) extinction responding reinforced by heroin-associated discrete cues in context B, (3) context A-induced reinstatement of heroin seeking, and (4) reacquisition of heroin self-administration in context A. RESULTS In females, AT-201 modestly increased reacquisition of heroin self-administration and J-113397 modestly decreased incubation of heroin seeking. The compounds had no effect on the other relapse-related measures in females, and no effect on any of the measures in males. CONCLUSION The NOP/MOR partial agonist AT-201 and the NOP antagonist J-113397 did not mimic buprenorphine's inhibitory effects on relapse in a rat model of opioid maintenance treatment.
Collapse
MESH Headings
- Animals
- Male
- Female
- Rats
- Nociceptin Receptor
- Self Administration
- Receptors, Opioid/metabolism
- Receptors, Opioid/agonists
- Receptors, Opioid, mu/agonists
- Heroin/administration & dosage
- Recurrence
- Heroin Dependence/drug therapy
- Narcotic Antagonists/pharmacology
- Narcotic Antagonists/administration & dosage
- Piperidines/pharmacology
- Piperidines/administration & dosage
- Disease Models, Animal
- Rats, Sprague-Dawley
- Drug-Seeking Behavior/drug effects
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacology
- Pyrimidines/pharmacology
- Pyrimidines/administration & dosage
- Extinction, Psychological/drug effects
- Dose-Response Relationship, Drug
- Benzimidazoles
Collapse
Affiliation(s)
| | - Kiera E Caldwell
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Hannah Korah
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Ashley Batista
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Hannah Bonbrest
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Ida Fredriksson
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | | | - Agnieszka Sulima
- Molecular Targets and Medications Discovery Branch, IRP/NIDA, NIAAA/NIH, Baltimore, MD, U.S.A
| | - Kenner C Rice
- Molecular Targets and Medications Discovery Branch, IRP/NIDA, NIAAA/NIH, Baltimore, MD, U.S.A
| | | | - Yavin Shaham
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| |
Collapse
|
5
|
Calvo F, Dos Anjos-Garcia T, Paschoalin-Maurin T, Bazaglia-de-Sousa G, de Paula Rodrigues BM, Lobão-Soares B, Almada RC, Wotjak CT, Coimbra NC. Kappa-opioid receptor blockade in the inferior colliculus of prey threatened by pit vipers decreases anxiety and panic-like behaviour. Acta Neuropsychiatr 2024:1-13. [PMID: 39370934 DOI: 10.1017/neu.2024.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The dorsal midbrain comprises dorsal columns of the periaqueductal grey matter and corpora quadrigemina. These structures are rich in beta-endorphinergic and leu-enkephalinergic neurons and receive GABAergic inputs from substantia nigra pars reticulata. Although the inferior colliculus (IC) is mainly involved in the acoustic pathways, the electrical and chemical stimulation of central and pericentral nuclei of the IC elicits a vigorous defensive behaviour. The defensive immobility and escape elicited by IC activation is commonly related to panic-like emotional states. To investigate the role of κ-opioid receptor of the IC in the antiaversive effects of endogenous opioid receptor blockade in a dangerous situation, male Wistar rats were pretreated in the IC with the κ-opioid receptor-selective antagonist nor-binaltorphimine at different concentrations and submitted to the non-enriched polygonal arena for a snake panic test in the presence of a rattlesnake and, after 24 h, prey were resubmitted to the experimental context. The snakes elicited in prey a set of antipredatory behaviours, such as the anxiety-like responses of defensive attention and risk assessment, and the panic-like reactions of defensive immobility and either escape or active avoidance during the elaboration of unconditioned and conditioned fear-related responses. Pretreatment of the IC with microinjections of nor-binaltorphimine at higher concentrations significantly decreased the frequency and duration of both anxiety- and panic-attack-like behaviours. These findings suggest that κ-opioid receptor blockade in the IC causes anxiolytic- and panicolytic-like responses in threatening conditions, and that kappa-opioid receptor-selective antagonists can be a putative coadjutant treatment for panic syndrome treatment.
Collapse
Affiliation(s)
- Fabrício Calvo
- Laboratory of Neuroanatomy & Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Ophidiarium LNN-FMRP-USP/INeC, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Tayllon Dos Anjos-Garcia
- Laboratory of Neuroanatomy & Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Behavioural Neurosciences Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- Ophidiarium LNN-FMRP-USP/INeC, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Department Physiological Sciences, Institute for Biomedical Sciences, Alfenas Federal University (ICB-UNIFAL), Alfenas, Minas Gerais, Brazil
| | - Tatiana Paschoalin-Maurin
- Laboratory of Neuroanatomy & Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Ophidiarium LNN-FMRP-USP/INeC, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Guilherme Bazaglia-de-Sousa
- Laboratory of Neuroanatomy & Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Behavioural Neurosciences Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- Ophidiarium LNN-FMRP-USP/INeC, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Bruno Mangili de Paula Rodrigues
- Laboratory of Neuroanatomy & Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Behavioural Neurosciences Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Ophidiarium LNN-FMRP-USP/INeC, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Bruno Lobão-Soares
- Laboratory of Neuroanatomy & Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte (UFRN), Natal (RN), Brazil
| | - Rafael Carvalho Almada
- Laboratory of Neuroanatomy & Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Laboratory of Neurobiology and Neurobiotechnology, Department of Biological Sciences, School of Science, Humanities and Languages, São Paulo State University (UNESP), Assis, São Paulo, Brazil
- Behavioural Neurosciences Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- Ophidiarium LNN-FMRP-USP/INeC, Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Carsten T Wotjak
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Laboratory of Neuronal Plasticity, Munich, Germany
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharmaceuticals Gesellschaft mit Beschränkter Haftung & Compagnie Kommanditgesellschaft, Biberach an der Riß, Germany
| | - Norberto Cysne Coimbra
- Laboratory of Neuroanatomy & Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Behavioural Neurosciences Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
6
|
Sandoval-Caballero C, Jara J, Luarte L, Jiménez Y, Teske JA, Perez-Leighton C. Control of motivation for sucrose in the paraventricular hypothalamic nucleus by dynorphin peptides and the kappa opioid receptor. Appetite 2024; 200:107504. [PMID: 38768926 DOI: 10.1016/j.appet.2024.107504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/14/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
The dynorphin peptides are the endogenous ligands for the kappa opioid receptor (KOR) and regulate food intake. Administration of dynorphin-A1-13 (DYN) in the paraventricular hypothalamic nucleus (PVN) increases palatable food intake, and this effect is blocked by co-administration of the orexin-A neuropeptide, which is co-released with DYN in PVN from neurons located in the lateral hypothalamus. While PVN administration of DYN increases palatable food intake, whether it increases food-seeking behaviors has yet to be examined. We tested the effects of DYN and norBNI (a KOR antagonist) on the seeking and consumption of sucrose using a progressive ratio (PR) and demand curve (DC) tasks. In PVN, DYN did not alter the sucrose breaking point (BP) in the PR task nor the elasticity or intensity of demand for sucrose in the DC task. Still, DYN reduced the delay in obtaining sucrose and increased licks during sucrose intake in the PR task, irrespective of the co-administration of orexin-A. In PVN, norBNI increased the delay in obtaining sucrose and reduced licks during sucrose intake in the PR task while increasing elasticity without altering intensity of demand in the DC task. However, subcutaneous norBNI reduced the BP for sucrose and increased the delay in obtaining sucrose in the PR task while reducing the elasticity of demand. Together, these data show different effects of systemic and PVN blockade of KOR on food-seeking, consummatory behaviors, and incentive motivation for sucrose and suggest that KOR activity in PVN is necessary but not sufficient to drive seeking behaviors for palatable food.
Collapse
Affiliation(s)
- C Sandoval-Caballero
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J Jara
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - L Luarte
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Y Jiménez
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J A Teske
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, Arizona, USA
| | - C Perez-Leighton
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
7
|
Shaykin JD, Denehy ED, Martin JR, Chandler CM, Luo D, Taylor CE, Sunshine MD, Turner JR, Alilain WJ, Prisinzano TE, Bardo MT. Targeting α 1- and α 2-adrenergic receptors as a countermeasure for fentanyl-induced locomotor and ventilatory depression. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 110:104527. [PMID: 39106924 PMCID: PMC11423298 DOI: 10.1016/j.etap.2024.104527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/12/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
This study assessed the ability of α1 and α2-adrenergic drugs to decrease fentanyl-induced locomotor and ventilatory depression. Rats were given saline or fentanyl, followed by: (1) naltrexone, (2) naloxone, (3) nalmefene, (4) α1 agonist phenylephrine, (5) α1 antagonist prazosin, (6) α1D antagonist BMY-7378, (7) α2 agonist clonidine, (8) α2 antagonist yohimbine or (9) vehicle. All µ-opioid antagonists dose-dependently reversed fentanyl-induced locomotor and ventilatory depression. While the α1 drugs did not alter the effects of fentanyl, clonidine dose-dependently decreased locomotion and respiration with and without fentanyl. Conversely, yohimbine given at a low dose (0.3-1 mg/kg) stimulated ventilation when given alone and higher doses (>1 mg/kg) partially reversed (∼50 %) fentanyl-induced ventilatory depression, but not locomotor depression. High doses of yohimbine in combination with a suboptimal dose of naltrexone reversed fentanyl-induced ventilatory depression, suggestive of additivity. Yohimbine may serve as an effective adjunctive countermeasure agent combined with naltrexone to rescue fentanyl-induced ventilatory depression.
Collapse
Affiliation(s)
- Jakob D Shaykin
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - Emily D Denehy
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - Jocelyn R Martin
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - Cassie M Chandler
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - Dan Luo
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Chase E Taylor
- Spinal Cord and Brain Injury Research Center (SCoBIRC) and Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Michael D Sunshine
- Spinal Cord and Brain Injury Research Center (SCoBIRC) and Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Jill R Turner
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Warren J Alilain
- Spinal Cord and Brain Injury Research Center (SCoBIRC) and Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | | | - Michael T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
8
|
Wang XM, Li XX, Jiang B, Wang TQ, Guo Z. Morphine timing-dependent modulation of TRPV1 phosphorylation correlates with differential morphine effects on myocardial ischemia/reperfusion injury. Eur J Pharmacol 2024; 975:176648. [PMID: 38759706 DOI: 10.1016/j.ejphar.2024.176648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Opioids are used for pain relief in patients suffering from acute myocardial ischemia or infarction. Clinical and laboratory studies demonstrate that morphine treated patients or the experimental animal model suffering acute myocardial ischemia and reperfusion, may worsen myocardial viability. As transient receptor potential vanilloid 1 (TRPV1) plays important roles in pain sensation and cardio-protection, we query whether opioids may exacerbate myocardial viability via interaction with TRPV1 activity in the pain relief. We found the co-expressions of TRPV1 and opioid μ, δ and κ receptors in adult rat cardiomyocytes. Intravenous injection of morphine (0.3 mg/kg) at 20 min after induction of myocardial ischemia, in the rat model of acute myocardial ischemia and reperfusion, induced significant reduction of phosphorylated TRPV1 (p-TRPV1) in the ventricular myocardium and increase in serum cardiac troponin I (cTnI), compared with the ischemia/reperfusion controls (all P < 0.05). The effects of morphine were completely reversed by selective opioid μ, δ and κ receptor antagonists. While significant upregulation of p-TRPV1 (P < 0.05) and improvement of ±dP/dt max (all P < 0.05) were detected in the animals giving the same dose of morphine before induction of myocardial ischemia. The changes in p-TRPV1 correlate with the alterations of cTnI (r = -0.5840, P = 0.0283) and ±dP/dt max (r = 0.8084, P = 0.0005 and r = -0.8133, P = 0.0004, respectively). The findings of this study may indicate that potentiation and attenuation of TRPV1 sensitivity correlate with the improvement of the cardiac performance and the aggravation of myocardial viability, respectively, by giving morphine before and during myocardial ischemia and reperfusion.
Collapse
Affiliation(s)
- Xin-Meng Wang
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Xiao-Xi Li
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China; Department of Anaesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Bo Jiang
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Tian-Qi Wang
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Zheng Guo
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China; Department of Anaesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), National Education Commission, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
9
|
Kajino K, Tokuda A, Saitoh T. Morphinan Evolution: The Impact of Advances in Biochemistry and Molecular Biology. J Biochem 2024; 175:337-355. [PMID: 38382631 DOI: 10.1093/jb/mvae021] [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: 11/05/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024] Open
Abstract
Morphinan-based opioids, derived from natural alkaloids like morphine, codeine and thebaine, have long been pivotal in managing severe pain. However, their clinical utility is marred by significant side effects and high addiction potential. This review traces the evolution of the morphinan scaffold in light of advancements in biochemistry and molecular biology, which have expanded our understanding of opioid receptor pharmacology. We explore the development of semi-synthetic and synthetic morphinans, their receptor selectivity and the emergence of biased agonism as a strategy to dissociate analgesic properties from undesirable effects. By examining the molecular intricacies of opioid receptors and their signaling pathways, we highlight how receptor-type selectivity and signaling bias have informed the design of novel analgesics. This synthesis of historical and contemporary perspectives provides an overview of the morphinan landscape, underscoring the ongoing efforts to mitigate the problems facing opioids through smarter drug design. We also highlight that most morphinan derivatives show a preference for the G protein pathway, although detailed experimental comparisons are still necessary. This fact underscores the utility of the morphinan skeleton in future opioid drug discovery.
Collapse
Affiliation(s)
- Keita Kajino
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Degree Programs in Pure and Applied Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Akihisa Tokuda
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
10
|
Karaki F, Takamori T, Kawakami K, Sakurai S, Hidaka K, Ishii K, Oki T, Sato N, Atsumi N, Ashizawa K, Taguchi A, Ura A, Naruse T, Hirayama S, Nonaka M, Miyano K, Uezono Y, Fujii H. Discovery of 7-Azanorbornane-Based Dual Agonists for the Delta and Kappa Opioid Receptors through an In Situ Screening Protocol. Molecules 2023; 28:6925. [PMID: 37836768 PMCID: PMC10574725 DOI: 10.3390/molecules28196925] [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: 09/07/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
In medicinal chemistry, the copper-catalyzed click reaction is used to prepare ligand candidates. This reaction is so clean that the bioactivities of the products can be determined without purification. Despite the advantages of this in situ screening protocol, the applicability of this method for transmembrane proteins has not been validated due to the incompatibility with copper catalysts. To address this point, we performed ligand screening for the µ, δ, and κ opioid receptors using this protocol. As we had previously reported the 7-azanorbornane skeleton as a privileged scaffold for the G protein-coupled receptors, we performed the click reactions between various 7-substituted 2-ethynyl-7-azanorbornanes and azides. Screening assays were performed without purification using the CellKeyTM system, and the putative hit compounds were re-synthesized and re-evaluated. Although the "hit" compounds for the µ and the δ receptors were totally inactive after purifications, three of the four "hits" for the κ receptor were true agonists for this receptor and also showed activities for the δ receptor. Although false positive/negative results exist as in other screening projects for soluble proteins, this in situ method is effective in identifying novel ligands for transmembrane proteins.
Collapse
Affiliation(s)
- Fumika Karaki
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Taro Takamori
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Koumei Kawakami
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Sae Sakurai
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kyoko Hidaka
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kei Ishii
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tomoya Oki
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Noriko Sato
- Analytical Unit for Organic Chemistry, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Nao Atsumi
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Department of Pain Control Research, Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Karin Ashizawa
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Department of Pain Control Research, Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Ai Taguchi
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Department of Pain Control Research, Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Asuka Ura
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Department of Pain Control Research, Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Toko Naruse
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Shigeto Hirayama
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Miki Nonaka
- Department of Pain Control Research, Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Kanako Miyano
- Department of Pain Control Research, Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yasuhito Uezono
- Department of Pain Control Research, Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hideaki Fujii
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
11
|
Zhang J, Lu Y, Jia M, Bai Y, Sun L, Dong Z, Tian W, Yin F, Wei S, Wang Y. Kappa opioid receptor in nucleus accumbens regulates depressive-like behaviors following prolonged morphine withdrawal in mice. iScience 2023; 26:107536. [PMID: 37636073 PMCID: PMC10448166 DOI: 10.1016/j.isci.2023.107536] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/12/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Prolonged withdrawal from opioids leads to negative emotions. Kappa opioid receptor (KOR) plays an important role in opioid addiction and affective disorders. However, the underlying mechanism of KOR in withdrawal-related depression is still lacking. We found that escitalopram treatment had a limited effect in improving depression symptoms in heroin-dependent patients. In mice, we demonstrated prolonged (4 weeks) but not acute (24 h) withdrawal from morphine induced depressive-like behaviors. The number of c-Fos positive cells and the expression of KOR in the nucleus accumbens (NAc), were significantly increased in the prolonged morphine withdrawal mice. Conditional KOR knockdown in NAc significantly improved depressive-like behaviors. Repeated but not acute treatment with the KOR antagonist norBNI improved depressive-like behaviors and reversed PSD95, synaptophysin, p-ERK, p-CREB, and BDNF in NAc. This study demonstrated the important role of striatal KOR in morphine withdrawal-related depressive-like behaviors and offered therapeutic potential for the treatment of withdrawal-related depression.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Ye Lu
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Min Jia
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Yuying Bai
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Lulu Sun
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Ziqing Dong
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Wenrong Tian
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Fangyuan Yin
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Shuguang Wei
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Yunpeng Wang
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| |
Collapse
|
12
|
Özkan-Kotiloğlu S, Kaya-Akyüzlü D, Yurdakul R, Yıldırım MA, Özgür-İlhan İ. Effect of PDYN and OPRK1 polymorphisms on the risk of alcohol use disorder and the intensity of depressive symptoms. Alcohol Alcohol 2023:7161013. [PMID: 37177778 DOI: 10.1093/alcalc/agad036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
AIMS The dynorphin (DYN)/Kappa Opioid Receptor (KOR) system has been suggested to be involved in both negative affective states and the action of alcohol. The present study was undertaken to explore whether the DYN/KOR system genes, PDYN and OPRK1, influence on individual differences in the intensity of depressive symptoms at admission as well as the risk of alcohol use disorder (AUD) risk in a sample of 101 individuals with AUD and 100 controls. METHODS PDYN (rs2281285, rs2225749 and rs910080) and OPRK1 (rs6473797, rs963549 and rs997917) polymorphisms were analyzed by PCR-RFLP. The intensity of depressive and anxiety symptoms and craving were measured by the Beck Depression Inventory-II (BDI-II), Beck Anxiety Inventory (BAI), and Penn Alcohol Craving Scale, respectively. RESULTS A significant association between the risk of AUD and OPRK1 rs6473797 (P < 0.05) at the gene level. OPRK1 rs6473797 CC genotype was found to lead to a 3.11 times greater alcohol dependence risk. In addition, the BDI-II score of the OPRK1 rs963549 CC genotype was found to be significantly lower (20.9 ± 11.2, min: 1.0, max: 48.0) than that of the CT + TT genotypes (27.04 ± 12.7, min: 0.0, max: 49.0) (t: -2.332, P = 0.022). None of the PDYN polymorphisms were associated with BDI-II score. CONCLUSION Variations in the KOR are associated with the risk of AUD and the intensity of depressive symptoms at admission at the gene level in Turkish males. On the other hand, PDYN gene seemed not to be associated with AUD, depression, anxiety, and craving.
Collapse
Affiliation(s)
- Selin Özkan-Kotiloğlu
- Department of Molecular Biology and Genetics, Faculty of Science and Art, Kırşehir Ahi Evran University, Kırşehir, 40100, Turkey
| | - Dilek Kaya-Akyüzlü
- Department of Forensic Biology, Institute of Forensic Sciences, Ankara University, Ankara, 06590, Turkey
| | - Rabia Yurdakul
- Department of Forensic Biology, Institute of Forensic Sciences, Ankara University, Ankara, 06590, Turkey
- Department of Interdisciplinary Forensic Sciences, Graduate School of Health Sciences, Ankara University, Ankara, 06110, Turkey
| | - Mukaddes Asena Yıldırım
- Department of Forensic Biology, Institute of Forensic Sciences, Ankara University, Ankara, 06590, Turkey
- Department of Interdisciplinary Forensic Sciences, Graduate School of Health Sciences, Ankara University, Ankara, 06110, Turkey
| | - İnci Özgür-İlhan
- Department of Mental Health and Diseases, Faculty of Medicine, Ankara University, Ankara, 06590, Turkey
| |
Collapse
|
13
|
Farahbakhsh ZZ, Song K, Branthwaite HE, Erickson KR, Mukerjee S, Nolan SO, Siciliano CA. Systemic kappa opioid receptor antagonism accelerates reinforcement learning via augmentation of novelty processing in male mice. Neuropsychopharmacology 2023; 48:857-868. [PMID: 36804487 PMCID: PMC10156709 DOI: 10.1038/s41386-023-01547-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/20/2023] [Accepted: 02/02/2023] [Indexed: 02/19/2023]
Abstract
Selective inhibition of kappa opioid receptors (KORs) is highly anticipated as a pharmacotherapeutic intervention for substance use disorders and depression. The accepted explanation for KOR antagonist-induced amelioration of aberrant behaviors posits that KORs globally function as a negative valence system; antagonism thereby blunts the behavioral influence of negative internal states such as anhedonia and negative affect. While effects of systemic KOR manipulations have been widely reproduced, explicit evaluation of negative valence as an explanatory construct is lacking. Here, we tested a series of falsifiable hypotheses generated a priori based on the negative valence model by pairing reinforcement learning tasks with systemic pharmacological KOR blockade in male C57BL/6J mice. The negative valence model failed to predict multiple experimental outcomes: KOR blockade accelerated contingency learning during both positive and negative reinforcement without altering innate responses to appetitive or aversive stimuli. We next proposed novelty processing, which influences learning independent of valence, as an alternative explanatory construct. Hypotheses based on novelty processing predicted subsequent observations: KOR blockade increased exploration of a novel, but not habituated, environment and augmented the reinforcing efficacy of novel visual stimuli in a sensory reinforcement task. Together, these results revise and extend long-standing theories of KOR system function.
Collapse
Affiliation(s)
- Zahra Z Farahbakhsh
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Keaton Song
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Hannah E Branthwaite
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kirsty R Erickson
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Snigdha Mukerjee
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Suzanne O Nolan
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
14
|
Khan MIH, Sawyer BJ, Akins NS, Le HV. A systematic review on the kappa opioid receptor and its ligands: New directions for the treatment of pain, anxiety, depression, and drug abuse. Eur J Med Chem 2022; 243:114785. [PMID: 36179400 DOI: 10.1016/j.ejmech.2022.114785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022]
Abstract
Kappa opioid receptor (KOR) is a member of the opioid receptor system, the G protein-coupled receptors that are expressed throughout the peripheral and central nervous systems and play crucial roles in the modulation of antinociception and a variety of behavioral states like anxiety, depression, and drug abuse. KOR agonists are known to produce potent analgesic effects and have been used clinically for the treatment of pain, while KOR antagonists have shown efficacy in the treatment of anxiety and depression. This review summarizes the history, design strategy, discovery, and development of KOR ligands. KOR agonists are classified as non-biased, G protein-biased, and β-arrestin recruitment-biased, according to their degrees of bias. The mechanisms and associated effects of the G protein signaling pathway and β-arrestin recruitment signaling pathway are also discussed. Meanwhile, KOR antagonists are classified as long-acting and short-acting, based on their half-lives. In addition, we have special sections for mixed KOR agonists and selective peripheral KOR agonists. The mechanisms of action and pharmacokinetic, pharmacodynamic, and behavioral studies for each of these categories are also discussed in this review.
Collapse
Affiliation(s)
- Md Imdadul H Khan
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Benjamin J Sawyer
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Nicholas S Akins
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Hoang V Le
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
15
|
Wedemeyer MJ, Jennings EM, Smith HR, Chavera TS, Jamshidi RJ, Berg KA, Clarke WP. 14-3-3γ mediates the long-term inhibition of peripheral kappa opioid receptor antinociceptive signaling by norbinaltorphimine. Neuropharmacology 2022; 220:109251. [PMID: 36126728 DOI: 10.1016/j.neuropharm.2022.109251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/16/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022]
Abstract
Long-term inhibition of kappa opioid receptor (KOR) signaling in peripheral pain-sensing neurons is a potential obstacle for development of peripherally-restricted KOR agonists that produce analgesia. Such a long-term inhibitory mechanism is invoked from activation of c-Jun N-terminal kinase (JNK) that follows a single injection of the KOR antagonist norbinaltorphimine (norBNI). This effect requires protein synthesis of an unknown mediator in peripheral pain-sensing neurons. Using 2D difference gel electrophoresis with tandem mass spectrometry, we have identified that the scaffolding protein 14-3-3γ is upregulated in peripheral sensory neurons following activation of JNK with norBNI. Knockdown of 14-3-3γ by siRNA eliminates the long-term reduction in KOR-mediated cAMP signaling by norBNI in peripheral sensory neurons in culture. Similarly, knockdown of 14-3-3γ in the rat hind paw abolished the norBNI-mediated long-term reduction in peripheral KOR-mediated antinociception. Further, overexpression of 14-3-3γ in KOR expressing CHO cells prevented KOR-mediated inhibition of cAMP signaling. These long-term effects are selective for KOR as heterologous regulation of other receptor systems was not observed. These data suggest that 14-3-3γ is both necessary and sufficient for the long-term inhibition of KOR by norBNI in peripheral sensory neurons.
Collapse
Affiliation(s)
- Michael J Wedemeyer
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Elaine M Jennings
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hudson R Smith
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Teresa S Chavera
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Raehannah J Jamshidi
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kelly A Berg
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - William P Clarke
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
16
|
Puls K, Olivé-Marti AL, Pach S, Pinter B, Erli F, Wolber G, Spetea M. In Vitro, In Vivo and In Silico Characterization of a Novel Kappa-Opioid Receptor Antagonist. Pharmaceuticals (Basel) 2022; 15:680. [PMID: 35745598 PMCID: PMC9229160 DOI: 10.3390/ph15060680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Kappa-opioid receptor (KOR) antagonists are promising innovative therapeutics for the treatment of the central nervous system (CNS) disorders. The new scaffold opioid ligand, Compound A, was originally found as a mu-opioid receptor (MOR) antagonist but its binding/selectivity and activation profile at the KOR and delta-opioid receptor (DOR) remain elusive. In this study, we present an in vitro, in vivo and in silico characterization of Compound A by revealing this ligand as a KOR antagonist in vitro and in vivo. In the radioligand competitive binding assay, Compound A bound at the human KOR, albeit with moderate affinity, but with increased affinity than to the human MOR and without specific binding at the human DOR, thus displaying a preferential KOR selectivity profile. Following subcutaneous administration in mice, Compound A effectively reverse the antinociceptive effects of the prototypical KOR agonist, U50,488. In silico investigations were carried out to assess the structural determinants responsible for opioid receptor subtype selectivity of Compound A. Molecular docking, molecular dynamics simulations and dynamic pharmacophore (dynophore) generation revealed differences in the stabilization of the chlorophenyl moiety of Compound A within the opioid receptor binding pockets, rationalizing the experimentally determined binding affinity values. This new chemotype bears the potential for favorable ADMET properties and holds promise for chemical optimization toward the development of potential therapeutics.
Collapse
Affiliation(s)
- Kristina Puls
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany; (K.P.); (S.P.)
| | - Aina-Leonor Olivé-Marti
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (A.-L.O.-M.); (B.P.); (F.E.)
| | - Szymon Pach
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany; (K.P.); (S.P.)
| | - Birgit Pinter
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (A.-L.O.-M.); (B.P.); (F.E.)
| | - Filippo Erli
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (A.-L.O.-M.); (B.P.); (F.E.)
| | - Gerhard Wolber
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany; (K.P.); (S.P.)
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (A.-L.O.-M.); (B.P.); (F.E.)
| |
Collapse
|
17
|
Yakhnitsa V, Ji G, Hein M, Presto P, Griffin Z, Ponomareva O, Navratilova E, Porreca F, Neugebauer V. Kappa Opioid Receptor Blockade in the Amygdala Mitigates Pain Like-Behaviors by Inhibiting Corticotropin Releasing Factor Neurons in a Rat Model of Functional Pain. Front Pharmacol 2022; 13:903978. [PMID: 35694266 PMCID: PMC9177060 DOI: 10.3389/fphar.2022.903978] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 01/06/2023] Open
Abstract
Functional pain syndromes (FPS) occur in the absence of identifiable tissue injury or noxious events and include conditions such as migraine, fibromyalgia, and others. Stressors are very common triggers of pain attacks in various FPS conditions. It has been recently demonstrated that kappa opioid receptors (KOR) in the central nucleus of amygdala (CeA) contribute to FPS conditions, but underlying mechanisms remain unclear. The CeA is rich in KOR and encompasses major output pathways involving extra-amygdalar projections of corticotropin releasing factor (CRF) expressing neurons. Here we tested the hypothesis that KOR blockade in the CeA in a rat model of FPS reduces pain-like and nocifensive behaviors by restoring inhibition of CeA-CRF neurons. Intra-CeA administration of a KOR antagonist (nor-BNI) decreased mechanical hypersensitivity and affective and anxiety-like behaviors in a stress-induced FPS model. In systems electrophysiology experiments in anesthetized rats, intra-CeA application of nor-BNI reduced spontaneous firing and responsiveness of CeA neurons to peripheral stimulation. In brain slice whole-cell patch-clamp recordings, nor-BNI increased feedforward inhibitory transmission evoked by optogenetic and electrical stimulation of parabrachial afferents, but had no effect on monosynaptic excitatory transmission. Nor-BNI decreased frequency, but not amplitude, of spontaneous inhibitory synaptic currents, suggesting a presynaptic action. Blocking KOR receptors in stress-induced FPS conditions may therefore represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Vadim Yakhnitsa
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Matthew Hein
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Zack Griffin
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Olga Ponomareva
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Edita Navratilova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
18
|
Cox BM, Toll L. Contributions of the International Narcotics Research Conference to Opioid Research Over the Past 50 years. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10115. [PMID: 38390618 PMCID: PMC10880772 DOI: 10.3389/adar.2022.10115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/14/2022] [Indexed: 02/24/2024]
Abstract
The International Narcotics Research Conference (INRC), founded in 1969, has been a successful forum for research into the actions of opiates, with an annual conference since 1971. Every year, scientists from around the world have congregated to present the latest data on novel opiates, opiate receptors and endogenous ligands, mechanisms of analgesic activity and unwanted side effects, etc. All the important discoveries in the opiate field were discussed, often first, at the annual INRC meeting. With an apology to important events and participants not discussed, this review presents a short history of INRC with a discussion of groundbreaking discoveries in the opiate field and the researchers who presented from the first meeting up to the present.
Collapse
Affiliation(s)
- Brian M. Cox
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Lawrence Toll
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
19
|
Blockade of kappa opioid receptors reduces mechanical hyperalgesia and anxiety-like behavior in a rat model of trigeminal neuropathic pain. Behav Brain Res 2022; 417:113595. [PMID: 34592375 DOI: 10.1016/j.bbr.2021.113595] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/12/2021] [Accepted: 09/20/2021] [Indexed: 12/18/2022]
Abstract
It has been shown that kappa opioid receptor (KOR) antagonists, such as nor-binaltorphimine (nor-BNI), have antinociceptive effects in some pain models that affect the trigeminal system. Also, its anxiolytic-like effect has been extensively demonstrated in the literature. The present study aimed to investigate the systemic, local, and central effect of nor-BNI on trigeminal neuropathic pain using the infraorbital nerve constriction model (CCI-ION), as well as to evaluate its effect on anxiety-like behavior associated with this model. Animals received nor-BNI systemically; in the trigeminal ganglion (TG); in the subarachnoid space to target the spinal trigeminal nucleus caudalis (Sp5C) or in the central amygdala (CeA) 14 days after CCI-ION surgery. Systemic administration of nor-BNI caused a significant reduction of facial mechanical hyperalgesia and promoted an anxiolytic-like effect, which was detected in the elevated plus-maze and the light-dark transition tests. When administered in the TG or CeA, the KOR antagonist was able to reduce facial mechanical hyperalgesia induced by CCI-ION, but without changing the anxiety-like behavior. Moreover, no change was observed on nociception and anxiety-like behavior after nor-BNI injection into the Sp5C. The present study demonstrated antinociceptive and anxiolytic-like effects of nor-BNI in a model of trigeminal neuropathic pain. The antinociceptive effect seems to be dissociated from the anxiolytic-like effect, at both the sites involved and at the dose need to achieve the effect. In conclusion, the kappa opioid system may represent a promising target to be explored for the control of trigeminal pain and associated anxiety. However, further studies are necessary to better elucidate its functioning and modulatory role in chronic trigeminal pain states.
Collapse
|
20
|
Fundamentals of the Dynorphins/Kappa Opioid Receptor System: From Distribution to Signaling and Function. Handb Exp Pharmacol 2022; 271:3-21. [PMID: 33754230 PMCID: PMC9013522 DOI: 10.1007/164_2021_433] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This chapter provides a general introduction to the dynorphins (DYNs)/kappa opioid receptor (KOR) system, including DYN peptides, neuroanatomy of the DYNs/KOR system, cellular signaling, and in vivo behavioral effects of KOR activation and inhibition. It is intended to serve as a primer for the book and to provide a basic background for the chapters in the book.
Collapse
|
21
|
Baynard C, Prisinzano TE, Butelman ER. Rapid-Onset Anti-Stress Effects of a Kappa-Opioid Receptor Antagonist, LY2795050, Against Immobility in an Open Space Swim Paradigm in Male and Female Mice. Front Pharmacol 2021; 12:775317. [PMID: 34880762 PMCID: PMC8645979 DOI: 10.3389/fphar.2021.775317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The kappa-opioid receptor (KOR) / dynorphin system is implicated with behavioral and neurobiological effects of stress exposure (including heavy exposure to drugs of abuse) in translational animal models. Thus some KOR-antagonists can decrease the aversive, depressant-like and anxiety-like effects caused by stress exposure. The first generation of selective KOR-antagonists have slow onsets (hours) and extremely long durations of action (days-weeks), in vivo. A new generation of KOR antagonists with rapid onset and shorter duration of action can potentially decrease the effects of stress exposure in translational models, and may be of interest for medication development. This study examined the rapid onset anti-stress effects of one of the shorter acting novel KOR-antagonists (LY2795050, (3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide)) in a single-session open space swim (OSS) stress paradigm (15 min duration), in adult male and female C57BL/6 J mice. LY2795050 (0.32 mg/kg, i.p.) had rapid onset (within 15 min) and short duration (<3 h) of KOR-antagonist effects, based on its blockade of the locomotor depressant effects of the KOR-agonist U50,488 (10 mg/kg). LY2795050 (0.32 mg/kg), when administered only 1 min prior to the OSS stress paradigm, decreased immobility in males, but not females. With a slightly longer pretreatment time (15 min), this dose of LY2795050 decreased immobility in both males and females. A 10-fold smaller dose of LY2795050 (0.032 mg/kg) was inactive in the OSS, showing dose-dependence of this anti-stress effect. Overall, these studies show that a novel KOR-antagonist can produce very rapid onset anti-immobility effects in this model of acute stress exposure.
Collapse
Affiliation(s)
- Caroline Baynard
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| |
Collapse
|
22
|
Experience-dependent myelination following stress is mediated by the neuropeptide dynorphin. Neuron 2021; 109:3619-3632.e5. [PMID: 34536353 DOI: 10.1016/j.neuron.2021.08.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/14/2021] [Accepted: 08/13/2021] [Indexed: 11/22/2022]
Abstract
Emerging evidence implicates experience-dependent myelination in learning and memory. However, the specific signals underlying this process remain unresolved. We demonstrate that the neuropeptide dynorphin, which is released from neurons upon high levels of activity, promotes experience-dependent myelination. Following forced swim stress, an experience that induces striatal dynorphin release, we observe increased striatal oligodendrocyte precursor cell (OPC) differentiation and myelination, which is abolished by deleting dynorphin or blocking its endogenous receptor, kappa opioid receptor (KOR). We find that dynorphin also promotes developmental OPC differentiation and myelination and demonstrate that this effect requires KOR expression specifically in OPCs. We characterize dynorphin-expressing neurons and use genetic sparse labeling to trace their axonal projections. Surprisingly, we find that they are unmyelinated normally and following forced swim stress. We propose a new model whereby experience-dependent and developmental myelination is mediated by unmyelinated, neuropeptide-expressing neurons that promote OPC differentiation for the myelination of neighboring axons.
Collapse
|
23
|
Bossert JM, Townsend EA, Altidor LKP, Fredriksson I, Shekara A, Husbands S, Sulima A, Rice KC, Banks ML, Shaham Y. Sex differences in the effect of chronic delivery of the buprenorphine analogue BU08028 on heroin relapse and choice in a rat model of opioid maintenance. Br J Pharmacol 2021; 179:227-241. [PMID: 34505281 DOI: 10.1111/bph.15679] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Maintenance treatment with opioid agonists (buprenorphine, methadone) decreases opioid use and relapse. We recently modelled maintenance treatment in rats and found that chronic delivery of buprenorphine or the μ opioid receptor partial agonist TRV130 decreased relapse to oxycodone seeking and taking. Here, we tested the buprenorphine analogue BU08028 on different heroin relapse-related measures and heroin versus food choice. EXPERIMENTAL APPROACH For relapse assessment, we trained male and female rats to self-administer heroin (6 h·day-1 , 14 days) in Context A and then implanted osmotic minipumps containing BU08028 (0, 0.03 or 0.1 mg·kg-1 ·d-1 ). Effects of chronic BU08028 delivery were tested on (1) incubation of heroin-seeking in a non-drug Context B, (2) extinction responding reinforced by heroin-associated discrete cues in Context B, (3) reinstatement of heroin-seeking induced by re-exposure to Context A and (4) re-acquisition of heroin self-administration in Context A. For choice assessment, we tested the effect of chronic BU08028 delivery on heroin versus food choice. KEY RESULTS Chronic BU08028 delivery decreased incubation of heroin seeking. Unexpectedly, BU08028 increased re-acquisition of heroin self-administration selectively in females. Chronic BU08028 had minimal effects on context-induced reinstatement and heroin versus food choice in both sexes. Finally, exploratory post hoc analyses suggest that BU08028 decreased extinction responding selectively in males. CONCLUSIONS AND IMPLICATIONS Chronic BU08028 delivery had both beneficial and detrimental, sex-dependent, effects on different triggers of heroin relapse and minimal effects on heroin choice in both sexes. Results suggest that BU08028 would not be an effective opioid maintenance treatment in humans.
Collapse
Affiliation(s)
| | - E Andrew Townsend
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | - Ida Fredriksson
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA
| | - Aniruddha Shekara
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA
| | - Stephen Husbands
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Agnieszka Sulima
- Molecular Targets and Medications Discovery Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA.,Chemical Biology Research Branch, IRP/NIAAA/NIH, Rockville, Maryland, USA
| | - Kenner C Rice
- Molecular Targets and Medications Discovery Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA.,Chemical Biology Research Branch, IRP/NIAAA/NIH, Rockville, Maryland, USA
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yavin Shaham
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Hedrick SL, Luo D, Kaska S, Niloy KK, Jackson K, Sarma R, Horn J, Baynard C, Leggas M, Butelman ER, Kreek MJ, Prisinzano TE. Design, synthesis, and preliminary evaluation of a potential synthetic opioid rescue agent. J Biomed Sci 2021; 28:62. [PMID: 34503531 PMCID: PMC8427951 DOI: 10.1186/s12929-021-00758-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/20/2021] [Indexed: 11/20/2022] Open
Abstract
Background One of the most prominent opioid analgesics in the United States is the high potency agonist fentanyl. It is used in the treatment of acute and chronic pain and as an anesthetic adjuvant. When used inappropriately, however, ingestion of just a few milligrams of fentanyl or other synthetic opioid can cause opioid-induced respiratory depression (OIRD), often leading to death. Currently, the treatment of choice for OIRD is the opioid receptor antagonist naloxone. Recent reports, however, suggest that higher doses or repeated dosing of naloxone (due to recurrence of respiratory depression) may be required to reverse fully fentanyl-induced respiratory depression, rendering this treatment inadequate. To combat this synthetic opioid overdose crisis, this research aims at identifying a novel opioid reversal agent with enhanced efficacy towards fentanyl and other synthetic opioids. Methods A series of naltrexone analogues were characterized for their ability to antagonize the effects of fentanyl in vitro utilizing a modified forskolin-induced cAMP accumulation assay. Lead analogue 29 was chosen to undergo further PK studies, followed by in vivo pharmacological analysis to determine its ability to antagonize opioid-induced antinociception in the hot plate assay. Results A series of potent MOR antagonists were identified, including the highly potent analogue 29 (IC50 = 2.06 nM). Follow-up PK studies revealed 29 to possess near 100% bioavailability following IP administration. Brain concentrations of 29 surpassed plasma concentrations, with an apparent terminal half-life of ~ 80 min in mice. In the hot plate assay, 29 dose-dependently (0.01–0.1 mg/kg; IP) and fully antagonized the antinociception induced by oxycodone (5.6 mg/kg; IP). Furthermore, the dose of 29 that is fully effective in preventing oxycodone-induced antinociception (0.1 mg/kg) was ineffective against locomotor deficits caused by the KOR agonist U50,488. Conclusions Methods have been developed that have utility to identify enhanced rescue agents for the treatment of OIRD. Analogue 29, possessing potent MOR antagonist activity in vitro and in vivo, provides a promising lead in our search for an enhanced synthetic opioid rescue agent. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-021-00758-y.
Collapse
Affiliation(s)
- Sidnee L Hedrick
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA
| | - Dan Luo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA
| | - Sophia Kaska
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA
| | - Kumar Kulldeep Niloy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA
| | - Karen Jackson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA.,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - Rupam Sarma
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA.,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - Jamie Horn
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA.,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - Caroline Baynard
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, 10065, USA
| | - Markos Leggas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA.,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA
| | - Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, 10065, USA
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, 10065, USA
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40536, USA. .,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
25
|
Butelman ER, Baynard C, McElroy BD, Prisinzano TE, Kreek MJ. Profile of a short-acting κ-antagonist, LY2795050, on self-grooming behaviors, forced swim test and locomotor activity: sex comparison in mice. J Psychopharmacol 2021; 35:579-590. [PMID: 33769112 DOI: 10.1177/0269881121996883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Novel short-acting κ(kappa)-opioid receptor selective antagonists are translational tools to examine the impact of the κ-receptor/dynorphin system in assays related to central nervous system dysfunction (e.g., substance use disorders, anhedonia and depression). The effects of such compounds have been compared in males and females under very limited conditions. AIMS The goal of this study was to examine potential sex differences in the effects of a κ-agonist and a short-acting κ-antagonist in an ethologically relevant test of anhedonia, the "splash test" of self-grooming, and also in the forced swim test and in locomotor activity. METHODS We examined the dose-dependence of grooming deficits caused by the κ-agonist U50,488 (0.1-3.2 mg/kg intraperitoneal (i.p.)) in gonadally intact adult male and female C57BL/6J mice. We then compared the effects of the short-acting κ-antagonist LY2795050 ((3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide)); 0.032-0.1 mg/kg i.p.) in blocking grooming deficits caused by U50,488 (3.2 mg/kg). The effects of LY2795050 were also studied in the forced swim test (FST). The effects of LY2795050 in blocking the locomotor depressant effects of U50,488 (10 mg/kg) were also studied. RESULTS U50,488 produced dose-dependent grooming deficits in male and female mice, and LY2795050 prevented these effects. In contrast, LY2795050 decreased immobility in the FST in males at a dose of 0.1 mg/kg, but not in females, up to a dose of 0.32 mg/kg. Also, LY2795050 (0.32 mg/kg) prevented and also reversed the locomotor-depressant effects of U50,488 (10 mg/kg), in males and females. CONCLUSIONS This study further implicates the κ-receptor system in ethologically relevant aspects of anhedonia, and confirms sexual dimorphism in some behavioral effects of novel κ-antagonists.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, USA
| | - Caroline Baynard
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, USA
| | - Bryan D McElroy
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, USA
| | | | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, USA
| |
Collapse
|
26
|
Marchette RCN, Gregory-Flores A, Tunstall BJ, Carlson ER, Jackson SN, Sulima A, Rice KC, Koob GF, Vendruscolo LF. κ-Opioid receptor antagonism reverses heroin withdrawal-induced hyperalgesia in male and female rats. Neurobiol Stress 2021; 14:100325. [PMID: 33997152 PMCID: PMC8095052 DOI: 10.1016/j.ynstr.2021.100325] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 03/18/2021] [Accepted: 04/06/2021] [Indexed: 10/29/2022] Open
Abstract
Although opioids are potent analgesics, a consequence of chronic opioid use is hyperalgesia during withdrawal, which may contribute to opioid misuse. Dynorphin, the endogenous ligand of κ-opioid receptors (KORs), is upregulated in opioid-dependent rats and in animal models of chronic pain. However, the role of KORs in opioid withdrawal-induced hyperalgesia remains to be determined. We hypothesized that KOR antagonism would reverse opioid withdrawal-induced hyperalgesia in opioid-dependent rats. Male and female Wistar rats received daily injections of heroin (2-6 mg/kg, SC) and were tested for mechanical sensitivity in the electronic von Frey test 4-6 h into withdrawal. Female rats required significantly more heroin than male rats to reach comparable levels of both heroin-induced analgesia and hyperalgesia (6 mg/kg vs. 2 mg/kg). Once hyperalgesia was established, we tested the effects of the KOR antagonists nor-binaltorphimine (norBNI; 30 mg/kg, SC) and 5'-guanidinonaltrindole (5'GNTI; 30 mg/kg, SC). When the animals continued to receive their daily heroin treatment (or saline treatment in the repeated saline group) five times per week throughout the experiment, both KOR antagonists reversed heroin withdrawal-induced hyperalgesia. The anti-hyperalgesia effect of norBNI was more prolonged in males than in females (14 days vs. 7 days), whereas 5'GNTI had more prolonged effects in females than in males (14 days vs. 4 days). The behavioral effects of 5'GNTI coincided with higher 5'GNTI levels in the brain than in plasma when measured at 24 h, whereas 5'GNTI did not reverse hyperalgesia at 30 min posttreatment when 5'GNTI levels were higher in plasma than in the brain. Finally, we tested the effects of 5'GNTI on naloxone-induced and spontaneous signs of opioid withdrawal and found no effect in either male or female rats. These findings indicate a functional role for KORs in heroin withdrawal-induced hyperalgesia that is observed in rats of both sexes.
Collapse
Affiliation(s)
- Renata C N Marchette
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Adriana Gregory-Flores
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Brendan J Tunstall
- Department of Pharmacology, Addiction Science, and Toxicology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Erika R Carlson
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Shelley N Jackson
- Structural Biology Core, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, National Institute on Drug Abuse, Intramural Research Program, Bethesda, MD, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse, Intramural Research Program, Bethesda, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
27
|
Spetea M, Schmidhammer H. Kappa Opioid Receptor Ligands and Pharmacology: Diphenethylamines, a Class of Structurally Distinct, Selective Kappa Opioid Ligands. Handb Exp Pharmacol 2021; 271:163-195. [PMID: 33454858 DOI: 10.1007/164_2020_431] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The kappa opioid receptor (KOR), a G protein-coupled receptor, and its endogenous ligands, the dynorphins, are prominent members of the opioid neuromodulatory system. The endogenous kappa opioid system is expressed in the central and peripheral nervous systems, and has a key role in modulating pain in central and peripheral neuronal circuits and a wide array of physiological functions and neuropsychiatric behaviors (e.g., stress, reward, emotion, motivation, cognition, epileptic seizures, itch, and diuresis). We review the latest advances in pharmacology of the KOR, chemical developments on KOR ligands with advances and challenges, and therapeutic and potential applications of KOR ligands. Diverse discovery strategies of KOR ligands targeting natural, naturally derived, and synthetic compounds with different scaffolds, as small molecules or peptides, with short or long-acting pharmacokinetics, and central or peripheral site of action, are discussed. These research efforts led to ligands with distinct pharmacological properties, as agonists, partial agonists, biased agonists, and antagonists. Differential modulation of KOR signaling represents a promising strategy for developing pharmacotherapies for several human diseases, either by activating (treatment of pain, pruritus, and epilepsy) or blocking (treatment of depression, anxiety, and addiction) the receptor. We focus on the recent chemical and pharmacological advances on diphenethylamines, a new class of structurally distinct, selective KOR ligands. Design strategies and investigations to define structure-activity relationships together with in vivo pharmacology of diphenethylamines as agonists, biased agonists, and antagonists and their potential use as therapeutics are discussed.
Collapse
Affiliation(s)
- Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria.
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Bartlett MJ, Mabrouk OS, Szabò L, Flores AJ, Parent KL, Bidlack JM, Heien ML, Kennedy RT, Polt R, Sherman SJ, Falk T. The Delta-Specific Opioid Glycopeptide BBI-11008: CNS Penetration and Behavioral Analysis in a Preclinical Model of Levodopa-Induced Dyskinesia. Int J Mol Sci 2020; 22:ijms22010020. [PMID: 33374986 PMCID: PMC7792611 DOI: 10.3390/ijms22010020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
In previous work we evaluated an opioid glycopeptide with mixed μ/δ-opioid receptor agonism that was a congener of leu-enkephalin, MMP-2200. The glycopeptide analogue showed penetration of the blood-brain barrier (BBB) after systemic administration to rats, as well as profound central effects in models of Parkinson's disease (PD) and levodopa (L-DOPA)-induced dyskinesia (LID). In the present study, we tested the glycopeptide BBI-11008 with selective δ-opioid receptor agonism, an analogue of deltorphin, a peptide secreted from the skin of frogs (genus Phyllomedusa). We tested BBI-11008 for BBB-penetration after intraperitoneal (i.p.) injection and evaluated effects in LID rats. BBI-11008 (10 mg/kg) demonstrated good CNS-penetrance as shown by microdialysis and mass spectrometric analysis, with peak concentration levels of 150 pM in the striatum. While BBI-11008 at both 10 and 20 mg/kg produced no effect on levodopa-induced limb, axial and oral (LAO) abnormal involuntary movements (AIMs), it reduced the levodopa-induced locomotor AIMs by 50% after systemic injection. The N-methyl-D-aspartate receptor antagonist MK-801 reduced levodopa-induced LAO AIMs, but worsened PD symptoms in this model. Co-administration of MMP-2200 had been shown prior to block the MK-801-induced pro-Parkinsonian activity. Interestingly, BBI-11008 was not able to block the pro-Parkinsonian effect of MK-801 in the LID model, further indicating that a balance of mu- and delta-opioid agonism is required for this modulation. In summary, this study illustrates another example of meaningful BBB-penetration of a glycopeptide analogue of a peptide to achieve a central behavioral effect, providing additional evidence for the glycosylation technique as a method to harness therapeutic potential of peptides.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacokinetics
- Analgesics, Opioid/pharmacology
- Animals
- Corpus Striatum/metabolism
- Disease Models, Animal
- Dizocilpine Maleate/pharmacology
- Dyskinesia, Drug-Induced/metabolism
- Dyskinesia, Drug-Induced/physiopathology
- Glycopeptides/administration & dosage
- Glycopeptides/pharmacokinetics
- Glycopeptides/pharmacology
- Levodopa
- Male
- Motor Activity/drug effects
- Motor Activity/physiology
- Neuroprotective Agents/pharmacology
- Parkinson Disease, Secondary/chemically induced
- Parkinson Disease, Secondary/metabolism
- Parkinson Disease, Secondary/physiopathology
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
Collapse
Affiliation(s)
- Mitchell J. Bartlett
- Department of Neurology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (M.J.B.); (S.J.S.)
| | - Omar S. Mabrouk
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; (O.S.M.); (R.T.K.)
| | - Lajos Szabò
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85721, USA; (L.S.); (K.L.P.); (M.L.H.); (R.P.)
| | - Andrew J. Flores
- Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, AZ 85724, USA;
| | - Kate L. Parent
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85721, USA; (L.S.); (K.L.P.); (M.L.H.); (R.P.)
| | - Jean M. Bidlack
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA;
| | - Michael L. Heien
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85721, USA; (L.S.); (K.L.P.); (M.L.H.); (R.P.)
| | - Robert T. Kennedy
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; (O.S.M.); (R.T.K.)
| | - Robin Polt
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85721, USA; (L.S.); (K.L.P.); (M.L.H.); (R.P.)
| | - Scott J. Sherman
- Department of Neurology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (M.J.B.); (S.J.S.)
| | - Torsten Falk
- Department of Neurology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (M.J.B.); (S.J.S.)
- Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, AZ 85724, USA;
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA
- Correspondence: ; Tel.: +1-520-626-3927
| |
Collapse
|
29
|
Bossert JM, Kiyatkin E, Korah H, Hoots JK, Afzal A, Perekopskiy D, Thomas S, Fredriksson I, Blough BE, Negus SS, Epstein DH, Shaham Y. In a Rat Model of Opioid Maintenance, the G Protein-Biased Mu Opioid Receptor Agonist TRV130 Decreases Relapse to Oxycodone Seeking and Taking and Prevents Oxycodone-Induced Brain Hypoxia. Biol Psychiatry 2020; 88:935-944. [PMID: 32305216 PMCID: PMC7483192 DOI: 10.1016/j.biopsych.2020.02.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Maintenance treatment with opioid agonists (buprenorphine, methadone) is effective for opioid addiction but does not eliminate opioid use in all patients. We modeled maintenance treatment in rats that self-administered the prescription opioid oxycodone. The maintenance medication was either buprenorphine or the G protein-biased mu opioid receptor agonist TRV130. We then tested prevention of oxycodone seeking and taking during abstinence using a modified context-induced reinstatement procedure, a rat relapse model. METHODS We trained rats to self-administer oxycodone (6 hours/day, 14 days) in context A; infusions were paired with discrete tone-light cues. We then implanted osmotic pumps containing buprenorphine or TRV130 (0, 3, 6, or 9 mg/kg/day) and performed 3 consecutive tests: lever pressing reinforced by oxycodone-associated discrete cues in nondrug context B (extinction responding), context-induced reinstatement of oxycodone seeking in context A, and reacquisition of oxycodone self-administration in context A. We also tested whether TRV130 maintenance would protect against acute oxycodone-induced decreases in nucleus accumbens oxygen levels. RESULTS In male rats, buprenorphine and TRV130 decreased extinction responding and reacquisition of oxycodone self-administration but had a weaker (nonsignificant) effect on context-induced reinstatement. In female rats, buprenorphine decreased responding in all 3 tests, while TRV130 decreased only extinction responding. In both sexes, TRV130 prevented acute brain hypoxia induced by moderate doses of oxycodone. CONCLUSIONS TRV130 decreased oxycodone seeking and taking during abstinence in a partly sex-specific manner and prevented acute oxycodone-induced brain hypoxia. We propose that G protein-biased mu opioid receptor agonists, currently in development as analgesics, should be considered as relapse prevention maintenance treatment for opioid addiction.
Collapse
Affiliation(s)
- Jennifer M. Bossert
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A.,Correspondence: Jennifer M. Bossert ()
| | - Eugene Kiyatkin
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Hannah Korah
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | | | - Anum Afzal
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | | | - Shruthi Thomas
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Ida Fredriksson
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Bruce E. Blough
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC, U.S.A
| | - S. Stevens Negus
- Dept. of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - David H. Epstein
- Clinical Pharmacology and Therapeutics Research Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| | - Yavin Shaham
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, U.S.A
| |
Collapse
|
30
|
Odagaki Y, Kinoshita M, Javier Meana J, Callado LF, García-Sevilla JA. Fundamental features of receptor-mediated Gα i/o activation in human prefrontal cortical membranes: A postmortem study. Brain Res 2020; 1747:147032. [PMID: 32745659 DOI: 10.1016/j.brainres.2020.147032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/12/2020] [Accepted: 07/23/2020] [Indexed: 11/19/2022]
Abstract
To elucidate possible abnormalities in transmembrane signal transduction in psychiatric diseases, use of autopsy brain is a feasible approach. However, postmortem studies should be interpreted with caution concerning such factors as age, gender, psychotropic drug history, agonal state, postmortem delay (PMD), and storage period. In this study, agonist-induced [35S]GTPγS binding was performed in postmortem dorsolateral prefrontal cortical membranes of 40 control subjects. In addition to the previously reported G protein-coupled receptor (GPCR)-mediated Gi/o activation, κ-opioid receptor-mediated [35S]GTPγS binding was detected by using U-50,448. The responses elicited by 16 different agonists were determined, and the effects of several factors were investigated. Gender difference was negligible. Concentration-response curve of histamine H3 receptor-mediated [35S]GTPγS binding was shifted rightward in the subjects with some drugs detected at toxicological screening. Age-related alterations were minimal, except for the age-dependent supersensitivity of μ-opioid receptor-mediated Gαi/o activation, revealed by endomorphin-1- and DAMGO-stimulated [35S]GTPγS binding. Age-related increase in %Emax values was also detected as to DPDPE-induced [35S]GTPγS binding through δ-opioid receptors. With an exception of NOP receptor/G-protein coupling, GPCR-mediated [35S]GTPγS binding is relatively stable irrespective of PMD or storage period. There were many positive correlations among the %Emax values for different receptor subtypes, which might reflect formation of heterodimer complex of such GPCRs coupled to the same Gi/o proteins. These results provide us with important fundamental data in the future project using human postmortem brains from patients with psychiatric disorders.
Collapse
Affiliation(s)
- Yuji Odagaki
- Department of Psychiatry, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan.
| | - Masakazu Kinoshita
- Department of Psychiatry, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Jesús A García-Sevilla
- Laboratory of Neuropharmacology, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), University of the Balearic Islands (UIB), and Institut d'investigació Sanitària Illes Balears (IdISBa), Palma de Mallorca, Spain
| |
Collapse
|
31
|
Barnett ME, Knapp BI, Bidlack JM. Unique Pharmacological Properties of the Kappa Opioid Receptor Signaling Through G αz as Shown with Bioluminescence Resonance Energy Tranfer. Mol Pharmacol 2020; 98:462-474. [PMID: 32958572 PMCID: PMC7562983 DOI: 10.1124/mol.120.119404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
Opioid receptors (ORs) convert extracellular messages to signaling events by coupling to the heterotrimeric G proteins, Gα•βγ Classic pharmacological methods, such as [35S]GTPγS binding and inhibition of cyclic AMP production, allow for general opioid characterization, but they are subject to the varying endogenous Gα proteins in a given cell type. Bioluminescence resonance energy transfer (BRET) technology offers new insight by allowing the direct observation of Gα subunit-specific effects on opioid pharmacology. Using a Venus-tagged Gβγ and nanoluciferase-tagged truncated G protein receptor kinase 3, an increase in BRET signal correlated with OR activation mediated by a specific Gα protein. The magnitude of the BRET signal was normalized to the maximum response obtained with 10 µM 2-(3,4-dichlorophenyl)-N-methyl-N-[(1R,2R)-2-pyrrolidin-1-ylcyclohexyl]acetamide (U50,488) for the kappa OR (KOR). Opioids reached equilibrium with the KOR, and concentration-response curves were generated. Although the full agonists U50,488, salvinorin A, nalfurafine, and dynorphin peptides were equally efficacious regardless of the Gα subunit present, the concentration-response curves were leftward shifted when the KOR was signaling through Gαz compared with other Gαi/o subunits. In contrast, the Gα subunit distinctly affected both the efficacy and potency of partial kappa agonists, such as the benzomorphans, and the classic mu opioid antagonists, naloxone, naltrexone, and nalmefene. For example, (-)pentazocine had EC50 values of 7.3 and 110 nM and maximal stimulation values of 79% and 35% when the KOR signaled through Gαz and Gαi1, respectively. Together, these observations suggest KOR pharmacology varies based on the specific Gα subunit coupled to the KOR. SIGNIFICANCE STATEMENT: Opioid receptors couple to various heterotrimeric Gαβγ proteins to convert extracellular cues to precise intracellular events. This paper focuses on how the various inhibitory Gα subunits influence the pharmacology of full and partial agonists at the kappa opioid receptor. Using a bioluminescent assay, the efficacy and potency of kappa opioids was determined. Opioid signaling was more potent through Gαz compared with other Gα proteins. These observations suggest that Gαz may impact opioid pharmacology and cellular physiology more than previously thought.
Collapse
Affiliation(s)
- Miriam E Barnett
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York
| | - Brian I Knapp
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York
| | - Jean M Bidlack
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York
| |
Collapse
|
32
|
Akgün E, Lunzer MM, Tian D, Ansonoff M, Pintar J, Bruce D, Hawkinson JE, Wilcox GL, Portoghese PS. FBNTI, a DOR-Selective Antagonist That Allosterically Activates MOR within a MOR-DOR Heteromer. Biochemistry 2020; 60:1413-1419. [PMID: 32930576 DOI: 10.1021/acs.biochem.0c00498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This report describes the unique pharmacological profile of FBNTI, a potent DOR antagonist that acts as a MOR agonist via an allosteric mechanism. Binding of FBNTI to opioid receptors expressed in HEK 293 cells revealed a 190-fold greater affinity for DOR (Ki = 0.84 nM) over MOR (Ki = 160 nM). In mice, intrathecal FBNTI produced potent antinociception (ED50 = 46.9 pmol/mouse), which was antagonized by selective MOR antagonists (CTOP, β-FNA). Autoantagonism of the MOR agonism by FBNTI was observed above the ED75 dose, suggesting antagonism of activated MOR. That FBNTI is devoid of agonism in DOR knockout mice is consistent with allosteric activation of the MOR protomer via FBNTI bound to within a MOR-DOR heteromer. This proposed mechanism is supported by calcium mobilization assays, which indicate that FBNTI selectively activates the MOR-DOR heteromer and functionally antagonizes the MOR protomer at >ED75. The unprecedented mode of MOR activation by FBNTI may be responsible for the lack of tolerance after intrathecal (i.t.) administration. FBNTI was highly effective upon topical administration to the ipsolateral hind paw in the Hargreaves assay (EC50 = 0.17 ± 0.08 μM) and without significant contralateral activity, suggesting a lack of systemic exposure.
Collapse
Affiliation(s)
- Eyup Akgün
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary M Lunzer
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Defeng Tian
- University of Minnesota Institute for Therapeutics Discovery & Development (ITDD), 717 Delaware Street SE Room 519E, Minneapolis, Minnesota 55414, United States
| | - Michael Ansonoff
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, United States
| | - John Pintar
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Daniel Bruce
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jon E Hawkinson
- University of Minnesota Institute for Therapeutics Discovery & Development (ITDD), 717 Delaware Street SE Room 519E, Minneapolis, Minnesota 55414, United States
| | - George L Wilcox
- Departments of Neuroscience, Pharmacology and Dermatology, Medical School, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Philip S Portoghese
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
33
|
Calvo F, Almada RC, da Silva JA, Medeiros P, da Silva Soares R, de Paiva YB, Roncon CM, Coimbra NC. The Blockade of µ1- and κ-Opioid Receptors in the Inferior Colliculus Decreases the Expression of Panic Attack-Like Behaviours Induced by Chemical Stimulation of the Dorsal Midbrain. Neuropsychobiology 2020; 78:218-228. [PMID: 31514182 DOI: 10.1159/000502439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/22/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Gamma-aminobutyric acid (GABA)ergic and opioid systems play a crucial role in the neural modulation of innate fear organised by the inferior colliculus (IC). In addition, the IC is rich in GABAergic fibres and opioid neurons, which are also connected to other mesencephalic structures, such as the superior colliculus and the substantia nigra. However, the contribution of distinct opioid receptors (ORs) in the IC during the elaboration and expression of innate fear and panic-like responses is unclear. The purpose of the present work was to investigate a possible integrated action exerted by ORs and the GABAA receptor-mediated system in the IC on panic-like responses. METHODS The effect of the blockade of either µ1- or κ-ORs in the IC was evaluated in the unconditioned fear-induced responses elicited by GABAA antagonism with bicuculline. Microinjections of naloxonazine, a µ1-OR antagonist, or nor-binaltorphimine (nor-BNI), a κ-OR antagonist, were made into the IC, followed by intramesencephalic administration of the GABAA-receptor antagonist bicuculline. The defensive behaviours elicited by the treatments in the IC were quantitatively analysed, recording the number of escapes expressed as running (crossing), jumps, and rotations, over a 30-min period in a circular arena. The exploratory behaviour of rearing was also recorded. RESULTS GABAA-receptor blockade with bicuculline in the IC increased defensive behaviours. However, pretreatment of the IC with higher doses (5 µg) of naloxonazine or nor-BNI followed by bicuculline resulted in a significant decrease in unconditioned fear-induced responses. CONCLUSIONS These findings suggest a role played by µ1- and κ-OR-containing connexions and GABAA receptor-mediated neurotransmission on the organisation of panic attack-related responses elaborated by the IC neurons.
Collapse
Affiliation(s)
- Fabrício Calvo
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil.,Department of Pharmacology, São Lucas College, Porto Velho, Brazil.,Aparício Carvalho Integrative College (FIMCA), Porto Velho, Brazil
| | - Rafael Carvalho Almada
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil.,Institute of Neuroscience and Behaviour (INeC), Ribeirão Preto, Brazil
| | - Juliana Almeida da Silva
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil.,Institute of Neuroscience and Behaviour (INeC), Ribeirão Preto, Brazil
| | - Priscila Medeiros
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Raimundo da Silva Soares
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Yara Bezerra de Paiva
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Camila Marroni Roncon
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil.,Assis County Educational Foundation (FEMA), Assis, Brazil
| | - Norberto Cysne Coimbra
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil, .,Institute of Neuroscience and Behaviour (INeC), Ribeirão Preto, Brazil, .,NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), School of Medicine of Ribeirão Preto of the University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil,
| |
Collapse
|
34
|
Fernandez TJ, De Maria M, Lobingier BT. A cellular perspective of bias at G protein-coupled receptors. Protein Sci 2020; 29:1345-1354. [PMID: 32297394 DOI: 10.1002/pro.3872] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/17/2022]
Abstract
G protein-coupled receptors (GPCRs) modulate cell function over short- and long-term timescales. GPCR signaling depends on biochemical parameters that define the what, when, and where of receptor function: what proteins mediate and regulate receptor signaling, where within the cell these interactions occur, and how long these interactions persist. These parameters can vary significantly depending on the activating ligand. Collectivity, differential agonist activity at a GPCR is called bias or functional selectivity. Here we review agonist bias at GPCRs with a focus on ligands that show dramatically different cellular responses from their unbiased counterparts.
Collapse
Affiliation(s)
- Thomas J Fernandez
- Department of Chemical Physiology and Biochemistry, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Monica De Maria
- Department of Chemical Physiology and Biochemistry, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Braden T Lobingier
- Department of Chemical Physiology and Biochemistry, Oregon Health and Sciences University, Portland, Oregon, USA
| |
Collapse
|
35
|
Reed B, Butelman ER, Kreek MJ. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Mood and Substance Use Disorders. Handb Exp Pharmacol 2020; 271:473-491. [PMID: 33174064 DOI: 10.1007/164_2020_401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kappa opioid receptor (KOR) and its primary cognate ligands, the dynorphin peptides, are involved in diverse physiological processes. Disruptions to the KOR/dynorphin system have been found to likely play a role in multiple neuropsychological disorders, and hence KOR has emerged as a potential therapeutic target. Targeting KOR is complicated by close homology to the mu and delta opioid receptors (MOR and DOR), and many KOR ligands have at least moderate affinity to MOR and/or DOR. Animal models utilizing primarily very long-lasting selective KOR antagonists (>3 weeks following a single dose) have demonstrated that KOR antagonism attenuates certain anxiety-like and depression-like behaviors and blocks stress- and cue-induced reinstatement to drug seeking. Recently, relatively selective KOR antagonists with medication-like pharmacokinetic and pharmacodynamic properties and durations of action have been developed. One of these, JNJ-67953964 (also referred to as CERC-501, LY2456302, OpraKappa or Aticaprant) has been studied in humans, and shown to be safe, relatively KOR selective, and able to substantially attenuate binding of a KOR PET tracer to CNS localized KOR for greater than 24 h. While animal studies have indicated that compounds of this structural class are capable of normalizing withdrawal signs in animal models of cocaine and alcohol dependence and reducing cocaine and alcohol intake/seeking, additional studies are needed to determine the value of these second generation KOR antagonists in treating mood disorders and substance use disorders in humans.
Collapse
Affiliation(s)
- Brian Reed
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA.
| | - Eduardo R Butelman
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| |
Collapse
|
36
|
Kong C, Miao F, Wu Y, Wang T. Oxycodone suppresses the apoptosis of hippocampal neurons induced by oxygen-glucose deprivation/recovery through caspase-dependent and caspase-independent pathways via κ- and δ-opioid receptors in rats. Brain Res 2019; 1721:146319. [PMID: 31276638 DOI: 10.1016/j.brainres.2019.146319] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/21/2019] [Accepted: 07/01/2019] [Indexed: 02/05/2023]
Abstract
Cerebral ischemia/reperfusion injury (CIRI) can lead to perioperative neurocognitive disorders (PND) during clinical recanalization procedures in cerebral vessels, principally due to neuronal apoptosis in the hippocampus. Oxycodone appears to be a multiple opioid receptor agonist and exerts intrinsic antinociception activity via κ-opioid receptor (KOR). Recent evidence has revealed that activation of both δ-opioid receptor (DOR) and KOR can provide neuroprotection against CIRI in vivo and in vitro. In our study, we established an oxygen-glucose deprivation/recovery (OGD/R) model with fetal hippocampal neurons and found that oxycodone could induce CIRI tolerance in these neurons, primarily through KOR and DOR. Possible mechanisms might involve the regulatory effect of oxycodone on the MAPK-Bcl2/Bax-caspase-9-caspase-3 pathway, as well as its inhibitory effect on cellular reactive oxygen species (ROS) production and mitochondrial membrane potential activation. Taken together, our findings may indicate a potential method for the prevention and treatment of PND associated with CIRI.
Collapse
Affiliation(s)
- Cuicui Kong
- Department of Anesthesiology, Capital Medical University Xuanwu Hospital, Beijing 100053, China
| | - Fangfang Miao
- Department of Anesthesiology, Capital Medical University Xuanwu Hospital, Beijing 100053, China
| | - Yan Wu
- Department of Anatomy, Beijing Institute for Brain Disorders, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China
| | - Tianlong Wang
- Department of Anesthesiology, Capital Medical University Xuanwu Hospital, Beijing 100053, China.
| |
Collapse
|
37
|
Butelman ER, McElroy BD, Prisinzano TE, Kreek MJ. Impact of Pharmacological Manipulation of the κ-Opioid Receptor System on Self-grooming and Anhedonic-like Behaviors in Male Mice. J Pharmacol Exp Ther 2019; 370:1-8. [PMID: 30975792 PMCID: PMC6538891 DOI: 10.1124/jpet.119.256354] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/09/2019] [Indexed: 12/11/2022] Open
Abstract
The kappa (κ) opioid receptor/dynorphin system modulates depression-like states and anhedonia, as well adaptations to stress and exposure to drugs of abuse. Several relatively short-acting small molecule κ-receptor antagonists have been synthesized, and their behavioral profile has been examined under some conditions. The hypothesis of this study is that pharmacological manipulations of the κ-receptor system will result in changes in ethologically relevant anhedonic-like behaviors in mice. Adult male C57BL/6j mice (n = 6-8) were examined for self-grooming behavior in the splash test (in which robust self-grooming is elicited by spraying the dorsum of the mouse with a sucrose solution). The κ-agonist salvinorin A (0.56-1.8 mg/kg) produced dose-dependent decreases in self-grooming, a marker of anhedonia. The selectivity, potency, and duration of action of two relatively short-acting κ-antagonists, LY2444296 [(S)-3-fluoro-4-(4-((2-(3-fluorophenyl) pyrrolidin-1-yl)methyl)phenoxy)benzamide] and LY2795050 [3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide], were studied for their effectiveness in preventing grooming deficits caused by salvinorin A (1.8 mg/kg). κ-selective doses of both LY2444296 (0.032-1 mg/kg) and LY2795050 (0.032-0.32 mg/kg) dose- and time-dependently prevented the grooming deficits caused by salvinorin A (1.8 m/kg). We also found that a κ-selective dose of each of these antagonists decreased immobility in the forced swim test, a common test of anti-anhedonia effects. This study shows that the κ-receptor system is involved in an ethologically relevant measure of anhedonia, and that κ-selective doses of these antagonists can produce effects consistent with rapid anti-anhedonia. SIGNIFICANCE STATEMENT: Activation of the κ-opioid receptor system results in grooming deficits in mice, an ethologically relevant marker of anhedonia. Shorter acting κ-antagonists are able to cause effects consistent with rapid antianhedonia.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Bryan D McElroy
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Thomas E Prisinzano
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| |
Collapse
|
38
|
Placzek MS, Schroeder FA, Che T, Wey HY, Neelamegam R, Wang C, Roth BL, Hooker JM. Discrepancies in Kappa Opioid Agonist Binding Revealed through PET Imaging. ACS Chem Neurosci 2019; 10:384-395. [PMID: 30212182 DOI: 10.1021/acschemneuro.8b00293] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Kappa opioid receptor (KOR) modulation has been pursued in many conceptual frameworks for the treatment of human pain, depression, and anxiety. As such, several imaging tools have been developed to characterize the density of KORs in the human brain and its occupancy by exogenous drug-like compounds. While exploring the pharmacology of KOR tool compounds using positron emission tomography (PET), we observed discrepancies in the apparent competition binding as measured by changes in binding potential (BPND, binding potential with respect to non-displaceable uptake). This prompted us to systematically look at the relationships between baseline BPND maps for three common KOR PET radioligands, the antagonists [11C]LY2795050 and [11C]LY2459989, and the agonist [11C]GR103545. We then measured changes in BPND using kappa antagonists (naloxone, naltrexone, LY2795050, JDTic, nor-BNI), and found BPND was affected similarly between [11C]GR103545 and [11C]LY2459989. Longitudinal PET studies with nor-BNI and JDTic were also examined, and we observed a persistent decrease in [11C]GR103545 BPND up to 25 days after drug administration for both nor-BNI and JDTic. Kappa agonists were also administered, and butorphan and GR89696 (racemic GR103545) impacted binding to comparable levels between the two radiotracers. Of greatest significance, kappa agonists salvinorin A and U-50488 caused dramatic reductions in [11C]GR103545 BPND but did not change [11C]LY2459989 binding. This discrepancy was further examined in dose-response studies with each radiotracer as well as in vitro binding experiments.
Collapse
Affiliation(s)
- Michael S. Placzek
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Frederick A. Schroeder
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Tao Che
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27516, United States
| | - Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Ramesh Neelamegam
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Bryan L. Roth
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27516, United States
- National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27516, United States
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516, United States
| | - Jacob M. Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
39
|
Kappa opioid receptors mediate yohimbine-induced increases in impulsivity in the 5-choice serial reaction time task. Behav Brain Res 2018; 359:258-265. [PMID: 30414973 DOI: 10.1016/j.bbr.2018.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 11/20/2022]
Abstract
Dynorphin (DYN), and its receptor, the kappa opioid receptor (KOR) are involved in drug seeking and relapse but the mechanisms are poorly understood. One hypothesis is that DYN/KOR activation promotes drug seeking through increased impulsivity, because many stimuli that induce DYN release increase impulsivity. Here, we systematically compare the effects of drugs that activate DYN/KOR on performance on the 5-choice serial reaction time task (5-CSRTT), a test of sustained attention and impulsivity. In Experiment 1, we determined the effects of U50,488 (0, 2.5, 5 mg/kg), yohimbine (0, 1.25, 2.5 mg/kg), and nicotine (0, 0.15, 0.3 mg/kg) on 5-CSRTT performance. In Experiment 2, we determined the effects of alcohol (0, 0.5, 1.0, 1.5 g/kg) on 5-CSRTT performance before and after voluntary, intermittent alcohol exposure. In Experiment 3, we determined the potential role of KOR in the pro-impulsive effects of yohimbine (1.25 mg/kg) and nicotine (0.3 mg/kg) by the prior administration of the KOR antagonist nor-BNI (10 mg/kg). Premature responding, the primary measure of impulsivity, was reduced by U50,488 and alcohol, but these drugs had a general suppressive effect. Yohimbine and nicotine increased premature responding. Yohimbine-, but not nicotine-induced increases in premature responding were blocked by nor-BNI, suggesting that impulsivity induced by yohimbine is KOR dependent. This may suggests a potential role for KOR-mediated increases in impulsivity in yohimbine-induced reinstatement.
Collapse
|
40
|
Flores AJ, Bartlett MJ, Root BK, Parent KL, Heien ML, Porreca F, Polt R, Sherman SJ, Falk T. The combination of the opioid glycopeptide MMP-2200 and a NMDA receptor antagonist reduced l-DOPA-induced dyskinesia and MMP-2200 by itself reduced dopamine receptor 2-like agonist-induced dyskinesia. Neuropharmacology 2018; 141:260-271. [PMID: 30201210 PMCID: PMC6309213 DOI: 10.1016/j.neuropharm.2018.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/01/2018] [Accepted: 09/06/2018] [Indexed: 02/08/2023]
Abstract
Dopamine (DA)-replacement therapy utilizing l-DOPA is the gold standard symptomatic treatment for Parkinson's disease (PD). A critical complication of this therapy is the development of l-DOPA-induced dyskinesia (LID). The endogenous opioid peptides, including enkephalins and dynorphin, are co-transmitters of dopaminergic, GABAergic, and glutamatergic transmission in the direct and indirect striatal output pathways disrupted in PD, and alterations in expression levels of these peptides and their precursors have been implicated in LID genesis and expression. We have previously shown that the opioid glycopeptide drug MMP-2200 (a.k.a. Lactomorphin), a glycosylated derivative of Leu-enkephalin mediates potent behavioral effects in two rodent models of striatal DA depletion. In this study, the mixed mu-delta agonist MMP-2200 was investigated in standard preclinical rodent models of PD and of LID to evaluate its effects on abnormal involuntary movements (AIMs). MMP-2200 showed antiparkinsonian activity, while increasing l-DOPA-induced limb, axial, and oral (LAO) AIMs by ∼10%, and had no effect on dopamine receptor 1 (D1R)-induced LAO AIMs. In contrast, it markedly reduced dopamine receptor 2 (D2R)-like-induced LAO AIMs. The locomotor AIMs were reduced by MMP-2200 in all three conditions. The N-methyl-d-aspartate receptor (NMDAR) antagonist MK-801 has previously been shown to be anti-dyskinetic, but only at doses that induce parkinsonism. When MMP-2200 was co-administered with MK-801, MK-801-induced pro-parkinsonian activity was suppressed, while a robust anti-dyskinetic effect remained. In summary, the opioid glycopeptide MMP-2200 reduced AIMs induced by a D2R-like agonist, and MMP-2200 modified the effect of MK-801 to result in a potent reduction of l-DOPA-induced AIMs without induction of parkinsonism.
Collapse
Affiliation(s)
- Andrew J Flores
- Department of Neurology, The University of Arizona, Tucson, AZ, 85724, USA; Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ, 85724, USA
| | - Mitchell J Bartlett
- Department of Neurology, The University of Arizona, Tucson, AZ, 85724, USA; Graduate Program in Medical Pharmacology, The University of Arizona, Tucson, AZ, 85724, USA
| | - Brandon K Root
- Department of Neurology, The University of Arizona, Tucson, AZ, 85724, USA
| | - Kate L Parent
- Department of Chemistry & Biochemistry and BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
| | - Michael L Heien
- Department of Chemistry & Biochemistry and BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
| | - Frank Porreca
- Department of Pharmacology, The University of Arizona, Tucson, AZ, 85724, USA
| | - Robin Polt
- Department of Chemistry & Biochemistry and BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
| | - Scott J Sherman
- Department of Neurology, The University of Arizona, Tucson, AZ, 85724, USA
| | - Torsten Falk
- Department of Neurology, The University of Arizona, Tucson, AZ, 85724, USA; Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ, 85724, USA; Department of Pharmacology, The University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
41
|
Timing Mechanisms Underlying Gate Control by Feedforward Inhibition. Neuron 2018; 99:941-955.e4. [PMID: 30122375 DOI: 10.1016/j.neuron.2018.07.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 05/20/2018] [Accepted: 07/17/2018] [Indexed: 01/20/2023]
Abstract
The gate control theory proposes that Aβ mechanoreceptor inputs to spinal pain transmission T neurons are gated via feedforward inhibition, but it remains unclear how monosynaptic excitation is gated by disynaptic inhibitory inputs that arrive later. Here we report that Aβ-evoked, non-NMDAR-dependent EPSPs in T neurons are subthreshold, allowing time for inhibitory inputs to prevent action potential firing that requires slow-onset NMDAR activation. Potassium channel activities-including IA, whose sizes are established constitutively by PreprodynorphinCre-derived inhibitory neurons-either completely filter away Aβ inputs or make them subthreshold, thereby creating a permissive condition to achieve gate control. Capsaicin-activated nociceptor inputs reduce IA and sensitize the T neurons, allowing Aβ inputs to cause firing before inhibitory inputs arrive. Thus, distinct kinetics of glutamate receptors and electric filtering by potassium channels solve the timing problem underlying the gating by feedforward inhibition, and their modulation offers a way to bypass the gate control.
Collapse
|
42
|
Munanairi A, Liu XY, Barry DM, Yang Q, Yin JB, Jin H, Li H, Meng QT, Peng JH, Wu ZY, Yin J, Zhou XY, Wan L, Mo P, Kim S, Huo FQ, Jeffry J, Li YQ, Bardoni R, Bruchas MR, Chen ZF. Non-canonical Opioid Signaling Inhibits Itch Transmission in the Spinal Cord of Mice. Cell Rep 2018; 23:866-877. [PMID: 29669290 PMCID: PMC5937707 DOI: 10.1016/j.celrep.2018.03.087] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/28/2018] [Accepted: 03/20/2018] [Indexed: 01/20/2023] Open
Abstract
Chronic itch or pruritus is a debilitating disorder that is refractory to conventional anti-histamine treatment. Kappa opioid receptor (KOR) agonists have been used to treat chronic itch, but the underlying mechanism remains elusive. Here, we find that KOR and gastrin-releasing peptide receptor (GRPR) overlap in the spinal cord, and KOR activation attenuated GRPR-mediated histamine-independent acute and chronic itch in mice. Notably, canonical KOR-mediated Gαi signaling is not required for desensitizing GRPR function. In vivo and in vitro studies suggest that KOR activation results in the translocation of Ca2+-independent protein kinase C (PKC)δ from the cytosol to the plasma membrane, which in turn phosphorylates and inhibits GRPR activity. A blockade of phospholipase C (PLC) in HEK293 cells prevented KOR-agonist-induced PKCδ translocation and GRPR phosphorylation, suggesting a role of PLC signaling in KOR-mediated GRPR desensitization. These data suggest that a KOR-PLC-PKCδ-GRPR signaling pathway in the spinal cord may underlie KOR-agonists-induced anti-pruritus therapies.
Collapse
MESH Headings
- Animals
- Cell Membrane/metabolism
- Chloroquine/toxicity
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- HEK293 Cells
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Phosphorylation
- Protein Kinase C-delta/antagonists & inhibitors
- Protein Kinase C-delta/genetics
- Protein Kinase C-delta/metabolism
- Pruritus/chemically induced
- Pruritus/pathology
- RNA Interference
- RNA, Small Interfering/metabolism
- Receptors, Bombesin/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/deficiency
- Receptors, Opioid, kappa/genetics
- Signal Transduction
- Spinal Cord/metabolism
- Type C Phospholipases/antagonists & inhibitors
- Type C Phospholipases/metabolism
Collapse
Affiliation(s)
- Admire Munanairi
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xian-Yu Liu
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Devin M Barry
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Qianyi Yang
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun-Bin Yin
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anatomy and K. K. Leung Brain Research Centre, The Fourth Military Medical University, 710032 Xi'an, PRC
| | - Hua Jin
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hui Li
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anatomy and K. K. Leung Brain Research Centre, The Fourth Military Medical University, 710032 Xi'an, PRC
| | - Qing-Tao Meng
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jia-Hang Peng
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhen-Yu Wu
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anatomy and K. K. Leung Brain Research Centre, The Fourth Military Medical University, 710032 Xi'an, PRC
| | - Jun Yin
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuan-Yi Zhou
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Li Wan
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anatomy and K. K. Leung Brain Research Centre, The Fourth Military Medical University, 710032 Xi'an, PRC
| | - Ping Mo
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anatomy and K. K. Leung Brain Research Centre, The Fourth Military Medical University, 710032 Xi'an, PRC
| | - Seungil Kim
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Fu-Quan Huo
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph Jeffry
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yun-Qing Li
- Department of Anatomy and K. K. Leung Brain Research Centre, The Fourth Military Medical University, 710032 Xi'an, PRC; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, PRC
| | - Rita Bardoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Michael R Bruchas
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhou-Feng Chen
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
43
|
Analysis of natural product regulation of opioid receptors in the treatment of human disease. Pharmacol Ther 2018; 184:51-80. [DOI: 10.1016/j.pharmthera.2017.10.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
44
|
Lê AD, Funk D, Coen K, Tamadon S, Shaham Y. Role of κ-Opioid Receptors in the Bed Nucleus of Stria Terminalis in Reinstatement of Alcohol Seeking. Neuropsychopharmacology 2018; 43:838-850. [PMID: 28589966 PMCID: PMC5809779 DOI: 10.1038/npp.2017.120] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 12/21/2022]
Abstract
κ-Opioid receptors (KORs) and their endogenous ligand dynorphin are involved in stress-induced alcohol seeking but the mechanisms involved are largely unknown. We previously showed that systemic injections of the KOR agonist U50,488, which induce stress-like aversive states, reinstate alcohol seeking after extinction of the alcohol-reinforced responding. Here, we used the neuronal activity marker Fos and site-specific injections of the KOR antagonist nor-BNI and U50,488 to study brain mechanisms of U50,488-induced reinstatement of alcohol seeking. We trained male Long-Evans rats to self-administer alcohol (12% w/v) for 23-30 days. After extinction of the alcohol-reinforced responding, we tested the effect of U50,488 (0, 1.25, 2.5, and 5 mg/kg) on reinstatement of alcohol seeking. Next, we correlated regional Fos expression with reinstatement induced by the most effective U50,488 dose (5 mg/kg). Based on the correlational Fos results, we determined the effect of bed nucleus of the stria terminalis (BNST) injections of nor-BNI (4 μg/side) on U50,488-induced reinstatement of alcohol seeking, and reinstatement induced by injections of U50,488 (0, 0.3, 1, and 3 μg/side) into the BNST. U50,488-induced reinstatement of alcohol seeking was associated with increased Fos expression in multiple brain areas, including the BNST, where it was significantly correlated with lever pressing. U50,488-induced reinstatement was blocked by BNST nor-BNI injections, and BNST U50,488 injections partially mimicked the drug's systemic effect on reinstatement. Our data indicate that the BNST is a critical site for U50,488-induced reinstatement of alcohol seeking and suggest that KOR/dynorphin mechanisms in this brain area play a key role in stress-induced alcohol seeking.
Collapse
Affiliation(s)
- A D Lê
- Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Douglas Funk
- Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Kathleen Coen
- Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Sahar Tamadon
- Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Yavin Shaham
- Behavioral Neuroscience Branch, Intramural Research Program, NIDA-NIH, Baltimore, MD, USA
| |
Collapse
|
45
|
Dhar D, Poree LR, Yaksh TL. Evolution of the Spinal Delivery of Opiate Analgesics. Neuromodulation 2018. [DOI: 10.1016/b978-0-12-805353-9.00065-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
46
|
Journigan VB, Polgar WE, Tuan EW, Lu J, Daga PR, Zaveri NT. Probing ligand recognition of the opioid pan antagonist AT-076 at nociceptin, kappa, mu, and delta opioid receptors through structure-activity relationships. Sci Rep 2017; 7:13255. [PMID: 29038479 PMCID: PMC5643385 DOI: 10.1038/s41598-017-13129-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/18/2017] [Indexed: 11/15/2022] Open
Abstract
Few opioid ligands binding to the three classic opioid receptor subtypes, mu, kappa and delta, have high affinity at the fourth opioid receptor, the nociceptin/orphanin FQ receptor (NOP). We recently reported the discovery of AT-076 (1), (R)-7-hydroxy-N-((S)-1-(4-(3-hydroxyphenyl)piperidin-1-yl)-3-methylbutan-2-yl)-1,2,3,4-tetrahydroisoquinoline-3-carboxamide, a pan antagonist with nanomolar affinity for all four subtypes. Since AT-076 binds with high affinity at all four subtypes, we conducted a structure-activity relationship (SAR) study to probe ligand recognition features important for pan opioid receptor activity, using chemical modifications of key pharmacophoric groups. SAR analysis of the resulting analogs suggests that for the NOP receptor, the entire AT-076 scaffold is crucial for high binding affinity, but the binding mode is likely different from that of NOP antagonists C-24 and SB-612111 bound in the NOP crystal structure. On the other hand, modifications of the 3-hydroxyphenyl pharmacophore, but not the 7-hydroxy Tic pharmacophore, are better tolerated at kappa and mu receptors and yield very high affinity multifunctional (e.g. 12) or highly selective (e.g. 16) kappa ligands. With the availability of the opioid receptor crystal structures, our SAR analysis of the common chemotype of AT-076 suggests rational approaches to modulate binding selectivity, enabling the design of multifunctional or selective opioid ligands from such scaffolds.
Collapse
MESH Headings
- Humans
- Narcotic Antagonists/chemistry
- Narcotic Antagonists/pharmacology
- Opioid Peptides/chemistry
- Receptors, Opioid/chemistry
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/chemistry
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/chemistry
- Structure-Activity Relationship
- Nociceptin
Collapse
Affiliation(s)
- V Blair Journigan
- Astraea Therapeutics, 320 Logue Avenue, Suite 142, Mountain View, CA, 94043, USA
- Marshall University School of Pharmacy, Department of Pharmaceutical Sciences, One John Marshall Drive, Huntington, WV 25755, USA
| | - Willma E Polgar
- Astraea Therapeutics, 320 Logue Avenue, Suite 142, Mountain View, CA, 94043, USA
| | - Edward W Tuan
- Astraea Therapeutics, 320 Logue Avenue, Suite 142, Mountain View, CA, 94043, USA
| | - James Lu
- Astraea Therapeutics, 320 Logue Avenue, Suite 142, Mountain View, CA, 94043, USA
| | - Pankaj R Daga
- Astraea Therapeutics, 320 Logue Avenue, Suite 142, Mountain View, CA, 94043, USA
| | - Nurulain T Zaveri
- Astraea Therapeutics, 320 Logue Avenue, Suite 142, Mountain View, CA, 94043, USA.
| |
Collapse
|
47
|
Helal MA, Habib ES, Chittiboyina AG. Selective kappa opioid antagonists for treatment of addiction, are we there yet? Eur J Med Chem 2017; 141:632-647. [PMID: 29107424 DOI: 10.1016/j.ejmech.2017.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/06/2017] [Accepted: 10/07/2017] [Indexed: 11/30/2022]
Abstract
Kappa opioid receptor (KOP) is a G-protein coupled receptor mainly expressed in the cerebral cortex and hypothalamus. It is implicated in nociception, diuresis, emotion, cognition, and immune system functions. KOP agonists possess a strong analgesic effect accompanied by a feeling of dysphoria. On the other hand, antagonists of this receptor were found to block depression, anxiety, and drug-seeking behaviors in animal models. Recently, great interest has been given to the development of selective KOP antagonists as an addiction treatment that does not cause dependence itself or show high relapse rates like the currently used agents. This review provides a comprehensive survey of the KOP antagonists developed for this purpose together with their in vivo studies and clinical trials. In addition, a future perspective and recommendations for the work needed to develop clinically relevant KOP antagonists are presented.
Collapse
Affiliation(s)
- Mohamed A Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza 12588, Egypt; Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| | - Eman S Habib
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Amar G Chittiboyina
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| |
Collapse
|
48
|
Owens SM, Pollard GT, Howard JL, Fennell TR, Snyder RW, Carroll FI. Pharmacodynamic Relationships between Duration of Action of JDTic-like Kappa-Opioid Receptor Antagonists and Their Brain and Plasma Pharmacokinetics in Rats. ACS Chem Neurosci 2016; 7:1737-1745. [PMID: 27700049 DOI: 10.1021/acschemneuro.6b00249] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
JDTic is a potent and selective κ-opioid receptor (KOR) antagonist that reverses U50,488-induced diuresis in rats. It partitions into brain with a duration of action lasting for weeks. In a search for KOR antagonists that do not accumulate in the brain, we compared single doses of five methylated JDTic analogs (RTI-97, -194, -212, -240, and -241) for reversal of U50,488 diuresis and pharmacokinetic (PK) properties. All six compounds showed potent and selective KOR antagonism in a [35S]GTPγS binding assay. Plasma half-lives ranged from 24 to 41 h and brain half-lives from 24 to 76 h. JDTic and RTI-194 showed increasing brain to plasma ratios over time, indicating increasing partitioning into brain and a longer duration of action for reversal of diuresis than did RTI-97. RTI-240 did not show significant brain accumulation. RTI-212 showed no substantive difference between brain and plasma levels and was inactive against diuresis. RTI-241, with a lower brain to plasma ratio than JDTic and RTI-194, formed JDTic as a metabolite, which still reduced diuresis after 9 weeks. The fact that the duration of action was correlated with the brain to blood plasma ratios and area under the concentration-time curves suggests that PK properties could help to predict safety and acceptable duration of action for KOR antagonists.
Collapse
Affiliation(s)
- S. Michael Owens
- University of Arkansas for Medical Sciences, College
of Medicine, Department of Pharmacology and Toxicology, Little Rock, Arkansas 72205, United States
| | - Gerald T. Pollard
- Howard Associates, LLC, 3040
East Cornwallis Road, P.O. Box 12194, Research
Triangle Park, North Carolina 27709-2194, United States
| | - James L. Howard
- Howard Associates, LLC, 3040
East Cornwallis Road, P.O. Box 12194, Research
Triangle Park, North Carolina 27709-2194, United States
| | - Timothy R. Fennell
- Research Triangle Institute, 3040
East Cornwallis Road, P.O. Box 12194, Research
Triangle Park, North Carolina 27709-2194, United States
| | - Rodney W. Snyder
- Research Triangle Institute, 3040
East Cornwallis Road, P.O. Box 12194, Research
Triangle Park, North Carolina 27709-2194, United States
| | - F. Ivy Carroll
- Research Triangle Institute, 3040
East Cornwallis Road, P.O. Box 12194, Research
Triangle Park, North Carolina 27709-2194, United States
| |
Collapse
|
49
|
Lopez JA, Bedenbaugh MN, McCosh RB, Weems PW, Meadows LJ, Wisman B, Coolen LM, Goodman RL, Hileman SM. Does Dynorphin Play a Role in the Onset of Puberty in Female Sheep? J Neuroendocrinol 2016; 28:10.1111/jne.12445. [PMID: 28328155 PMCID: PMC5412962 DOI: 10.1111/jne.12445] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 10/05/2016] [Accepted: 11/01/2016] [Indexed: 11/30/2022]
Abstract
Puberty onset involves increased gonadotrophin-release (GnRH) release as a result of decreased sensitivity to oestrogen (E2 )-negative feedback. Because GnRH neurones lack E2 receptor α, this pathway must contain interneurones. One likely candidate is KNDy neurones (kisspeptin, neurokinin B, dynorphin). The overarching hypothesis of the present study was that the prepubertal hiatus in luteinising hormone (LH) release involves reduced kisspeptin and/or heightened dynorphin input. We first tested the specific hypothesis that E2 would reduce kisspeptin-immunopositive cell numbers and increase dynorphin-immunopositive cell numbers. We found that kisspeptin cell numbers were higher in ovariectomised (OVX) lambs than OVX lambs treated with E2 (OVX+ E2 ) or those left ovary-intact. Very few arcuate dynorphin cells were identified in any group. Next, we hypothesised that central blockade of κ-opioid receptor (KOR) would increase LH secretion at a prepubertal (6 months) but not postpubertal (10 months) age. Luteinising hormone pulse frequency and mean LH increased during infusion of a KOR antagonist, norbinaltorphimine, in OVX + E2 lambs at the prepubertal age but not in the same lambs at the postpubertal age. We next hypothesised that E2 would increase KOR expression in GnRH neurones or alter synaptic input to KNDy neurones in prepubertal ewes. Oestrogen treatment decreased the percentage of GnRH neurones coexpressing KOR (approximately 68%) compared to OVX alone (approximately 78%). No significant differences in synaptic contacts per cell between OVX and OVX + E2 groups were observed. Although these initial data are consistent with dynorphin inhibiting pulsatile LH release prepubertally, additional work will be necessary to define the source and mechanisms of this inhibition.
Collapse
Affiliation(s)
- J A Lopez
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - M N Bedenbaugh
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - R B McCosh
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - P W Weems
- Department of Neurobiology and Anatomical Sciences, The University of Mississippi Medical Center, Jackson, MS, USA
| | - L J Meadows
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - B Wisman
- Department of Biology, Alderson-Broaddus University, Philippi, WV, USA
| | - L M Coolen
- Department of Neurobiology and Anatomical Sciences, The University of Mississippi Medical Center, Jackson, MS, USA
| | - R L Goodman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - S M Hileman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
50
|
Jamshidi RJ, Sullivan LC, Jacobs BA, Chavera TA, Berg KA, Clarke WP. Long-Term Reduction of Kappa Opioid Receptor Function by the Biased Ligand, Norbinaltorphimine, Requires c-Jun N-Terminal Kinase Activity and New Protein Synthesis in Peripheral Sensory Neurons. J Pharmacol Exp Ther 2016; 359:319-328. [PMID: 27605628 PMCID: PMC5074480 DOI: 10.1124/jpet.116.235184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 09/02/2016] [Indexed: 01/10/2023] Open
Abstract
A single administration of the κ opioid receptor (KOR) antagonist, norbinaltorphimine (norBNI), produces long-term reduction in KOR function in heterologous expression systems and brain that is mediated by activation of c-Jun N-terminal kinase (JNK). In this study, we examined the long-term effects of norBNI on adult rat peripheral sensory neurons in vivo and ex vivo. Following a single intraplantar (i.pl.) injection of norBNI into the hind paw, peripheral KOR-mediated antinociception in the ipsilateral, but not the contralateral, hindpaw was abolished for at least 9 days. By contrast, the antinociceptive response to mu and delta opioid receptor agonists was unaltered. The long-term inhibitory effect on antinociception produced by pretreatment with norBNI required occupancy of peripheral KOR and was completely blocked by i.pl. injection of the JNK inhibitor, SP600125. In cultures of peripheral sensory neurons, norBNI activated JNK for at least 30 minutes. Furthermore, norBNI blocked KOR-mediated inhibition of adenylyl cyclase activity measured 24 hours later in a JNK-dependent manner, but did not block activation of extracellular signal-regulated kinase (ERK). The long-term inhibitory effect of norBNI on KOR function in vivo and ex vivo was blocked by inhibitors of mRNA translation, cycloheximide and rapamycin. These data suggest that in peripheral sensory neurons norBNI is a KOR-biased ligand for activation of JNK signaling, resulting in long-term blockade of some (antinociception, inhibition of adenylyl cyclase activity), but not all (ERK), KOR signaling. Importantly, norBNI elicits de novo protein synthesis in sensory neuron terminals that produces selective long-term regulation of KOR.
Collapse
Affiliation(s)
- Raehannah J Jamshidi
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Laura C Sullivan
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Blaine A Jacobs
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Teresa A Chavera
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Kelly A Berg
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - William P Clarke
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|