1
|
Rogers A, Castro EM, Lotfipour S, Leslie FM. Dynorphinergic lateral hypothalamus to posterior ventral tegmental area pathway matures after adolescence in male rats. Neuropharmacology 2025; 270:110350. [PMID: 39938860 DOI: 10.1016/j.neuropharm.2025.110350] [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: 11/25/2024] [Revised: 01/23/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
The highly plastic nature of the adolescent brain is well-known, and is thought to contribute to the unique susceptibility of adolescents to drugs of abuse. However, much investigation of adolescent plasticity has been focused on synaptic plasticity, as synapses are strengthened and pruned. Here, we show that dynorphin+ neurons in the lateral hypothalamus of adolescent male rats do not respond to low doses of intravenous combined nicotine + ethanol, while male adult lateral hypothalamus dynorphin+ neurons do. We also provide evidence that the dynorphinergic projection from the lateral hypothalamus to the posterior ventral tegmental area is not present in adolescent males, suggesting that axons are still extending during this time. Together, these results suggest a mechanism for the increased susceptibility of adolescent male rats to drug reward.
Collapse
Affiliation(s)
- Alexandra Rogers
- Department of Pharmaceutical Sciences, University of California, Irvine, 856 Health Sciences Road, Suite 5400, 92697-3958, Irvine, CA, USA.
| | - Emily M Castro
- Department of Pharmaceutical Sciences, University of California, Irvine, 856 Health Sciences Road, Suite 5400, 92697-3958, Irvine, CA, USA
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, University of California, Irvine, 856 Health Sciences Road, Suite 5400, 92697-3958, Irvine, CA, USA
| | - Frances M Leslie
- Department of Pharmaceutical Sciences, University of California, Irvine, 856 Health Sciences Road, Suite 5400, 92697-3958, Irvine, CA, USA
| |
Collapse
|
2
|
Mathew SJ, Cutler AJ, Visitacion NC, Gold M, Yuan J, Aurora B. Navacaprant, a Novel and Highly Selective Kappa Opioid Receptor Antagonist, in Adults With Major Depressive Disorder: A Randomized, Double-Blind Phase 2 Clinical Trial. J Clin Psychopharmacol 2025:00004714-990000000-00378. [PMID: 40199329 DOI: 10.1097/jcp.0000000000001967] [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: 04/10/2025]
Abstract
PURPOSE/BACKGROUND This phase 2a randomized, double-blind, placebo-controlled, 8-week trial assessed the efficacy and safety of navacaprant, a highly selective kappa opioid receptor antagonist, in adults with major depressive disorder (MDD). METHODS/PROCEDURES Participants with 17-Item Hamilton Depression Rating Scale (HAMD-17) scores of 14 to 30 were randomized 1:1 to once-daily navacaprant 80 mg or placebo (n = 102 each). The primary endpoint was HAMD-17 change from baseline (CFB) to week 8. Secondary endpoints included CFB in Snaith-Hamilton Pleasure Scale (SHAPS). No adjustment for multiple comparisons was made. FINDINGS/RESULTS At week 8, HAMD-17 CFB was not statistically significantly improved with navacaprant vs placebo (least squares mean difference -1.7 [standard error, 1.08], P = 0.121; mixed-models repeated-measures) in the efficacy population. In a prespecified sensitivity analysis using last-observation-carried-forward, navacaprant statistically significantly improved HAMD-17 CFB (-2.9 [0.88], P = 0.002; -2.2 [0.98], P = 0.024) and SHAPS CFB (-2.8 [0.96], P = 0.004; -3.4 [1.10], P = 0.002) vs placebo at weeks 4 and 8. In the prespecified subgroup with moderate-to-severe MDD (baseline HAMD-17 score ≥22; n = 100), navacaprant statistically significantly improved HAMD-17 CFB at both timepoints (-3.0 [1.20], P = 0.015; -2.8 [1.33], P = 0.037) and SHAPS CFB at week 8 (-4.8 [1.35], P = 0.001) vs placebo. Most frequently reported adverse events (AEs) included headache (4.9% both) and nausea (4.9% navacaprant, 1.0% placebo); no serious AEs were reported with navacaprant. IMPLICATIONS/CONCLUSIONS Although the primary endpoint was not met in the efficacy population, which included participants with mild depression, statistically significant improvements with navacaprant on depressive symptoms including anhedonia in the moderate-to-severe MDD subgroup, along with a favorable safety profile, support further study of navacaprant for the treatment of MDD.
Collapse
Affiliation(s)
- Sanjay J Mathew
- From the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX
| | - Andrew J Cutler
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY
| | | | | | - Jason Yuan
- Neumora Therapeutics, Inc., Watertown, MA
| | | |
Collapse
|
3
|
Spodnick MB, McElderry SC, Diaz MR. Opioid receptor signaling throughout ontogeny: Shaping neural and behavioral trajectories. Neurosci Biobehav Rev 2025; 170:106033. [PMID: 39894419 PMCID: PMC11851333 DOI: 10.1016/j.neubiorev.2025.106033] [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: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Due to the recent and ongoing opioid crisis in the United States, exposure to opioid drugs in utero is becoming more common, including during medication-assisted therapy used to treat opioid use disorder. As such, careful consideration of opioidergic signaling in utero and beyond, as well as alterations to this signaling via introduction of exogenous opioids, is warranted. This review explores the ontogeny and function of the Mu, Kappa and Delta opioid receptor systems throughout the lifespan, highlighting their importance in guiding neurobehavioral development. We argue for a paradigm shift in conceptualization of opioids as not only contributors within their own system, but also vital regulators of a multitude of downstream neurodevelopmental processes.
Collapse
Affiliation(s)
- Mary B Spodnick
- Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, USA.
| | | | - Marvin R Diaz
- Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, USA.
| |
Collapse
|
4
|
Marchette RCN, Vendruscolo LF, Koob GF. The Dynorphin/-Opioid Receptor System at the Interface of Hyperalgesia/Hyperkatifeia and Addiction. CURRENT ADDICTION REPORTS 2025; 12:11. [PMID: 40124896 PMCID: PMC11925990 DOI: 10.1007/s40429-025-00618-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 03/25/2025]
Abstract
Purpose of Review Drug addiction is characterized by compulsive drug seeking and use, accompanied by negative emotional states (hyperkatifeia) and heightened pain sensitivity (hyperalgesia) during withdrawal. Both hyperalgesia and hyperkatifeia are integral components of substance use disorders, negatively impacting treatment and recovery. The underlying neurobiological mechanisms of hyperalgesia and hyperkatifeia involve alterations of brain reward and stress circuits, including the dynorphin/κ-opioid receptor (KOR) system. The dynorphin/KOR system modulates pain perception, negative affect, and addictive behaviors. Here, we review the preclinical evidence of dynorphin/KOR signaling in opioid withdrawal-induced hyperalgesia and hyperkatifeia. Recent Findings In opioid dependence models, pharmacological and genetic interventions of the dynorphin/KOR system attenuate somatic and motivational signs of withdrawal and addictive-like behaviors, highlighting its therapeutic potential. Understanding the intricate interplay between dynorphin/KOR signaling, hyperalgesia, hyperkatifeia, and addiction offers novel insights into treatment strategies for opioid use disorder and other substance use disorders. Summary Further research is needed to elucidate precise mechanisms of the sexual dimorphism of dynorphin/KOR signaling and identify targeted interventions to mitigate hyperalgesia and hyperkatifeia and facilitate recovery from addiction.
Collapse
Affiliation(s)
- Renata C. N. Marchette
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, BRC Room 08A505.19, 251 Bayview Blvd, Baltimore, MD 21224 USA
| | - Leandro F. Vendruscolo
- Stress and Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, Division of Intramural Clinical and Biological Research, National Institute on Drug Abuse, Intramural Research Program, and National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, MD 21224 USA
| | - George F. Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, BRC Room 08A505.19, 251 Bayview Blvd, Baltimore, MD 21224 USA
| |
Collapse
|
5
|
van de Wetering R, Vu LY, Kornberger LD, Luo D, Scouller B, Hong S, Paton K, Prisinzano TE, Kivell BM. Effects of Biased Analogues of the Kappa Opioid Receptor Agonist, U50,488, in Preclinical Models of Pain and Side Effects. Molecules 2025; 30:604. [PMID: 39942708 PMCID: PMC11820436 DOI: 10.3390/molecules30030604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Kappa opioid receptor (KOR) agonists have well-established antinociceptive effects. However, many KOR agonists have negative side effects, which limit their therapeutic potential. Some researchers have suggested that the development of biased agonists that preferentially stimulate KOR G-protein pathways over β-arrestin pathways may yield drugs with fewer adverse side effects. This was investigated in the current study. We describe the synthesis and characterization of three U50,488 analogues, 1, 2, and 3. We evaluated the acute and chronic antinociceptive effects of these compounds in mice using the warm-water tail flick assay and in a paclitaxel-induced neuropathic pain model. Side effects were investigated using open-field, passive wire hang, rotarod, elevated zero maze, conditioned place aversion, and whole-body plethysmography, with some tests being conducted in KOR or β-arrestin2 knock out mice. All compounds were highly potent, full agonists of the KOR, with varying signaling biases in vitro. In the warm-water tail withdrawal assay, these agonists were ~10 times more potent than U50,488, but not more efficacious. All KOR agonists reversed paclitaxel-induced neuropathic pain, without tolerance. Compound 3 showed no significant side effects on any test. Signaling bias did not correlate with the antinociceptive or side effects of any compounds and knockout of β-arrestin2 had no effect on U50,488-induced sedation or motor incoordination. These findings highlight the therapeutic potential of 3, with its lack of side effects typically associated with KOR agonists, and also suggest that G-protein signaling bias is a poor predictor of KOR agonist-induced side effects.
Collapse
MESH Headings
- Animals
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Mice
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/adverse effects
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/chemistry
- Disease Models, Animal
- Neuralgia/drug therapy
- Neuralgia/chemically induced
- Male
- Mice, Knockout
- Paclitaxel/adverse effects
- Humans
- Analgesics/pharmacology
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Loan Y. Vu
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Lindsay D. Kornberger
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Dan Luo
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Brittany Scouller
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Sheein Hong
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Kelly Paton
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Thomas E. Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Bronwyn M. Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| |
Collapse
|
6
|
Salazar AL, Centanni SW. Sex Differences in Mouse Models of Voluntary Alcohol Drinking and Abstinence-Induced Negative Emotion. Alcohol 2024; 121:45-57. [PMID: 39053705 PMCID: PMC11637945 DOI: 10.1016/j.alcohol.2024.07.004] [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: 04/15/2024] [Revised: 07/03/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Alcohol Use Disorder (AUD) is a growing problem worldwide, causing an incredible burden on health and the economy. Though AUD impacts people of all backgrounds and demographics, increasing evidence has suggested robust sex differences in alcohol drinking patterns and AUD-induced negative emotionality or hyperkatifeia. Rates of problematic drinking have significantly risen among women, and women face more severe negative emotional consequences in abstinence such as increased risk of comorbidity with an anxiety or mood disorder and more severe symptoms of depression. As such, a bevy of preclinical literature using contingent methods of alcohol (ethanol) consumption has amassed in recent years to better understand sex as a biological variable in alcohol drinking and abstinence-induced negative emotionality. Mice are widely used to model alcohol drinking, as they are conducive to genetic manipulation strategies, and many strains will voluntarily consume alcohol. Sex-specific results from these mouse studies, however, have been inconsistent. Therefore, this review aims to summarize the current knowledge on sex differences in AUD-related contingent ethanol drinking and abstinence-induced negative emotionality in mice. Various contingent mouse drinking models and negative emotional-based behavioral paradigms are introduced and subsequently discussed in the context of sex differences to show increasing indications of sex specificity in mouse preclinical studies of AUD. With this review, we hope to inform future research on potential sex differences in preclinical mouse models of AUD and provide mounting evidence supporting the need for more widespread inclusion of preclinical female subjects in future studies.
Collapse
Affiliation(s)
- Amanda L Salazar
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Samuel W Centanni
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA.
| |
Collapse
|
7
|
Namchuk AB, Tsuda MC, Lucki I, Browne CA. Kappa opioid receptor mediated operant performance in male and female rats. Pharmacol Biochem Behav 2024; 244:173847. [PMID: 39151827 DOI: 10.1016/j.pbb.2024.173847] [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: 06/19/2024] [Revised: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Anhedonia and avolition are emotions frequently endorsed by individuals with stress related disorders. Kappa opioid receptor (KOR) activation can induce negative emotions and recent clinical evidence suggests that KOR antagonism can alleviate anhedonia in a transdiagnostic cohort of patients. However, the behavioral consequences of KOR activation and antagonism in modulating motivation, as assessed by schedule-controlled behavioral performance without preexisting conditions (stress or substance use), have not been formally assessed. To address this gap in the literature, this report utilized male and female Sprague Dawley rats to (1) evaluate the impact of the selective KOR agonist U50,488, on the performance of animals responding for sucrose pellets under a progressive ratio (PR) schedule and (2) determine the effects of the short-acting KOR antagonist LY2444296 alone and on U50,488 mediated reductions in PR performance. Overall, U50,488 5 mg/kg significantly reduced the breakpoint and number of rewards obtained by animals. This occurred in the absence of motor impairment and independent of evidence for satiation. LY2444296 did not alter PR performance when administered alone but effectively blocked the deficits induced by U50,488. To further delineate the behavioral alterations that underlie these reductions in responding, a more detailed analysis was conducted on PR performance in the first 15 min of the session, the period of time when animals obtained the most reinforcers. During this period, U50,488 increased the length of the post-reinforcement pause and reduced the running rate on PR schedules. These changes in behavior produced by acute activation of KORs are consistent with a reduction of effort-related motivation in rodents. These data contribute to the understanding of how KORs modulate motivation, which is critical to future efforts to evaluate performance in the context of stress and assess how KOR antagonists alleviate anhedonic behaviors associated with stress.
Collapse
Affiliation(s)
- Amanda B Namchuk
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Mumeko C Tsuda
- Preclinical Behavior & Modelling Core, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America; Preclinical Behavior & Modelling Core, Uniformed Services University, Bethesda, MD 20814, United States of America; Department of Psychiatry, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Caroline A Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America.
| |
Collapse
|
8
|
Lepreux G, Henricks AM, Wei G, Go BS, Erikson CM, Abella RM, Pham A, Walker BM. Kappa-opioid receptor antagonism in the nucleus accumbens shell distinguishes escalated alcohol consumption and negative affective-like behavior from physiological withdrawal in alcohol-dependence. Pharmacol Biochem Behav 2024; 243:173840. [PMID: 39096973 DOI: 10.1016/j.pbb.2024.173840] [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: 05/29/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Alcohol use disorder (AUD) is a chronic relapsing disease that is deleterious at individual, familial, and societal levels. Although AUD is one of the highest preventable causes of death in the USA, therapies for the treatment of AUD are not sufficient given the heterogeneity of the disorder and the limited number of approved medications. To provide better pharmacological strategies, it is important to understand the neurological underpinnings of AUD. Evidence implicates the endogenous dynorphin (DYN)/κ-opioid receptor (KOR) system recruitment in dysphoric and negative emotional states in AUD to promote maladaptive behavioral regulation. The nucleus accumbens shell (AcbSh), mediating motivational and emotional processes that is a component of the mesolimbic dopamine system and the extended amygdala, is an important site related to alcohol's reinforcing actions (both positive and negative) and neuroadaptations in the AcbSh DYN/KOR system have been documented to induce maladaptive symptoms in AUD. We have previously shown that in other nodes of the extended amygdala, site-specific KOR antagonism can distinguish different symptoms of alcohol dependence and withdrawal. In the current study, we examined the role of the KOR signaling in the AcbSh of male Wistar rats in operant alcohol self-administration, measures of negative affective-like behavior, and physiological symptoms during acute alcohol withdrawal in alcohol-dependence. To induce alcohol dependence, rats were exposed to chronic intermittent ethanol vapor for 14 h/day for three months, during which stable escalation of alcohol self-administration was achieved and pharmacological AcbSh KOR antagonism ensued. The results showed that AcbSh KOR antagonism significantly reduced escalated alcohol intake and negative affective-like states but did not alter somatic symptoms of withdrawal. Understanding the relative contribution of these different drivers is important to understand and inform therapeutic efficacy approaches in alcohol dependence and further emphasis the importance of the KOR/DYN system as a target for AUD therapeutics.
Collapse
Affiliation(s)
- Gaetan Lepreux
- Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Angela M Henricks
- Department of Psychology, Washington State University, Pullman, WA, USA
| | - Gengze Wei
- Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Department of Psychology, Washington State University, Pullman, WA, USA
| | - Bok Soon Go
- Department of Life Sciences, Korea University, Seoul, South Korea
| | - Chloe M Erikson
- Department of Psychology, Washington State University, Pullman, WA, USA
| | - Rachel M Abella
- Department of Psychology, Washington State University, Pullman, WA, USA
| | - Amy Pham
- Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Brendan M Walker
- Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Department of Psychology, Washington State University, Pullman, WA, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; USF Neuroscience Institute, USF Health, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
9
|
Morrison FG, Van Orden LJ, Zeitz K, Kuijer EJ, Smith SL, Heal DJ, Wallace TL. Navacaprant, a novel and selective kappa opioid receptor antagonist, has no agonist properties implicated in opioid-related abuse. Neuropharmacology 2024; 257:110037. [PMID: 38876309 DOI: 10.1016/j.neuropharm.2024.110037] [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: 11/21/2023] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Kappa opioid receptors (KORs) are implicated in the pathophysiology of various psychiatric and neurological disorders creating interest in targeting the KOR system for therapeutic purposes. Accordingly, navacaprant (NMRA-140) is a potent, selective KOR antagonist being evaluated as a treatment for major depressive disorder. In the present report, we have extended the pharmacological characterization of navacaprant by further demonstrating its selective KOR antagonist properties and confirming its lack of agonist activity at KORs and related targets involved in opioid-related abuse. Using CHO-K1 cells expressing human KOR, mu (MOR), or delta (DOR) opioid receptors, navacaprant demonstrated selective antagonist properties at KOR (IC50 = 0.029 μM) versus MOR (IC50 = 3.3 μM) and DOR (IC50 > 10 μM) in vitro. In vivo, navacaprant (10-30 mg/kg, i.p.) dose-dependently abolished KOR-agonist induced analgesia in the mouse tail-flick assay. Additionally, navacaprant (10, 30 mg/kg, p.o.) significantly reduced KOR agonist-stimulated prolactin release in mice and rats, confirming KOR antagonism in vivo. Navacaprant showed no agonist activity at any opioid receptor subtype (EC50 > 10 μM) in vitro and exhibited no analgesic effect in the tail-flick assays at doses ≤100 mg/kg, p.o. thereby confirming a lack of opioid receptor agonist activity in vivo. Importantly, navacaprant did not alter extracellular dopamine concentrations in the nucleus accumbens shell of freely-moving rats following doses ≤100 mg/kg, p.o., whereas morphine (10, 20 mg/kg, i.p.) significantly increased dopamine levels. These results demonstrate that navacaprant is a KOR-selective antagonist with no pharmacological properties implicated in opioid-related abuse.
Collapse
MESH Headings
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Animals
- CHO Cells
- Cricetulus
- Humans
- Male
- Mice
- Rats
- Analgesics, Opioid/pharmacology
- Cricetinae
- Opioid-Related Disorders/drug therapy
- Narcotic Antagonists/pharmacology
- Dose-Response Relationship, Drug
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Nucleus Accumbens/drug effects
- Nucleus Accumbens/metabolism
- Mice, Inbred C57BL
- Dopamine/metabolism
Collapse
Affiliation(s)
| | | | - Karla Zeitz
- Neumora Therapeutics, Inc., 490 Arsenal Way, Watertown, MA, 02472, USA
| | - Eloise J Kuijer
- Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
| | | | - David J Heal
- Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK; DevelRx Ltd., BioCity, Nottingham, NG1 1GF, UK
| | - Tanya L Wallace
- Neumora Therapeutics, Inc., 490 Arsenal Way, Watertown, MA, 02472, USA.
| |
Collapse
|
10
|
Hampsey E, Jelen L, Young AH. Aticaprant: (a κ-opioid receptor antagonist) for major depressive disorder. Expert Opin Emerg Drugs 2024; 29:193-204. [PMID: 38682267 DOI: 10.1080/14728214.2024.2345645] [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: 02/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION Major depression is a common, disabling mental health condition associated with the highest disease burden for any neuropsychiatric disorder worldwide, according to the WHO. Due to the imperfect efficacy and tolerability profiles of existing treatments, investigational compounds in novel treatment classes are needed. Opioid-receptor antagonists are a potential new class of treatments currently under investigation. AREAS COVERED Major depressive disorder is first overviewed. Existing treatments, both their mechanisms of action and their place within the antidepressant space, are discussed herein. Then, the profile of Aticaprant and the wider context of kappa-opioid antagonism for depression are discussed in focus. EXPERT OPINION Early evidence indicates that Aticaprant may possess desirable pharmacodynamic and pharmacokinetic properties. A lack of convincing efficacy data at the time of writing precludes any definitive statement on its potential as an antidepressant.
Collapse
Affiliation(s)
- Elliot Hampsey
- Centre for Affective Disorders, King's College London, London, UK
| | - Luke Jelen
- Centre for Affective Disorders, King's College London, London, UK
| | - Allan H Young
- Centre for Affective Disorders, King's College London, London, UK
- South London & Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
11
|
Cole RH, Moussawi K, Joffe ME. Opioid modulation of prefrontal cortex cells and circuits. Neuropharmacology 2024; 248:109891. [PMID: 38417545 PMCID: PMC10939756 DOI: 10.1016/j.neuropharm.2024.109891] [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: 11/10/2023] [Revised: 01/30/2024] [Accepted: 02/26/2024] [Indexed: 03/01/2024]
Abstract
Several neurochemical systems converge in the prefrontal cortex (PFC) to regulate cognitive and motivated behaviors. A rich network of endogenous opioid peptides and receptors spans multiple PFC cell types and circuits, and this extensive opioid system has emerged as a key substrate underlying reward, motivation, affective behaviors, and adaptations to stress. Here, we review the current evidence for dysregulated cortical opioid signaling in the pathogenesis of psychiatric disorders. We begin by providing an introduction to the basic anatomy and function of the cortical opioid system, followed by a discussion of endogenous and exogenous opioid modulation of PFC function at the behavioral, cellular, and synaptic level. Finally, we highlight the therapeutic potential of endogenous opioid targets in the treatment of psychiatric disorders, synthesizing clinical reports of altered opioid peptide and receptor expression and activity in human patients and summarizing new developments in opioid-based medications. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Rebecca H Cole
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience University of Pittsburgh, Pittsburgh, PA, USA
| | - Khaled Moussawi
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience University of Pittsburgh, Pittsburgh, PA, USA
| | - Max E Joffe
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
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
|
13
|
Varastehmoradi B, Smith KL, Müller HK, Elfving B, Sanchez C, Wegener G. Kappa opioid activation changes protein profiles in different regions of the brain relevant to depression. Eur Neuropsychopharmacol 2023; 72:9-17. [PMID: 37040689 DOI: 10.1016/j.euroneuro.2023.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/18/2023] [Accepted: 03/23/2023] [Indexed: 04/13/2023]
Abstract
Depression is a widespread disorder with a significant burden on individuals and society. There are various available treatments for patients with depression. However, not all patients respond adequately to their treatment. Recently, the opioid system has regained interest in depression studies. Research in animals and humans suggest that blocking the kappa opioid receptor (KOR) may potentially alleviate the symptoms of depression. The mechanism behind this effect is not fully understood. Stress and alterations in hypothalamic-pituitary-adrenal axis (HPA-axis) activity are thought to play a crucial role in depression. This study aimed to characterize stress hormones and stress-related protein expression following activation of KOR using a selective agonist. The longitudinal effect was investigated 24 h after KOR activation using the selective agonist U50,488 in Sprague Dawley rats. Stress-related hormones and protein expression patterns were explored using multiplex bead-based assays and western blotting. We found that KOR activation caused an increase in both adrenocorticotropic hormone (ACTH) and corticosterone (CORT) in serum. Regarding protein assays in different brain regions, phosphorylated glucocorticoid receptors also increased significantly in thalamus (THL), hypothalamus (HTH), and striatum (STR). C-Fos increased time-dependently in THL following KOR activation, extracellular signal-regulated kinases 1/2 (ERK1/2) increased significantly in STR and amygdala (AMG), while phosphorylated ERK1/2 decreased during the first 2 h and then increased again in AMG and prefrontal cortex (PFC). This study shows that KOR activation alters the HPA axis and ERK signaling which may cause to develop mood disorders.
Collapse
Affiliation(s)
- Bardia Varastehmoradi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karen L Smith
- Alkermes, Inc., Biology, Waltham, MA, United States of America
| | - Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Alkermes, Inc., Biology, Waltham, MA, United States of America
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
14
|
van de Wetering R, Ewald A, Welsh S, Kornberger L, Williamson SE, McElroy BD, Butelman ER, Prisinzano TE, Kivell BM. The Kappa Opioid Receptor Agonist 16-Bromo Salvinorin A Has Anti-Cocaine Effects without Significant Effects on Locomotion, Food Reward, Learning and Memory, or Anxiety and Depressive-like Behaviors. Molecules 2023; 28:4848. [PMID: 37375403 PMCID: PMC10304272 DOI: 10.3390/molecules28124848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Kappa opioid receptor (KOR) agonists have preclinical antipsychostimulant effects; however, adverse side effects have limited their therapeutic development. In this preclinical study, conducted in Sprague Dawley rats, B6-SJL mice, and non-human primates (NHPs), we evaluated the G-protein-biased analogue of salvinorin A (SalA), 16-bromo salvinorin A (16-BrSalA), for its anticocaine effects, side effects, and activation of cellular signaling pathways. 16-BrSalA dose-dependently decreased the cocaine-primed reinstatement of drug-seeking behavior in a KOR-dependent manner. It also decreased cocaine-induced hyperactivity, but had no effect on responding for cocaine on a progressive ratio schedule. Compared to SalA, 16-BrSalA had an improved side effect profile, with no significant effects in the elevated plus maze, light-dark test, forced swim test, sucrose self-administration, or novel object recognition; however, it did exhibit conditioned aversive effects. 16-BrSalA increased dopamine transporter (DAT) activity in HEK-293 cells coexpressing DAT and KOR, as well as in rat nucleus accumbens and dorsal striatal tissue. 16-BrSalA also increased the early phase activation of extracellular-signal-regulated kinases 1 and 2, as well as p38 in a KOR-dependent manner. In NHPs, 16-BrSalA caused dose-dependent increases in the neuroendocrine biomarker prolactin, similar to other KOR agonists, at doses without robust sedative effects. These findings highlight that G-protein-biased structural analogues of SalA can have improved pharmacokinetic profiles and fewer side effects while maintaining their anticocaine effects.
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Amy Ewald
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Susan Welsh
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Lindsay Kornberger
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA (T.E.P.)
| | - Samuel E. Williamson
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bryan D. McElroy
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Eduardo R. Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Thomas E. Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA (T.E.P.)
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bronwyn M. Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| |
Collapse
|
15
|
Lepreux G, Shinn GE, Wei G, Suko A, Concepcion G, Sirohi S, Soon Go B, Bruchas MR, Walker BM. Recapitulating phenotypes of alcohol dependence via overexpression of Oprk1 in the ventral tegmental area of non-dependent TH::Cre rats. Neuropharmacology 2023; 228:109457. [PMID: 36764577 PMCID: PMC10034863 DOI: 10.1016/j.neuropharm.2023.109457] [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: 12/14/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The dynorphin (DYN)/kappa-opioid receptor (KOR) system is involved in dysphoria and negative emotional states. Dysregulation of KOR function promotes maladaptive behavioral regulation during withdrawal associated with alcohol dependence. Mesolimbic dopaminergic (DA) projections from the ventral tegmental area (VTA) innervate the extended amygdala circuitry and presynaptic KORs attenuate DA in these regions leading to an excessive alcohol consumption and negative affective-like behavior, whereas mesocortical KOR-regulated DA projections have been implicated in executive function and decision-making. Thus, the neuroadaptations occurring in DYN/KOR systems are important aspects to consider for the development of personalized therapeutic solutions. Herein, we study the contribution of the VTA DA neuron Oprk1 (KOR gene) in excessive alcohol consumption, negative emotional state, and executive function. To do so, Oprk1 mRNA expression and KOR function were characterized to confirm alcohol dependence-induced dysregulation in the VTA. Then, a transgenic Cre-Lox rat model (male and female TH::Cre rats) was used to allow for conditional and inducible overexpression of Oprk1 in VTA DA neurons. The effect of this overexpression was evaluated on operant alcohol self-administration, negative emotional states, and executive function. We found that VTA Oprk1 overexpression recapitulates some phenotypes of alcohol dependence including escalated alcohol self-administration and depressive-like behavior. However, working memory performance was not impacted following VTA Oprk1 overexpression in TH::Cre rats. This supports the hypothesis that dysregulated KOR signaling within the mesolimbic DA system is an important contributor to symptoms of alcohol dependence and shows that understanding Oprk1-mediated contributions to alcohol use disorder (AUD) should be an important future goal.
Collapse
Affiliation(s)
- Gaetan Lepreux
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Tampa, FL, USA
| | - Grace E Shinn
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Gengze Wei
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Tampa, FL, USA
| | - Azra Suko
- Department of Anesthesiology and Pain Medicine, Seattle, WA, USA
| | - George Concepcion
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Tampa, FL, USA
| | - Sunil Sirohi
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, USA
| | - Bok Soon Go
- Department of Life Sciences, Korea University, Seoul, 02841, South Korea
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, Seattle, WA, USA; Department of Pharmacology, Seattle, WA, USA; Center for the Neurobiology of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Brendan M Walker
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychiatry and Behavioral Neurosciences, Tampa, FL, USA; Department of Molecular Medicine, Tampa, FL, USA; USF Health Neuroscience Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
16
|
Chen Y, Wang CY, Zan GY, Yao SY, Deng YZ, Shu XL, Wu WW, Ma Y, Wang YJ, Yu CX, Liu JG. Upregulation of dynorphin/kappa opioid receptor system in the dorsal hippocampus contributes to morphine withdrawal-induced place aversion. Acta Pharmacol Sin 2023; 44:538-545. [PMID: 36127507 PMCID: PMC9958091 DOI: 10.1038/s41401-022-00987-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022]
Abstract
Aversive emotion of opioid withdrawal generates motivational state leading to compulsive drug seeking and taking. Kappa opioid receptor (KOR) and its endogenous ligand dynorphin have been shown to participate in the regulation of aversive emotion. In the present study, we investigated the role of dynorphin/KOR system in the aversive emotion following opioid withdrawal in acute morphine-dependent mice. We found that blockade of KORs before pairing by intracerebroventricular injection of KOR antagonist norBNI (20, 40 μg) attenuated the development of morphine withdrawal-induced conditioned place aversion (CPA) behavior. We further found that morphine withdrawal increased dynorphin A expression in the dorsal hippocampus, but not in the amygdala, prefrontal cortex, nucleus accumbens, and thalamus. Microinjection of norBNI (20 μg) into the dorsal hippocampus significantly decreased morphine withdrawal-induced CPA behavior. We further found that p38 MAPK was significantly activated in the dorsal hippocampus after morphine withdrawal, and the activation of p38 MAPK was blocked by pretreatment with norBNI. Accordingly, microinjection of p38 MAPK inhibitor SB203580 (5 μg) into the dorsal hippocampus significantly decreased morphine withdrawal-produced CPA behavior. This study demonstrates that upregulation of dynorphin/KOR system in the dorsal hippocampus plays a critical role in the formation of aversive emotion associated with morphine withdrawal, suggesting that KOR antagonists may have therapeutic value for the treatment of opioid withdrawal-induced mood-related disorders.
Collapse
Affiliation(s)
- Yan Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Chen-Yao Wang
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, Beijing, 100049, China
| | - Gui-Ying Zan
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Song-Yu Yao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ying-Zhi Deng
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Xue-Lian Shu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, Beijing, 100049, China
| | - Wei-Wei Wu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Yan Ma
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Yu-Jun Wang
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China.
| | - Chang-Xi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Jing-Gen Liu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| |
Collapse
|
17
|
Yang R, Tuan RRL, Hwang FJ, Bloodgood DW, Kong D, Ding JB. Dichotomous regulation of striatal plasticity by dynorphin. Mol Psychiatry 2023; 28:434-447. [PMID: 36460726 PMCID: PMC10188294 DOI: 10.1038/s41380-022-01885-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022]
Abstract
Modulation of corticostriatal plasticity alters the information flow throughout basal ganglia circuits and represents a fundamental mechanism for motor learning, action selection, and reward. Synaptic plasticity in the striatal direct- and indirect-pathway spiny projection neurons (dSPNs and iSPNs) is regulated by two distinct networks of GPCR signaling cascades. While it is well-known that dopamine D2 and adenosine A2a receptors bi-directionally regulate iSPN plasticity, it remains unclear how D1 signaling modulation of synaptic plasticity is counteracted by dSPN-specific Gi signaling. Here, we show that striatal dynorphin selectively suppresses long-term potentiation (LTP) through Kappa Opioid Receptor (KOR) signaling in dSPNs. Both KOR antagonism and conditional deletion of dynorphin in dSPNs enhance LTP counterbalancing with different levels of D1 receptor activation. Behaviorally, mice lacking dynorphin in D1 neurons show comparable motor behavior and reward-based learning, but enhanced flexibility during reversal learning. These findings support a model in which D1R and KOR signaling bi-directionally modulate synaptic plasticity and behavior in the direct pathway.
Collapse
Affiliation(s)
- Renzhi Yang
- Biology Graduate Program, Stanford University, Stanford, CA, USA
| | - Rupa R Lalchandani Tuan
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
| | - Fuu-Jiun Hwang
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | | | - Dong Kong
- Division of Endocrinology, Department of Pediatrics, F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Stanford Bio-X, Stanford University, Stanford, CA, USA.
| |
Collapse
|
18
|
Liu-Chen LY, Huang P. Signaling underlying kappa opioid receptor-mediated behaviors in rodents. Front Neurosci 2022; 16:964724. [PMID: 36408401 PMCID: PMC9670127 DOI: 10.3389/fnins.2022.964724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/08/2022] [Indexed: 11/06/2022] Open
Abstract
Kappa opioid receptor (KOR) agonists are potentially useful as analgesic and anti-pruritic agents, for prevention and treatment of substance use disorders, and for treatment of demyelinating diseases. However, side effects of KOR agonists, including psychotomimesis, dysphoria, and sedation, have caused early termination of clinical trials. Understanding the signaling mechanisms underlying the beneficial therapeutic effects and the adverse side effects may help in the development of KOR agonist compounds. In this review, we summarize the current knowledge in this regard in five sections. First, studies conducted on mutant mouse lines (GRK3-/-, p38alpha MAPK-/-, β-arrestin2-/-, phosphorylation-deficient KOR) are summarized. In addition, the abilities of four distinct KOR agonists, which have analgesic and anti-pruritic effects with different side effect profiles, to cause KOR phosphorylation are discussed. Second, investigations on the KOR agonist nalfurafine, both in vitro and in vivo are reviewed. Nalfurafine was the first KOR full agonist approved for clinical use and in the therapeutic dose range it did not produce significant side effects associated with typical KOR agonists. Third, large-scale high-throughput phosphoproteomic studies without a priori hypotheses are described. These studies have revealed that KOR-mediated side effects are associated with many signaling pathways. Fourth, several novel G protein-biased KOR agonists that have been characterized for in vitro biochemical properties and agonist biases and in vivo behavior effects are described. Lastly, possible mechanisms underlying KOR-mediated CPA, hypolocomotion and motor incoordination are discussed. Overall, it is agreed upon that the analgesic and anti-pruritic effects of KOR agonists are mediated via G protein signaling. However, there is no consensus on the mechanisms underlying their side effects. GRK3, p38 MAPK, β-arrestin2, mTOR pathway, CB1 cannabinoid receptor and protein kinase C have been implicated in one side effect or another. For drug discovery, after initial in vitro characterization, in vivo pharmacological characterizations in various behavior tests are still the most crucial steps and dose separation between beneficial therapeutic effects and adverse side effects are the critical determinant for the compounds to be moved forward for clinical development.
Collapse
Affiliation(s)
- Lee-Yuan Liu-Chen
- Center for Substance Abuse Research, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | | |
Collapse
|
19
|
The Opioid System in Depression. Neurosci Biobehav Rev 2022; 140:104800. [PMID: 35914624 PMCID: PMC10166717 DOI: 10.1016/j.neubiorev.2022.104800] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 12/16/2022]
Abstract
Opioid receptors are widely distributed throughout the brain and play an essential role in modulating aspects of human mood, reward, and well-being. Accumulating evidence indicates the endogenous opioid system is dysregulated in depression and that pharmacological modulators of mu, delta, and kappa opioid receptors hold potential for the treatment of depression. Here we review animal and clinical data, highlighting evidence to support: dysregulation of the opioid system in depression, evidence for opioidergic modulation of behavioural processes and brain regions associated with depression, and evidence for opioidergic modulation in antidepressant responses. We evaluate clinical trials that have examined the safety and efficacy of opioidergic agents in depression and consider how the opioid system may be involved in the effects of other treatments, including ketamine, that are currently understood to exert antidepressant effects through non-opioidergic actions. Finally, we explore key neurochemical and molecular mechanisms underlying the potential therapeutic effects of opioid system engagement, that together provides a rationale for further investigation into this relevant target in the treatment of depression.
Collapse
|
20
|
Brice-Tutt AC, Eans SO, Yakovlev D, Aldrich JV, McLaughlin JP. An analog of [d-Trp]CJ-15,208 exhibits kappa opioid receptor antagonism following oral administration and prevents stress-induced reinstatement of extinguished morphine conditioned place preference. Pharmacol Biochem Behav 2022; 217:173405. [PMID: 35584724 PMCID: PMC11891885 DOI: 10.1016/j.pbb.2022.173405] [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: 12/11/2021] [Revised: 03/26/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
Opioid use disorder (OUD) relapse rates are discouragingly high, underscoring the need for new treatment options. The macrocyclic tetrapeptide natural product CJ-15,208 and its stereoisomer [d-Trp]CJ-15,208 demonstrate kappa opioid receptor (KOR) antagonist activity upon oral administration which prevents stress-induced reinstatement of cocaine-seeking behavior. In order to further explore the structure-activity relationships and expand the potential therapeutic applications of KOR antagonism for the treatment of OUD, we screened a series of 24 analogs of [d-Trp]CJ-15,208 with the goal of enhancing KOR antagonist activity. From this screening, analog 22 arose as a compound of interest, demonstrating dose-dependent KOR antagonism after central and oral administration lasting at least 2.5 h. In further oral testing, analog 22 lacked respiratory, locomotor, or reinforcing effects, consistent with the absence of opioid agonism. Pretreatment with analog 22 (30 mg/kg, p.o.) prevented stress-induced reinstatement of extinguished morphine conditioned place preference and reduced some signs of naloxone-precipitated withdrawal in mice physically dependent on morphine. Collectively, these data support the therapeutic potential of KOR antagonists to support abstinence in OUD and ameliorate opioid withdrawal.
Collapse
Affiliation(s)
- Ariana C Brice-Tutt
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Dmitry Yakovlev
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Jane V Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America.
| |
Collapse
|
21
|
French AR, van Rijn RM. An updated assessment of the translational promise of G-protein-biased kappa opioid receptor agonists to treat pain and other indications without debilitating adverse effects. Pharmacol Res 2022; 177:106091. [PMID: 35101565 PMCID: PMC8923989 DOI: 10.1016/j.phrs.2022.106091] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 01/22/2023]
Abstract
Kappa opioid receptor (κOR) agonists lack the abuse liability and respiratory depression effects of clinically used mu opioid receptor (μOR) analgesics and are hypothesized to be safer alternatives. However, κOR agonists have limiting adverse effects of their own, including aversion, sedation, and mood effects, that have hampered their clinical translation. Studies performed over the last 15 years have suggested that these adverse effects could result from activation of distinct intracellular signaling pathways that are dependent on β-arrestin, whereas signaling downstream of G protein activation produces antinociception. This led to the hypothesis that agonists biased away from β-arrestin signaling would have improved therapeutic windows over traditional unbiased agonists and allow for clinical development of analgesic G-protein-biased κOR agonists. Given a recent controversy regarding the benefits of G-protein-biased μOR agonists, it is timely to reassess the therapeutic promise of G-protein-biased κOR agonists. Here we review recent discoveries from preclinical κOR studies and critically evaluate the therapeutic windows of G-protein-biased κOR agonists in each of the adverse effects above. Overall, we find that G-protein-biased κOR agonists generally have improved therapeutic window relative to unbiased agonists, although frequently study design limits strong conclusions in this regard. However, a steady flow of newly developed biased κOR agonists paired with recently engineered behavioral and molecular tools puts the κOR field in a prime position to make major advances in our understanding of κOR function and fulfill the promise of translating a new generation of biased κOR agonists to the clinic.
Collapse
Affiliation(s)
- Alexander R French
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
22
|
Koob GF. Anhedonia, Hyperkatifeia, and Negative Reinforcement in Substance Use Disorders. Curr Top Behav Neurosci 2022; 58:147-165. [PMID: 35112332 DOI: 10.1007/7854_2021_288] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug addiction has been defined as a chronically relapsing disorder that is characterized by a compulsion to seek and take a drug or stimulus, the loss of control in limiting intake, and the emergence of a negative emotional state when access to the drug or stimulus is prevented, a component of which is anhedonia. The present review explores a heuristic framework for understanding the role of anhedonia in addiction, in which anhedonia is a key component of hyperkatifeia (conceptualized as the potentiated intensity of negative emotional/motivational symptoms during drug withdrawal) and negative reinforcement in addiction. The neural substrates that mediate such anhedonia and crosstalk between elements of hyperkatifeia that contribute to anhedonia are then explored, including crosstalk between physical pain and emotional pain systems. The present review explores current knowledge of neurochemical neurocircuitry changes that are associated with conditioned hyperkatifeia/anhedonia. The overall hypothesis is that the shift in motivation toward negative reinforcement in addiction reflects the allostatic misregulation of hedonic tone, such that drug taking makes anhedonia worse during the process of seeking temporary relief by compulsive drug taking, thereby perpetuating the addiction cycle and hedonic comorbidities that are associated with addiction.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA. .,National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
23
|
Estave PM, Spodnick MB, Karkhanis AN. KOR Control over Addiction Processing: An Exploration of the Mesolimbic Dopamine Pathway. Handb Exp Pharmacol 2022; 271:351-377. [PMID: 33301050 PMCID: PMC8192597 DOI: 10.1007/164_2020_421] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Drug addiction is a complex, persistent, and chronically relapsing neurological disorder exacerbated by acute and chronic stress. It is well known that the dynorphin/kappa opioid receptor (KOR) system regulates stress perception and responsivity, while the mesolimbic dopamine system plays a role in reward and reinforcement associated with alcohol and substance use disorders. Interestingly, the dopamine and dynorphin/KOR systems are highly integrated in mesolimbic areas, with KOR activation leading to inhibition of dopamine release, further altering the perception of reinforcing and aversive stimuli. Chronic or repeated exposure to stress or drugs potentiates KOR function ultimately contributing to a hypodopaminergic state. This hypodopaminergic state is one of the hallmarks of hyperkatifeia, defined as the hypersensitivity to emotional distress that is exacerbated during drug withdrawal and abstinence. The relationship between stress and drug addiction is bidirectional; repeated/chronic stress promotes pro-addictive behaviors, and repeated cycles of drug exposure and withdrawal, across various drug classes, produces stress. Neuroadaptations driven by this bidirectional relationship ultimately influence the perception of the reinforcing value of rewarding stimuli. In this chapter, we address the involvement of the dopamine and dynorphin/KOR systems and their interactions in shaping reinforcement value processing after drug and stress exposure, as well as a combinatorial impact of both drugs and stress.
Collapse
Affiliation(s)
- Paige M Estave
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Mary B Spodnick
- Department of Psychology, Developmental Exposure Alcohol Research Center, Center for Developmental and Behavioral Neuroscience, Binghamton University - SUNY, Binghamton, NY, USA
| | - Anushree N Karkhanis
- Department of Psychology, Developmental Exposure Alcohol Research Center, Center for Developmental and Behavioral Neuroscience, Binghamton University - SUNY, Binghamton, NY, USA.
| |
Collapse
|
24
|
Anderson EM, Lopez MF, Kastner A, Mulholland PJ, Becker HC, Cowan CW. The histone methyltransferase G9a mediates stress-regulated alcohol drinking. Addict Biol 2022; 27:e13060. [PMID: 34013595 PMCID: PMC8602448 DOI: 10.1111/adb.13060] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/23/2021] [Accepted: 05/04/2021] [Indexed: 01/03/2023]
Abstract
The epigenetic enzyme G9a is a histone methyltransferase that dimethylates lysine 9 on histone H3 (H3K9me2), and in the adult nucleus accumbens (NAc), G9a regulates multiple behaviors associated with substance use disorder. We show here that chronic intermittent ethanol (CIE) exposure in male mice reduced both G9a and H3K9me2 levels in the adult NAc, but not dorsal striatum. Viral-mediated reduction of G9a in the NAc had no effects on baseline volitional ethanol drinking or escalated alcohol drinking produced by CIE exposure; however, NAc G9a was required for stress-regulated changes in ethanol drinking, including potentiated alcohol drinking produced by activation of the kappa-opioid receptor. In addition, we observed that chronic systemic administration of a G9a inhibitor, UNC0642, also blocked stress-potentiated alcohol drinking. Together, our findings suggest that chronic alcohol use, similar to other abused substances, produces a NAc-selective reduction in G9a levels that serves to limit stress-regulated alcohol drinking. Moreover, our findings suggest that pharmacological inhibition of G9a might provide a novel therapeutic approach to treat stress-induced alcohol drinking, which is a major trigger of relapse in individuals suffering from AUD.
Collapse
Affiliation(s)
- Ethan M. Anderson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Marcelo F. Lopez
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Abigail Kastner
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Patrick J. Mulholland
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Howard C. Becker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| | - Christopher W. Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Department of Psychiatry and Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
25
|
GSK3β Activity in Reward Circuit Functioning and Addiction. NEUROSCI 2021. [DOI: 10.3390/neurosci2040033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glycogen synthase kinase-3β (GSK3β), primarily described as a regulator of glycogen metabolism, is a molecular hub linking numerous signaling pathways and regulates many cellular processes like cytoskeletal rearrangement, cell migration, apoptosis, and proliferation. In neurons, the kinase is engaged in molecular events related to the strengthening and weakening of synapses, which is a subcellular manifestation of neuroplasticity. Dysregulation of GSK3β activity has been reported in many neuropsychiatric conditions, like schizophrenia, major depressive disorder, bipolar disorder, and Alzheimer’s disease. In this review, we describe the kinase action in reward circuit-related structures in health and disease. The effect of pharmaceuticals used in the treatment of addiction in the context of GSK3β activity is also discussed.
Collapse
|
26
|
Zan GY, Wang YJ, Li XP, Fang JF, Yao SY, Du JY, Wang Q, Sun X, Liu R, Shao XM, Long JD, Chai JR, Deng YZ, Chen YQ, Li QL, Fang JQ, Liu ZQ, Liu JG. Amygdalar κ-opioid receptor-dependent upregulating glutamate transporter 1 mediates depressive-like behaviors of opioid abstinence. Cell Rep 2021; 37:109913. [PMID: 34731618 DOI: 10.1016/j.celrep.2021.109913] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/06/2021] [Accepted: 10/09/2021] [Indexed: 11/17/2022] Open
Abstract
Opiates produce a strong rewarding effect, but abstinence from opiate use emerges with severe negative emotions. Depression is one of the most frequent emotion disorders associated with opiate abstinence, which is thought to be a main cause for relapse. However, neurobiological bases of such an aversive emotion processing are poorly understood. Here, we find that morphine abstinence activates κ-opioid receptors (KORs) by increasing endogenous KOR ligand dynorphin expression in the amygdala, which in turn facilitates glutamate transporter 1 (GLT1) expression by activation of p38 mitogen-activated protein kinase (MAPK). Upregulation of GLT1 expression contributes to opiate-abstinence-elicited depressive-like behaviors through modulating amygdalar glutamatergic inputs to the nucleus accumbens (NAc). Intra-amygdala injection of GLT1 inhibitor DHK or knockdown of GLT1 expression in the amygdala significantly suppresses morphine-abstinence-induced depressive-like behaviors. Pharmacological and pharmacogenetic activation of amygdala-NAc projections prevents morphine-abstinence-induced behaviors. Overall, our study provides key molecular and circuit insights into the mechanisms of depression associated with opiate abstinence.
Collapse
Affiliation(s)
- Gui-Ying Zan
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yu-Jun Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Xue-Ping Li
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Jun-Fan Fang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Song-Yu Yao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jun-Ying Du
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Qian Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiang Sun
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230038, China
| | - Rui Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xiao-Mei Shao
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Jian-Dong Long
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing-Rui Chai
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ying-Zhi Deng
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ye-Qing Chen
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Qing-Lin Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230038, China
| | - Jian-Qiao Fang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China.
| | - Zhi-Qiang Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Jing-Gen Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China; Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
27
|
Avila JA, Memos N, Aslan A, Andrejewski T, Luine VN, Serrano PA. Voluntary oral methamphetamine increases memory deficits and contextual sensitization during abstinence associated with decreased PKMζ and increased κOR in the hippocampus of female mice. J Psychopharmacol 2021; 35:1240-1252. [PMID: 34587831 PMCID: PMC9083019 DOI: 10.1177/02698811211048285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Female populations exhibit vulnerabilities to psychostimulant addiction, as well as cognitive dysfunction following bouts of abuse. AIMS The goal for this study was to advance our understanding of the mechanisms that produce sex disparities in drug addiction. METHODS We used an animal model for voluntary oral methamphetamine administration (VOMA) and focused on male and female mice that consumed 7.6-8.2 mg/kg of methamphetamine (MA) per day during the last 18 days of the paradigm. RESULTS The VOMA-exposed female mice displayed increased locomotor activity in the drug-administration context compared to male mice, demonstrating sex-specific changes in contextual sensitization. During 2 weeks of forced abstinence, mice underwent further behavioral testing. We show that abstinence increased open-arm entries on the elevated plus maze in both sexes. There were no differences in immobility on the tail suspension test. In a hippocampal-dependent radial arm maze task, VOMA-treated female mice, but not male mice, showed working memory deficits. Hippocampal tissue was collected and analyzed using Western blotting. VOMA-exposed female mice exhibited increased kappa opioid receptor (κOR) expression in the hippocampus compared to male mice, suggesting a vulnerability toward abstinence-induced dysphoria. Female VOMA mice also exhibited a decrease in the memory protein marker, protein kinase M zeta (PKMζ), in the hippocampus. CONCLUSIONS Our study reveals sex-specific effects following abstinence from chronic MA consumption on hippocampal κOR and PKMζ expression, suggesting that these neural changes in female mice may underlie spatial memory deficits and identify an increased susceptibility to dysregulated neural mechanisms. These data validate VOMA as a model sensitive to sex differences in behavior and hippocampal neurochemistry following chronic MA exposure.
Collapse
Affiliation(s)
- Jorge A. Avila
- Department of Psychology, Hunter College, City University of New York, New York, NY 10065, USA.,The Graduate Center of CUNY, New York, NY 10016, USA
| | - Nicoletta Memos
- Department of Psychology, Hunter College, City University of New York, New York, NY 10065, USA.,The Graduate Center of CUNY, New York, NY 10016, USA
| | - Abdurrahman Aslan
- Department of Psychology, Hunter College, City University of New York, New York, NY 10065, USA.,Department of Pharmacology, Health Sciences Center, Louisiana State University, New Orleans, LA,Department of Pharmacology, Istinye University, Istanbul, Turkey
| | - Tytus Andrejewski
- Department of Psychology, Hunter College, City University of New York, New York, NY 10065, USA
| | - Victoria N. Luine
- Department of Psychology, Hunter College, City University of New York, New York, NY 10065, USA.,The Graduate Center of CUNY, New York, NY 10016, USA
| | - Peter A. Serrano
- Department of Psychology, Hunter College, City University of New York, New York, NY 10065, USA.,The Graduate Center of CUNY, New York, NY 10016, USA
| |
Collapse
|
28
|
Yu C, Zhang T, Shi S, Wei T, Wang Q. Potential biomarkers: differentially expressed proteins of the extrinsic coagulation pathway in plasma samples from patients with depression. Bioengineered 2021; 12:6318-6331. [PMID: 34488523 PMCID: PMC8806736 DOI: 10.1080/21655979.2021.1971037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Depression is a severe disabling psychiatric illness and the pathophysiological mechanisms remain unknown. In previous work, we found the changes in extrinsic coagulation (EC) pathway proteins in depressed patients compared with healthy subjects were significant. In this study, we screened differentially expressed proteins (DEPs) in the EC pathway, and explored the molecular mechanism by constructing a protein-protein interaction (PPI) network. The DEPs of the EC pathwaywere initially screened by isobaric tags for relative and absolute quantification (iTRAQ) in plasma samples obtained from 20 depression patients and 20 healthy controls, and were then identified by Enzyme-linked immunosorbent assays (ELISAs). Ingenuity Pathway Analysis (IPA) software was used to analyse pathway. The differentially expressed genes (DEGs) were identified by analyzing the GSE98793 microarray data from the Gene Expression Omnibus database using the Significance Analysis for Microarrays (SAM, version 4.1) statistical method. Cytoscape version 3.4.0 software was used to construct and visualize PPI networks. The results show that Fibrinogen alpha chain (FGA), Fibrinogen beta chain (FGB), Fibrinogen gamma chain (FGG) and Coagulation factor VII (FVII) were screened in the EC pathway from depression patient samples. FGA, FGB, and FGG were significantly up-regulated, and FVII was down-regulated. Thirteen DEGs related to depression and EC pathways were identified from the microarray database. Among them NF-κB Inhibitor Beta (NFKBIB) and Heat shock protein family B (small) member 1 (HSPB1) were highly correlated with EC pathway. We conclude that EC pathway is associated with depression, which provided clues for the biomarker development and the pathogenesis of depression.
Collapse
Affiliation(s)
- Chunyue Yu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Teli Zhang
- Department of Pharmacy, The People's Hospital of Daqing, Daqing, China
| | - Shanshan Shi
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Taiming Wei
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Qi Wang
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| |
Collapse
|
29
|
Nguyen JD, Grant Y, Taffe MA. Paradoxical changes in brain reward status during oxycodone self-administration in a novel test of the negative reinforcement hypothesis. Br J Pharmacol 2021; 178:3797-3812. [PMID: 33948939 PMCID: PMC8387405 DOI: 10.1111/bph.15520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE The extra medical use of, and addiction to, prescription opioid analgesics is a growing health problem. To characterize how prescription opioid abuse develops, this study investigated the affective consequences of escalating prescription opioid use using intracranial self-stimulation (ICSS) reward and oxycodone intravenous self-administration (IVSA) models. EXPERIMENTAL APPROACH Male Wistar rats were given access to oxycodone IVSA (0.15 mg·kg-1 per infusion, i.v.) in short-access (ShA; 1 h) or long-access (LgA; 12 h) sessions for five sessions per week followed by intermittent 60-h discontinuations from drug access, a novel explicit test of the negative reinforcement hypothesis. Separate groups were first trained in the ICSS procedure and then in oxycodone IVSA in 11-h LgA sessions. KEY RESULTS Rats given LgA to oxycodone escalated their responding more than ShA rats, with further significant increases observed following each 60-h discontinuation. Presession brain reward thresholds increased with sequential daily LgA IVSA sessions, consistent with a growing negative affective state consequent to successive daily intoxication/abstinence cycles. A 1-h oxycodone IVSA interval was sufficient to normalize these elevated reward thresholds, as was, paradoxically, a 60-h weekend abstinence. The increase in ICSS thresholds was attenuated in a group treated with the long-acting κ-opioid antagonist norbinaltorphimine prior to IVSA training. CONCLUSION AND IMPLICATIONS Changes in brain reward function during escalation of oxycodone self-administration are driven by an interplay between κ-opioid receptor-mediated negative affective state associated with escalated oxycodone intake and dynamic restoration of brain reward status during longer periods of abstinence.
Collapse
Affiliation(s)
- Jacques D. Nguyen
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Yanabel Grant
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Michael A. Taffe
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| |
Collapse
|
30
|
Cameron CM, Nieto S, Bosler L, Wong M, Bishop I, Mooney L, Cahill CM. Mechanisms Underlying the Anti-Suicidal Treatment Potential of Buprenorphine. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2021; 1. [PMID: 35265942 PMCID: PMC8903193 DOI: 10.3389/adar.2021.10009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Death by suicide is a global epidemic with over 800 K suicidal deaths worlwide in 2012. Suicide is the 10th leading cause of death among Americans and more than 44 K people died by suicide in 2019 in the United States. Patients with chronic pain, including, but not limited to, those with substance use disorders, are particularly vulnerable. Chronic pain patients have twice the risk of death by suicide compared to those without pain, and 50% of chronic pain patients report that they have considered suicide at some point due to their pain. The kappa opioid system is implicated in negative mood states including dysphoria, depression, and anxiety, and recent evidence shows that chronic pain increases the function of this system in limbic brain regions important for affect and motivation. Additionally, dynorphin, the endogenous ligand that activates the kappa opioid receptor is increased in the caudate putamen of human suicide victims. A potential treatment for reducing suicidal ideation and suicidal attempts is buprenorphine. Buprenorphine, a partial mu opioid agonist with kappa opioid antagonist properties, reduced suicidal ideation in chronic pain patients with and without an opioid use disorder. This review will highlight the clinical and preclinical evidence to support the use of buprenorphine in mitigating pain-induced negative affective states and suicidal thoughts, where these effects are at least partially mediated via its kappa antagonist properties.
Collapse
Affiliation(s)
- Courtney M. Cameron
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven Nieto
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lucienne Bosler
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Megan Wong
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Isabel Bishop
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Larissa Mooney
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Catherine M. Cahill
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Correspondence: Catherine M. Cahill,
| |
Collapse
|
31
|
Nosova O, Bazov I, Karpyak V, Hallberg M, Bakalkin G. Epigenetic and Transcriptional Control of the Opioid Prodynorphine Gene: In-Depth Analysis in the Human Brain. Molecules 2021; 26:molecules26113458. [PMID: 34200173 PMCID: PMC8201134 DOI: 10.3390/molecules26113458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
Neuropeptides serve as neurohormones and local paracrine regulators that control neural networks regulating behavior, endocrine system and sensorimotor functions. Their expression is characterized by exceptionally restricted profiles. Circuit-specific and adaptive expression of neuropeptide genes may be defined by transcriptional and epigenetic mechanisms controlled by cell type and subtype sequence-specific transcription factors, insulators and silencers. The opioid peptide dynorphins play a critical role in neurological and psychiatric disorders, pain processing and stress, while their mutations cause profound neurodegeneration in the human brain. In this review, we focus on the prodynorphin gene as a model for the in-depth epigenetic and transcriptional analysis of expression of the neuropeptide genes. Prodynorphin studies may provide a framework for analysis of mechanisms relevant for regulation of neuropeptide genes in normal and pathological human brain.
Collapse
Affiliation(s)
- Olga Nosova
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (I.B.); (M.H.)
- Correspondence: (O.N.); (G.B.)
| | - Igor Bazov
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (I.B.); (M.H.)
| | | | - Mathias Hallberg
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (I.B.); (M.H.)
| | - Georgy Bakalkin
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (I.B.); (M.H.)
- Correspondence: (O.N.); (G.B.)
| |
Collapse
|
32
|
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
|
33
|
Wang H, Cao D, Gillespie JC, Mendez RE, Selley DE, Liu-Chen LY, Zhang Y. Exploring the putative mechanism of allosteric modulations by mixed-action kappa/mu opioid receptor bitopic modulators. Future Med Chem 2021; 13:551-573. [PMID: 33590767 PMCID: PMC8027703 DOI: 10.4155/fmc-2020-0308] [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] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/14/2021] [Indexed: 12/26/2022] Open
Abstract
The modulation and selectivity mechanisms of seven mixed-action kappa opioid receptor (KOR)/mu opioid receptor (MOR) bitopic modulators were explored. Molecular modeling results indicated that the 'message' moiety of seven bitopic modulators shared the same binding mode with the orthosteric site of the KOR and MOR, whereas the 'address' moiety bound with different subdomains of the allosteric site of the KOR and MOR. The 'address' moiety of seven bitopic modulators bound to different subdomains of the allosteric site of the KOR and MOR may exhibit distinguishable allosteric modulations to the binding affinity and/or efficacy of the 'message' moiety. Moreover, the 3-hydroxy group on the phenolic moiety of the seven bitopic modulators induced selectivity to the KOR over the MOR.
Collapse
Affiliation(s)
- Huiqun Wang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Danni Cao
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - James C Gillespie
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Rolando E Mendez
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Dana E Selley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Lee-Yuan Liu-Chen
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
34
|
The Role of Dynorphin and the Kappa Opioid Receptor in Schizophrenia and Major Depressive Disorder: A Translational Approach. Handb Exp Pharmacol 2021; 271:525-546. [PMID: 33459877 DOI: 10.1007/164_2020_396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kappa opioid receptor (KOR) and its endogenous ligands dynorphins (DYN) have been implicated in the development or symptomatology of a variety of neuropsychiatric disorders. This review covers a brief history of the development of KOR agonists and antagonists, their effects in healthy volunteers, and the potential role of DYN/KOR dysfunction in schizophrenia and major depressive disorder from a translational perspective. The potential role of DYN/KOR dysfunction in schizophrenia is based on several lines of evidence. Selective KOR agonists induce affective states in healthy volunteers with similarities to the symptoms of schizophrenia. Studies have shown increased DYN in patients with schizophrenia, although the data have been mixed. Finally, meta-analytic data have shown that opioid antagonists are associated with reductions in the symptoms of schizophrenia. The potential role of DYN/KOR dysfunction in major depressive disorder is also based on a combination of preclinical and clinical data. Selective KOR agonists have shown pro-depressive effects in human volunteers, while selective KOR antagonists have shown robust efficacy in several preclinical models of antidepressant activity. Small studies have shown that nonselective KOR antagonists may have efficacy in treatment-resistant depression. Additionally, recent clinical data have shown that the KOR may be an effective target for treating anhedonia, a finding relevant to both schizophrenia and depression. Finally, recommendations are provided for translating preclinical models for schizophrenia and major depressive disorder into the clinic.
Collapse
|
35
|
Koob GF. Drug Addiction: Hyperkatifeia/Negative Reinforcement as a Framework for Medications Development. Pharmacol Rev 2021; 73:163-201. [PMID: 33318153 PMCID: PMC7770492 DOI: 10.1124/pharmrev.120.000083] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Compulsive drug seeking that is associated with addiction is hypothesized to follow a heuristic framework that involves three stages (binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation) and three domains of dysfunction (incentive salience/pathologic habits, negative emotional states, and executive function, respectively) via changes in the basal ganglia, extended amygdala/habenula, and frontal cortex, respectively. This review focuses on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the addiction cycle. Hyperkatifeia provides an additional source of motivation for compulsive drug seeking via negative reinforcement. Negative reinforcement reflects an increase in the probability of a response to remove an aversive stimulus or drug seeking to remove hyperkatifeia that is augmented by genetic/epigenetic vulnerability, environmental trauma, and psychiatric comorbidity. Neurobiological targets for hyperkatifeia in addiction involve neurocircuitry of the extended amygdala and its connections via within-system neuroadaptations in dopamine, enkephalin/endorphin opioid peptide, and γ-aminobutyric acid/glutamate systems and between-system neuroadaptations in prostress corticotropin-releasing factor, norepinephrine, glucocorticoid, dynorphin, hypocretin, and neuroimmune systems and antistress neuropeptide Y, nociceptin, endocannabinoid, and oxytocin systems. Such neurochemical/neurocircuitry dysregulations are hypothesized to mediate a negative hedonic set point that gradually gains allostatic load and shifts from a homeostatic hedonic state to an allostatic hedonic state. Based on preclinical studies and translational studies to date, medications and behavioral therapies that reset brain stress, antistress, and emotional pain systems and return them to homeostasis would be promising new targets for medication development. SIGNIFICANCE STATEMENT: The focus of this review is on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the drug addiction cycle and a driving force for negative reinforcement in addiction. Medications and behavioral therapies that reverse hyperkatifeia by resetting brain stress, antistress, and emotional pain systems and returning them to homeostasis would be promising new targets for medication development.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
36
|
Jacobson ML, Wulf HA, Tsuda MC, Browne CA, Lucki I. Sex differences in the modulation of mouse nest building behavior by kappa opioid receptor signaling. Neuropharmacology 2020; 177:108254. [PMID: 32726598 PMCID: PMC11423493 DOI: 10.1016/j.neuropharm.2020.108254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022]
Abstract
Emerging evidence suggests that females are less sensitive than males to the effects of kappa opioid receptor (KOR) ligands across multiple behavioral measures. The effects of the KOR agonist U50,488 and the KOR antagonist aticaprant were assessed on nest building behavior, an ethologically relevant indicator of overall well-being and affect, in adult male and female C57BL/6J mice. Females required a higher dose of U50,488 to suppress nesting, and a higher dose of aticaprant to restore U50,488-induced impairment of nesting. Females also required a higher dose of aticaprant to decrease immobility scores in the forced swim test. Pretreatment with the estrogen receptor modulator tamoxifen, at a dose which blocked estrogen receptors, augmented the effect of U50,488 on nesting in female mice, suggesting that estrogen receptors play a key role in attenuating the effects of KOR ligands in female mice. Together, these results suggest that females are less sensitive to KOR mediation, requiring a higher dose to achieve comparable results to males. This behavioral sensitivity, as measured by nesting, may be mediated by estrogen receptors. Together these studies highlight the importance of comparing sex differences in response to KOR regulation on behaviors related to affective states.
Collapse
Affiliation(s)
- Moriah L Jacobson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Service University, Bethesda, MD, 20814, USA
| | - Hildegard A Wulf
- Department of Pharmacology & Molecular Therapeutics, Uniformed Service University, Bethesda, MD, 20814, USA
| | - Mumeko C Tsuda
- Department of Pharmacology & Molecular Therapeutics, Uniformed Service University, Bethesda, MD, 20814, USA; Rat Behavior Core, Uniformed Service University, Bethesda, MD, 20814, USA
| | - Caroline A Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Service University, Bethesda, MD, 20814, USA
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Service University, Bethesda, MD, 20814, USA; Department of Psychiatry, Uniformed Service University, Bethesda, MD, 20814, USA.
| |
Collapse
|
37
|
Huang P, Gentile TA, Muschamp JW, Liu-Chen LY. The kappa opioid receptor agonist U50,488H did not affect brain-stimulation reward while it elicited conditioned place aversion in mice. BMC Res Notes 2020; 13:384. [PMID: 32799930 PMCID: PMC7429879 DOI: 10.1186/s13104-020-05227-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/06/2020] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Selective kappa opioid receptor (KOR) agonists were shown to produce a dose-dependent depression of brain-stimulation reward (BSR) in the rat intracranial self-stimulation (ICSS) tests. However, limited studies using mice produced less conclusive results. Here the effects of U50,488H were re-examined on BSR in mice with a larger cohort of animals. RESULTS Forty C57BL/6J male mice were implanted with the electrodes in medial forebrain bundle. About a week after surgery, mice were subject to ICSS training. Only eighteen passed the two-phase procedures, at which point they readily spun the wheels to obtain reinforcing effect of BSR, and were used for the ICSS tests. Compared with saline (s.c.), U50,488H (2 mg/kg, s.c.) did not have effects on the BSR thresholds within 1 h post-treatment, while it decreased the maximum wheel-spinning rates in a time-dependent manner. In contrast, cocaine (5 mg/kg, s.c.) decreased the BSR thresholds time-dependently without affecting the maximum wheel-spinning rates in the same cohort of mice, demonstrating the validity of our mouse ICSS models. For comparison, U50,488H (2 mg/kg, s.c.) induced significant conditioned place aversion (CPA) in a different cohort of mice without surgeries. Thus, ICSS may not be an appropriate test for KOR agonist-induced aversion in mice.
Collapse
Affiliation(s)
- Peng Huang
- Center for Substance Abuse Research (CSAR) & Department of Pharmacology, Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, MERB 851, Philadelphia, PA, 19140, USA
| | - Taylor A Gentile
- Center for Substance Abuse Research (CSAR) & Department of Pharmacology, Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, MERB 851, Philadelphia, PA, 19140, USA
| | - John W Muschamp
- Center for Substance Abuse Research (CSAR) & Department of Pharmacology, Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, MERB 851, Philadelphia, PA, 19140, USA
| | - Lee-Yuan Liu-Chen
- Center for Substance Abuse Research (CSAR) & Department of Pharmacology, Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, MERB 851, Philadelphia, PA, 19140, USA.
| |
Collapse
|
38
|
Design, synthesis and biological evaluation of N-hydroxy-aminobenzyloxyarylamide analogues as novel selective κ opioid receptor antagonists. Bioorg Med Chem Lett 2020; 30:127236. [PMID: 32386980 DOI: 10.1016/j.bmcl.2020.127236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 01/05/2023]
Abstract
Aminobenzyloxyarylamide derivatives 1a-i and 2a-t were designed and synthesized as novel selective κ opioid receptor (KOR) antagonists. The benzoyl amide moiety of LY2456302 was changed into N-hydroxybenzamide and benzisoxazole-3(2H)-one to investigate whether it could increase the binding affinity or selectivity for KOR. All target compounds were evaluated in radioligand binding assays for opioid receptor binding affinity. These efforts led to the identification of compound 1c (κ Ki = 179.9 nM), which exhibited high affinity for KOR. Moreover, the selectivity of KOR over MOR and DOR increased nearly 2-fold and 7-fold, respectively, compared with (±)LY2456302.
Collapse
|
39
|
Kappa opioid agonists reduce oxycodone self-administration in male rhesus monkeys. Psychopharmacology (Berl) 2020; 237:1471-1480. [PMID: 32006048 PMCID: PMC7196516 DOI: 10.1007/s00213-020-05473-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
RATIONALE Combinations of mu and kappa opioid receptor (KOR) agonists have been proposed as potential analgesic formulations with reduced abuse liability. The current studies extend previous work by investigating the typical KOR agonist, salvinorin A, and the atypical KOR agonist, nalfurafine, as deterrents of oxycodone self-administration using a progressive ratio (PR) schedule of reinforcement. METHODS In separate experiments, adult male rhesus monkeys (N = 4/experiment) were trained under a PR schedule of reinforcement to self-administer cocaine (0.1 mg/kg/injection) and saline on alternating days. Oxycodone (0.01-0.1 mg/kg/injection) alone and combined with salvinorin A (experiment 1; 0.006, 0.012 mg/kg/injection) or nalfurafine (experiment 2; 0.0001-0.00032 mg/kg/injection) were tested within the alternating cocaine and saline baseline. The mechanism of nalfurafine's effects on oxycodone self-administration was investigated via pretreatment with the KOR antagonist, nor-binaltorphimine (nor-BNI; 10 mg/kg; i.m.). RESULTS All subjects self-administered oxycodone alone above saline levels at sufficiently large doses, and combining salvinorin A or nalfurafine with oxycodone reduced the mean number of injections per session to saline levels (experiment 1) or to levels that were significantly lower than oxycodone alone (experiment 2). The ability of nalfurafine to reduce oxycodone self-administration was reversed by pretreatment with nor-BNI. CONCLUSIONS These results demonstrate that KOR agonists, including the clinically used KOR agonist, nalfurafine, can punish self-administration of a prescription opioid analgesic, oxycodone, in rhesus monkeys and that nalfurafine's punishing effect is KOR-dependent. Combinations of KOR agonists with prescription opioids may have reduced abuse liability.
Collapse
|
40
|
Effects of Kappa opioid receptor blockade by LY2444296 HCl, a selective short-acting antagonist, during chronic extended access cocaine self-administration and re-exposure in rat. Psychopharmacology (Berl) 2020; 237:1147-1160. [PMID: 31915862 DOI: 10.1007/s00213-019-05444-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/27/2019] [Indexed: 12/14/2022]
Abstract
RATIONALE Cocaine addiction is a chronic brain disease characterized by compulsive drug intake and dysregulation of brain reward systems. Few preclinical studies have modeled the natural longitudinal course of cocaine addiction. Extended access self-administration protocols are powerful tools for modeling the advanced stages of addiction; however, few studies have duration of drug access longer than 12 h/session, potentially limiting their construct validity. Identification of changes in cocaine intake patterns during the development of addictive-like states may allow better treatments for vulnerable subjects. The kappa opioid receptor (KOPr) system has been implicated in the neurobiological regulation of addictive states as well as mood and stress disorders, with selective KOPr antagonists proposed as possible pharmacotherapeutic agents. Chronic cocaine exposure increases the expression of KOPr and its endogenous agonists, the dynorphins, in several brain areas in rodents. OBJECTIVES To examine the behavioral pattern of intake during chronic (14 days) 18 h intravenous cocaine self-administration (0.5 mg/kg/infusion) and the effect of a novel short-acting KOPr antagonist LY2444296 HCl (3 mg/kg) administered during sessions 8 to 14 of chronic 18 h/day cocaine self-administration and prior to a single re-exposure session after 2 cocaine-free withdrawal days. RESULTS Both daily and hourly cocaine intake patterns changed over 14 days of 18 h self-administration. LY pretreatment affected the pattern of self-administration across the second week of extended access cocaine self-administration and prevented the increase in cocaine intake during re-exposure. CONCLUSIONS Overall, the KOPr antagonist attenuated escalated cocaine consumption in a rat model of extended access cocaine self-administration.
Collapse
|
41
|
Riley AL, Nelson KH, To P, López-Arnau R, Xu P, Wang D, Wang Y, Shen HW, Kuhn DM, Angoa-Perez M, Anneken JH, Muskiewicz D, Hall FS. Abuse potential and toxicity of the synthetic cathinones (i.e., “Bath salts”). Neurosci Biobehav Rev 2020; 110:150-173. [DOI: 10.1016/j.neubiorev.2018.07.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/14/2018] [Accepted: 07/24/2018] [Indexed: 01/22/2023]
|
42
|
Jacobson ML, Browne CA, Lucki I. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Stress-Related Disorders. Annu Rev Pharmacol Toxicol 2020; 60:615-636. [DOI: 10.1146/annurev-pharmtox-010919-023317] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Exposure to stressful stimuli activates kappa opioid receptor (KOR) signaling, a process known to produce aversion and dysphoria in humans and other species. This endogenous opioid system is dysregulated in stress-related disorders, specifically in major depressive disorder (MDD). These findings serve as the foundation for a growing interest in the therapeutic potential of KOR antagonists as novel antidepressants. In this review, data supporting the hypothesis of dysregulated KOR function in MDD are considered. The clinical data demonstrating the therapeutic efficacy and safety of selective and mixed opioid antagonists are then presented. Finally, the preclinical evidence illustrating the induction of behaviors relevant to the endophenotypes of MDD and KOR antagonist activity in stress-naïve and stress-exposed animals is evaluated. Overall, this review highlights the emergent literature supporting the pursuit of KOR antagonists as novel therapeutics for MDD and other stress-related disorders.
Collapse
Affiliation(s)
- Moriah L. Jacobson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Caroline A. Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| |
Collapse
|
43
|
Puryear CB, Brooks J, Tan L, Smith K, Li Y, Cunningham J, Todtenkopf MS, Dean RL, Sanchez C. Opioid receptor modulation of neural circuits in depression: What can be learned from preclinical data? Neurosci Biobehav Rev 2020; 108:658-678. [DOI: 10.1016/j.neubiorev.2019.12.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
|
44
|
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
|
45
|
Simon MJ, Zafra MA, Puerto A. Differential rewarding effects of electrical stimulation of the lateral hypothalamus and parabrachial complex: Functional characterization and the relevance of opioid systems and dopamine. J Psychopharmacol 2019; 33:1475-1490. [PMID: 31282233 DOI: 10.1177/0269881119855982] [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: 11/17/2022]
Abstract
BACKGROUND Since the discovery of rewarding intracranial self-stimulation by Olds and Milner, extensive data have been published on the biological basis of reward. Although participation of the mesolimbic dopaminergic system is well documented, its precise role has not been fully elucidated, and some authors have proposed the involvement of other neural systems in processing specific aspects of reinforced behaviour. AIMS AND METHODS We reviewed published data, including our own findings, on the rewarding effects induced by electrical stimulation of the lateral hypothalamus (LH) and of the external lateral parabrachial area (LPBe) - a brainstem region involved in processing the rewarding properties of natural and artificial substances - and compared its functional characteristics as observed in operant and non-operant behavioural procedures. RESULTS Brain circuits involved in the induction of preferences for stimuli associated with electrical stimulation of the LBPe appear to functionally and neurochemically differ from those activated by electrical stimulation of the LH. INTERPRETATION We discuss the possible involvement of the LPBe in processing emotional-affective aspects of the brain reward system.
Collapse
Affiliation(s)
- Maria J Simon
- Department of Psychobiology, Mind, Brain and Behaviour Research Center (CIMCYC), University of Granada, Granada, Spain
| | - Maria A Zafra
- Department of Psychobiology, Mind, Brain and Behaviour Research Center (CIMCYC), University of Granada, Granada, Spain
| | - Amadeo Puerto
- Department of Psychobiology, Mind, Brain and Behaviour Research Center (CIMCYC), University of Granada, Granada, Spain
| |
Collapse
|
46
|
Page S, Mavrikaki MM, Lintz T, Puttick D, Roberts E, Rosen H, Carroll FI, Carlezon WA, Chartoff EH. Behavioral Pharmacology of Novel Kappa Opioid Receptor Antagonists in Rats. Int J Neuropsychopharmacol 2019; 22:735-745. [PMID: 31613314 PMCID: PMC7145521 DOI: 10.1093/ijnp/pyz054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/09/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND New treatments for stress-related disorders including depression, anxiety, and substance use disorder are greatly needed. Kappa opioid receptors are expressed in the central nervous system, including areas implicated in analgesia and affective state. Although kappa opioid receptor agonists share the antinociceptive effects of mu opioid receptor agonists, they also tend to produce negative affective states. In contrast, selective kappa opioid receptor antagonists have antidepressant- and anxiolytic-like effects, stimulating interest in their therapeutic potential. The prototypical kappa opioid receptor antagonists (e.g., norBNI, JDTic) have an exceptionally long duration of action that complicates their use in humans, particularly in tests to establish safety. This study was designed to test dose- and time-course effects of novel kappa opioid receptor antagonists with the goal of identifying short-acting lead compounds for future medication development. METHODS We screened 2 novel, highly selective kappa opioid receptor antagonists (CYM-52220 and CYM-52288) with oral efficacy in the warm water tail flick assay in rats to determine initial dose and time course effects. For comparison, we tested existing kappa opioid receptor antagonists JDTic and LY-2456302 (also known as CERC-501 or JNJ-67953964). RESULTS In the tail flick assay, the rank order of duration of action for the antagonists was LY-2456302 < CYM-52288 < CYM-52220 << JDTic. Furthermore, LY-2456302 blocked the depressive (anhedonia-producing) effects of the kappa opioid receptor agonist U50,488 in the intracranial self-stimulation paradigm, albeit at a higher dose than that needed for analgesic blockade in the tail flick assay. CONCLUSIONS These results suggest that structurally diverse kappa opioid receptor antagonists can have short-acting effects and that LY-2456302 reduces anhedonia as measured in the intracranial self-stimulation test.
Collapse
Affiliation(s)
- Sarah Page
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA
| | - Maria M Mavrikaki
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA
| | - Tania Lintz
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA
| | - Daniel Puttick
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA
| | - Edward Roberts
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Hugh Rosen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - F Ivy Carroll
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA
| | - Elena H Chartoff
- Research Triangle Institute, Research Triangle Park, NC,Correspondence: Dr E. H. Chartoff, PhD, McLean Hospital, MRC 218, 115 Mill Street, Belmont, MA 02478 ()
| |
Collapse
|
47
|
Emery MA, Eitan S. Members of the same pharmacological family are not alike: Different opioids, different consequences, hope for the opioid crisis? Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:428-449. [PMID: 30790677 DOI: 10.1016/j.pnpbp.2019.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/14/2023]
Abstract
Pain management is the specialized medical practice of modulating pain perception and thus easing the suffering and improving the life quality of individuals suffering from painful conditions. Since this requires the modulation of the activity of endogenous systems involved in pain perception, and given the large role that the opioidergic system plays in pain perception, opioids are currently the most effective pain treatment available and are likely to remain relevant for the foreseeable future. This contributes to the rise in opioid use, misuse, and overdose death, which is currently characterized by public health officials in the United States as an epidemic. Historically, the majority of preclinical rodent studies were focused on morphine. This has resulted in our understanding of opioids in general being highly biased by our knowledge of morphine specifically. However, recent in vitro studies suggest that direct extrapolation of research findings from morphine to other opioids is likely to be flawed. Notably, these studies suggest that different opioid analgesics (opioid agonists) engage different downstream signaling effects within the cell, despite binding to and activating the same receptors. This recognition implies that, in contrast to the historical status quo, different opioids cannot be made equivalent by merely dose adjustment. Notably, even at equianalgesic doses, different opioids could result in different beneficial and risk outcomes. In order to foster further translational research regarding drug-specific differences among opioids, here we review basic research elucidating differences among opioids in pharmacokinetics, pharmacodynamics, their capacity for second messenger pathway activation, and their interactions with the immune system and the dopamine D2 receptors.
Collapse
Affiliation(s)
- Michael A Emery
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA.
| |
Collapse
|
48
|
Dynorphin/kappa-opioid receptor control of dopamine dynamics: Implications for negative affective states and psychiatric disorders. Brain Res 2019; 1713:91-101. [DOI: 10.1016/j.brainres.2018.09.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/12/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023]
|
49
|
Sakloth F, Leggett E, Moerke MJ, Townsend EA, Banks ML, Negus SS. Effects of acute and repeated treatment with serotonin 5-HT2A receptor agonist hallucinogens on intracranial self-stimulation in rats. Exp Clin Psychopharmacol 2019; 27:215-226. [PMID: 30628811 PMCID: PMC6690189 DOI: 10.1037/pha0000253] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prototype 5-HT2A receptor agonist hallucinogens LSD, mescaline, and psilocybin are classified as Schedule 1 drugs of abuse by the U.S. Drug Enforcement Administration. Accumulating clinical evidence has also suggested that acute or repeated "microdosing" with these drugs may have utility for treatment of some mental health disorders, including drug abuse and depression. The goal of the present study was to evaluate LSD, mescaline, and psilocybin effects on intracranial self-stimulation (ICSS), a procedure that has been used to evaluate abuse-related effects of other classes of abused drugs. Effects of repeated LSD were also examined to evaluate potential changes in its own effects on ICSS or changes in effects produced by the abused psychostimulant methamphetamine or the prodepressant kappa opioid receptor (KOR) agonist U69,593. Male Sprague-Dawley rats were implanted with microelectrodes targeting the medial forebrain bundle and trained to respond under a "frequency-rate" ICSS procedure, in which many drugs of abuse increase (or "facilitate") ICSS. In acute dose-effect and time-course studies, evidence for abuse-related ICSS facilitation was weak and inconsistent; the predominant effect of all 3 drugs was dose- and time-dependent ICSS depression. Repeated LSD treatment failed to alter either its own ICSS depressant effects or the abuse-related effects of methamphetamine; however, repeated LSD did attenuate ICSS depression by U69,593. These results extend those of previous preclinical studies to suggest weak expression of abuse-related effects by 5-HT2A agonist hallucinogens and provide supportive evidence for therapeutic effects of repeated LSD dosing to attenuate KOR-mediated depressant effects but not abuse potential of psychostimulants. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
|
50
|
Phosphoproteomic approach for agonist-specific signaling in mouse brains: mTOR pathway is involved in κ opioid aversion. Neuropsychopharmacology 2019; 44:939-949. [PMID: 30082888 PMCID: PMC6462019 DOI: 10.1038/s41386-018-0155-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/05/2023]
Abstract
Kappa opioid receptor (KOR) agonists produce analgesic and anti-pruritic effects, but their clinical application was limited by dysphoria and hallucinations. Nalfurafine, a clinically used KOR agonist, does not cause dysphoria or hallucinations at therapeutic doses in humans. We found that in CD-1 mice nalfurafine produced analgesic and anti-scratch effects dose-dependently, like the prototypic KOR agonist U50,488H. In contrast, unlike U50,488H, nalfurafine caused no aversion, anhedonia, or sedation or and a low level of motor incoordination at the effective analgesia and anti-scratch doses. Thus, we established a mouse model that recapitulated important aspects of the clinical observations. We then employed a phosphoproteomics approach to investigate mechanisms underlying differential KOR-mediated effects. A large-scale mass spectrometry (MS)-based analysis on brains revealed that nalfurafine perturbed phosphoproteomes differently from U50,488H in a brain-region specific manner after 30-min treatment. In particular, U50,488H and nalfurafine imparted phosphorylation changes to proteins found in different cellular components or signaling pathways in different brain regions. Notably, we observed that U50,488H, but not nalfurafine, activated the mammalian target of rapamycin (mTOR) pathway in the striatum and cortex. Inhibition of the mTOR pathway by rapamycin abolished U50,488H-induced aversion, without affecting analgesic, anti-scratch, and sedative effects and motor incoordination. The results indicate that the mTOR pathway is involved in KOR agonist-induced aversion. This is the first demonstration that phosphoproteomics can be applied to agonist-specific signaling of G protein-coupled receptors (GPCRs) in mouse brains to unravel pharmacologically important pathways. Furthermore, this is one of the first two reports that the mTOR pathway mediates aversion caused by KOR activation.
Collapse
|