1
|
Flores-Ramirez FJ, Illenberger JM, Martin-Fardon R. Interaction between corticotropin-releasing factor, orexin, and dynorphin in the infralimbic cortex may mediate exacerbated alcohol-seeking behavior. Neurobiol Stress 2024; 33:100695. [PMID: 39640001 PMCID: PMC11617300 DOI: 10.1016/j.ynstr.2024.100695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
A major challenge for the treatment of alcohol use disorder (AUD) is relapse to alcohol use, even after protracted periods of self-imposed abstinence. Stress significantly contributes to the chronic relapsing nature of AUD, given its long-lasting ability to elicit intense craving and precipitate relapse. As individuals transition to alcohol dependence, compensatory allostatic mechanisms result in insults to hypothalamic-pituitary-adrenal axis function, mediated by corticotropin-releasing factor (CRF), which is subsequently hypothesized to alter brain reward pathways, influence affect, elicit craving, and ultimately perpetuate problematic drinking and relapse vulnerability. Orexin (OX; also called hypocretin) plays a well-established role in regulating diverse physiological processes, including stress, and has been shown to interact with CRF. Interestingly, most hypothalamic cells that express Ox mRNA also express Pdyn mRNA. Both dynorphin and OX are located in the same synaptic vesicles, and they are co-released. The infralimbic cortex (IL) of the medial prefrontal cortex (mPFC) has emerged as being directly involved in the compulsive nature of alcohol consumption during dependence. The IL is a CRF-rich region that receives OX projections from the hypothalamus and where OX receptor mRNA has been detected. Although not thoroughly understood, anatomical and behavioral pharmacology data suggest that CRF, OX, and dynorphin may interact, particularly in the IL, and that functional interactions between these three systems in the IL may be critical for the etiology and pervasiveness of compulsive alcohol seeking in dependent subjects that may render them vulnerable to relapse. The present review presents evidence of the role of the IL in AUD and discusses functional interactions between CRF, OX, and dynorphin in this structure and how they are related to exacerbated alcohol drinking and seeking.
Collapse
Affiliation(s)
- Francisco J. Flores-Ramirez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Psychology, California State University, San Marcos, CA, USA
| | | | - Rémi Martin-Fardon
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
2
|
Heesbeen EJ, van Kampen T, Verdouw PM, van Lissa C, Bijlsma EY, Groenink L. The effect of SSRIs on unconditioned anxiety: a systematic review and meta-analysis of animal studies. Psychopharmacology (Berl) 2024; 241:1731-1755. [PMID: 38980348 PMCID: PMC11339141 DOI: 10.1007/s00213-024-06645-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
RATIONALE Selective serotonin reuptake inhibitors (SSRIs) are the first choice of treatment for anxiety-like disorders. However, which aspects of anxiety are affected by SSRIs is not yet fully understood. OBJECTIVE We aimed to systematically review the effect of six clinically effective SSRIs on four aspects of unconditioned anxiety: approach-avoidance behaviour (elevated plus maze), repetitive behaviour (marble burying), distress behaviour (ultrasonic vocalization), and activation of the autonomous nervous system (stress-induced hyperthermia). METHODS We identified publications by searching Medline and Embase databases and assessed the risk of bias. A random effects meta-analysis was performed and moderator effects were analysed with Bayesian penalized meta-regression. RESULTS Our search yielded 105 elevated plus maze, 63 marble burying, 11 ultrasonic vocalization, and 7 stress-induced hyperthermia articles. Meta-analysis suggested that SSRIs reduce anxiety-like behaviour in the elevated plus maze, marble burying and ultrasonic vocalization test and that effects are moderated by pre-existing stress conditions (elevated plus maze) and dose dependency (marble burying) but not by duration of treatment or type of SSRI. The reporting quality was low, publication bias was likely, and heterogeneity was high. CONCLUSION SSRIs seem to reduce a broad range of unconditioned anxiety-associated behaviours. These results should be interpreted with caution due to a high risk of bias, likely occurrence of publication bias, substantial heterogeneity and limited moderator data availability. Our review demonstrates the importance of including bias assessments when interpreting meta-analysis results. We further recommend improving the reporting quality, the conduct of animal research, and the publication of all results regardless of significance.
Collapse
Affiliation(s)
- Elise J Heesbeen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Tatum van Kampen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - P Monika Verdouw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Caspar van Lissa
- Department of Methodology, Tilburg University, Tilburg, The Netherlands
| | - Elisabeth Y Bijlsma
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lucianne Groenink
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Ujcikova H, Lee YS, Roubalova L, Svoboda P. The impact of multifunctional enkephalin analogs and morphine on the protein changes in crude membrane fractions isolated from the rat brain cortex and hippocampus. Peptides 2024; 174:171165. [PMID: 38307418 DOI: 10.1016/j.peptides.2024.171165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Endogenous opioid peptides serve as potent analgesics through the opioid receptor (OR) activation. However, they often suffer from poor metabolic stability, low lipophilicity, and low blood-brain barrier permeability. Researchers have developed many strategies to overcome the drawbacks of current pain medications and unwanted biological effects produced by the interaction with opioid receptors. Here, we tested multifunctional enkephalin analogs LYS739 (MOR/DOR agonist and KOR partial antagonist) and LYS744 (MOR/DOR agonist and KOR full antagonist) under in vivo conditions in comparison with MOR agonist, morphine. We applied 2D electrophoretic resolution to investigate differences in proteome profiles of crude membrane (CM) fractions isolated from the rat brain cortex and hippocampus exposed to the drugs (10 mg/kg, seven days). Our results have shown that treatment with analog LYS739 induced the most protein changes in cortical and hippocampal samples. The identified proteins were mainly associated with energy metabolism, cell shape and movement, apoptosis, protein folding, regulation of redox homeostasis, and signal transduction. Among these, the isoform of mitochondrial ATP synthase subunit beta (ATP5F1B) was the only protein upregulation in the hippocampus but not in the brain cortex. Contrarily, the administration of analog LYS744 caused a small number of protein alterations in both brain parts. Our results indicate that the KOR full antagonism, together with MOR/DOR agonism of multifunctional opioid ligands, can be beneficial in treating chronic pain states by reducing changes in protein expression levels but retaining analgesic efficacy.
Collapse
Affiliation(s)
- Hana Ujcikova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic.
| | - Yeon Sun Lee
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA
| | - Lenka Roubalova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Petr Svoboda
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| |
Collapse
|
4
|
Nisbett KE, Vendruscolo LF, Koob GF. µ-Opioid receptor antagonism facilitates the anxiolytic-like effect of oxytocin in mice. Transl Psychiatry 2024; 14:125. [PMID: 38413576 PMCID: PMC10899625 DOI: 10.1038/s41398-024-02830-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/29/2024] Open
Abstract
Mood and anxiety disorders are leading causes of disability worldwide and are major contributors to the global burden of diseases. Neuropeptides, such as oxytocin and opioid peptides, are important for emotion regulation. Previous studies have demonstrated that oxytocin reduced depression- and anxiety-like behavior in male and female mice, and opioid receptor activation reduced depression-like behavior. However, it remains unclear whether the endogenous opioid system interacts with the oxytocin system to facilitate emotion regulation in male and female mice. We hypothesized that opioid receptor blockade would inhibit the anxiolytic- and antidepressant-like effects of oxytocin. In this study, we systemically administered naloxone, a preferential μ-opioid receptor antagonist, and then intracerebroventricularly administered oxytocin. We then tested mice on the elevated zero maze and the tail suspension tests, respective tests of anxiety- and depression-like behavior. Contrary to our initial hypothesis, naloxone potentiated the anxiolytic-like, but not the antidepressant-like, effect of oxytocin. Using a selective μ-opioid receptor antagonist, D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2, and a selective κ-opioid receptor antagonist, norbinaltorphimine, we demonstrate that μ-opioid receptor blockade potentiated the anxiolytic-like effect of oxytocin, whereas κ-opioid receptor blockade inhibited the oxytocin-induced anxiolytic-like effects. The present results suggest that endogenous opioids can regulate the oxytocin system to modulate anxiety-like behavior. Potential clinical implications of these findings are discussed.
Collapse
Affiliation(s)
- Khalin E Nisbett
- Graduate Program in Neuroscience, Graduate College, University of Illinois Chicago, Chicago, IL, 60607, USA.
- Stress & Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, 21224, USA.
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Leandro F Vendruscolo
- Stress & Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, 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, Baltimore, MD, 21224, USA
| |
Collapse
|
5
|
Todorov P, Assenov B, Angelov D, Dzhambazova E, Pechlivanova D. Behavioral Effects and Analgesic Profile of Hemoglobin-Derived Valorphin and Its Synthetic Analog in Rodents. Biomedicines 2023; 11:2783. [PMID: 37893157 PMCID: PMC10603931 DOI: 10.3390/biomedicines11102783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Valorphin (V1) is a naturally occurring peptide derived from hemoglobin that has been found to have an affinity for opioid receptors and exhibits antinociceptive and anticonvulsant activity. Some of its synthetic analogs containing an aminophosphonate moiety show structure-dependent potent antinociceptive effects. This study aimed to reveal a detailed picture of the antinociceptive mechanisms and behavioral effects of V1 and its recently synthesized phosphopeptide analog V2p in rodents using a range of methods. The studied peptides significantly reduced acute (mean V1-9.0, V2p-5.8 vs. controls-54.1 s) and inflammatory (mean V1-57.9 and V2p-53.3 vs. controls-107.6 s) nociceptive pain in the formalin test, as well as carrageenan-induced hyperalgesia (mean V1-184.7 and V2p-107.3 vs. controls-61.8 g) in the paw pressure test. These effects are mediated by activation of opioid receptors with a predominance of kappa in V1 antinociception and by delta, kappa, and mu receptors in V2p-induced antinociception. Both peptides did not change the levels of TNF-alpha and IL-1-beta in blood serum. V1 induces depression-like behavior, and V2p shows a tendency toward anxiolysis and short-term impairment of motor coordination without affecting exploratory behavior. The results characterize valorphin and its derivative as promising analgesics that exert their effects both centrally and peripherally, without causing severe behavioral changes in experimental animals. These encouraging data are a foundation for future studies focusing on the effects of hemorphins after long-term treatment.
Collapse
Affiliation(s)
- Petar Todorov
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 1756 Sofia, Bulgaria;
| | - Borislav Assenov
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 23, 1113 Sofia, Bulgaria; (B.A.); (D.A.)
- Faculty of Medicine, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria;
| | - Dimo Angelov
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 23, 1113 Sofia, Bulgaria; (B.A.); (D.A.)
| | - Elena Dzhambazova
- Faculty of Medicine, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria;
| | - Daniela Pechlivanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 23, 1113 Sofia, Bulgaria; (B.A.); (D.A.)
- Faculty of Medicine, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria;
| |
Collapse
|
6
|
Vázquez-León P, Miranda-Páez A, Sánchez-Castillo H, Marichal-Cancino BA. Pharmacologic hyperreactivity of kappa opioid receptors in periaqueductal gray matter during alcohol withdrawal syndrome in rats. Pharmacol Rep 2023; 75:1299-1308. [PMID: 37658980 DOI: 10.1007/s43440-023-00522-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND Periaqueductal gray matter (PAG) is a brain region rich in kappa-opioid receptors (KOR). KOR in PAG mediates behavioral responses related to pain integration, and panic response, among others. Its participation in the addiction phenomena has been poorly studied. Hence, this preliminary study explored the pharmacological effects of KOR stimulation/blockade in dorsal-PAG (D-PAG) during alcohol withdrawal on anxiety-type behaviors and alcohol intake/preference. METHODS Juvenile male Wistar rats were unexposed (A-naïve group) or exposed to alcohol for 5 weeks and then restricted (A-withdrawal group). Posteriorly, animals received intra D-PAG injections of vehicle (10% DMSO), salvinorin A (SAL-A; a selective KOR agonist), or 2-Methyl-N-((2'-(pyrrolidin-1-ylsulfonyl)biphenyl-4-yl)methyl)propan-1-amine (PF-04455242; a highly selective KOR-antagonist). Subsequently, the defensive burying behavior (DBB) and alcohol intake/preference paradigms were evaluated. RESULTS SAL-A markedly increased burying time, the height of bedding, and alcohol consumption/preference in A-withdrawal, while slightly increased the height of bedding in A-näive rats. PF-04455242 decreased both burying and immobility duration, whereas increases latency to burying, frequency of rearing, and the number of stretches attempts with no action on alcohol intake/preference in A-withdrawal rats. CONCLUSIONS In general, stimulation/blockade of KOR in A-withdrawal animals exert higher responses compared to A-naïve ones. SAL-A produced anxiety-like behaviors and increased alcohol consumption/preference, especially/solely in the alcohol-withdrawal condition, while PF-04455242 augmented exploration with no effects on alcohol intake/preference. Our findings suggest a possible pharmacologic hyperreactivity of the KOR in PAG during alcohol withdrawal.
Collapse
Affiliation(s)
- Priscila Vázquez-León
- Departamento de Fisiología Y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, 20131, Aguascalientes, Ags, Mexico
- Laboratorio de Neuropsicofarmacología, Facultad de Psicología, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Abraham Miranda-Páez
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, 07738, Mexico City, Mexico
| | - Hugo Sánchez-Castillo
- Laboratorio de Neuropsicofarmacología, Facultad de Psicología, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Bruno A Marichal-Cancino
- Departamento de Fisiología Y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, 20131, Aguascalientes, Ags, Mexico.
| |
Collapse
|
7
|
Salas-Estrada L, Provasi D, Qiu X, Kaniskan HÜ, Huang XP, DiBerto JF, Lamim Ribeiro JM, Jin J, Roth BL, Filizola M. De Novo Design of κ-Opioid Receptor Antagonists Using a Generative Deep-Learning Framework. J Chem Inf Model 2023; 63:5056-5065. [PMID: 37555591 PMCID: PMC10466374 DOI: 10.1021/acs.jcim.3c00651] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Indexed: 08/10/2023]
Abstract
Likely effective pharmacological interventions for the treatment of opioid addiction include attempts to attenuate brain reward deficits during periods of abstinence. Pharmacological blockade of the κ-opioid receptor (KOR) has been shown to abolish brain reward deficits in rodents during withdrawal, as well as to reduce the escalation of opioid use in rats with extended access to opioids. Although KOR antagonists represent promising candidates for the treatment of opioid addiction, very few potent selective KOR antagonists are known to date and most of them exhibit significant safety concerns. Here, we used a generative deep-learning framework for the de novo design of chemotypes with putative KOR antagonistic activity. Molecules generated by models trained with this framework were prioritized for chemical synthesis based on their predicted optimal interactions with the receptor. Our models and proposed training protocol were experimentally validated by binding and functional assays.
Collapse
Affiliation(s)
- Leslie Salas-Estrada
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Davide Provasi
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Xing Qiu
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Husnu Ümit Kaniskan
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Xi-Ping Huang
- National
Institute of Mental Health, Psychoactive Drug Screening Program, Department
of Pharmacology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Jeffrey F. DiBerto
- National
Institute of Mental Health, Psychoactive Drug Screening Program, Department
of Pharmacology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - João Marcelo Lamim Ribeiro
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jian Jin
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
- Mount
Sinai Center for Therapeutics Discovery, Departments of Oncological
Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Bryan L. Roth
- National
Institute of Mental Health, Psychoactive Drug Screening Program, Department
of Pharmacology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
- Division
of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Marta Filizola
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
8
|
Farahbakhsh ZZ, Song K, Branthwaite HE, Erickson KR, Mukerjee S, Nolan SO, Siciliano CA. Systemic kappa opioid receptor antagonism accelerates reinforcement learning via augmentation of novelty processing in male mice. Neuropsychopharmacology 2023; 48:857-868. [PMID: 36804487 PMCID: PMC10156709 DOI: 10.1038/s41386-023-01547-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/20/2023] [Accepted: 02/02/2023] [Indexed: 02/19/2023]
Abstract
Selective inhibition of kappa opioid receptors (KORs) is highly anticipated as a pharmacotherapeutic intervention for substance use disorders and depression. The accepted explanation for KOR antagonist-induced amelioration of aberrant behaviors posits that KORs globally function as a negative valence system; antagonism thereby blunts the behavioral influence of negative internal states such as anhedonia and negative affect. While effects of systemic KOR manipulations have been widely reproduced, explicit evaluation of negative valence as an explanatory construct is lacking. Here, we tested a series of falsifiable hypotheses generated a priori based on the negative valence model by pairing reinforcement learning tasks with systemic pharmacological KOR blockade in male C57BL/6J mice. The negative valence model failed to predict multiple experimental outcomes: KOR blockade accelerated contingency learning during both positive and negative reinforcement without altering innate responses to appetitive or aversive stimuli. We next proposed novelty processing, which influences learning independent of valence, as an alternative explanatory construct. Hypotheses based on novelty processing predicted subsequent observations: KOR blockade increased exploration of a novel, but not habituated, environment and augmented the reinforcing efficacy of novel visual stimuli in a sensory reinforcement task. Together, these results revise and extend long-standing theories of KOR system function.
Collapse
Affiliation(s)
- Zahra Z Farahbakhsh
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Keaton Song
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Hannah E Branthwaite
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kirsty R Erickson
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Snigdha Mukerjee
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Suzanne O Nolan
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
9
|
Salas-Estrada L, Provasi D, Qui X, Kaniskan HÜ, Huang XP, DiBerto JF, Ribeiro JML, Jin J, Roth BL, Filizola M. De Novo Design of κ-Opioid Receptor Antagonists Using a Generative Deep Learning Framework. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.537995. [PMID: 37162828 PMCID: PMC10168226 DOI: 10.1101/2023.04.25.537995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Likely effective pharmacological interventions for the treatment of opioid addiction include attempts to attenuate brain reward deficits during periods of abstinence. Pharmacological blockade of the κ-opioid receptor (KOR) has been shown to abolish brain reward deficits in rodents during withdrawal, as well as to reduce the escalation of opioid use in rats with extended access to opioids. Although KOR antagonists represent promising candidates for the treatment of opioid addiction, very few potent selective KOR antagonists are known to date and most of them exhibit significant safety concerns. Here, we used a generative deep learning framework for the de novo design of chemotypes with putative KOR antagonistic activity. Molecules generated by models trained with this framework were prioritized for chemical synthesis based on their predicted optimal interactions with the receptor. Our models and proposed training protocol were experimentally validated by binding and functional assays.
Collapse
|
10
|
Zhou S, Yin Y, Sheets PL. Mouse models of surgical and neuropathic pain produce distinct functional alterations to prodynorphin expressing neurons in the prelimbic cortex. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 13:100121. [PMID: 36864928 PMCID: PMC9971546 DOI: 10.1016/j.ynpai.2023.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
The medial prefrontal cortex (mPFC) consists of a heterogeneous population of neurons that respond to painful stimuli, and our understanding of how different pain models alter these specific mPFC cell types remains incomplete. A distinct subpopulation of mPFC neurons express prodynorphin (Pdyn+), the endogenous peptide agonist for kappa opioid receptors (KORs). Here, we used whole cell patch clamp for studying excitability changes to Pdyn expressing neurons in the prelimbic region of the mPFC (PLPdyn+ neurons) in mouse models of surgical and neuropathic pain. Our recordings revealed that PLPdyn+ neurons consist of both pyramidal and inhibitory cell types. We find that the plantar incision model (PIM) of surgical pain increases intrinsic excitability only in pyramidal PLPdyn+ neurons one day after incision. Following recovery from incision, excitability of pyramidal PLPdyn+ neurons did not differ between male PIM and sham mice, but was decreased in PIM female mice. Moreover, the excitability of inhibitory PLPdyn+ neurons was increased in male PIM mice, but was with no difference between female sham and PIM mice. In the spared nerve injury model (SNI), pyramidal PLPdyn+ neurons were hyperexcitable at both 3 days and 14 days after SNI. However, inhibitory PLPdyn+ neurons were hypoexcitable at 3 days but hyperexcitable at 14 days after SNI. Our findings suggest different subtypes of PLPdyn+ neurons manifest distinct alterations in the development of different pain modalities and are regulated by surgical pain in a sex-specific manner. Our study provides information on a specific neuronal population that is affected by surgical and neuropathic pain.
Collapse
Affiliation(s)
- Shudi Zhou
- Medical Neurosciences Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuexi Yin
- Medical Neurosciences Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Patrick L. Sheets
- Medical Neurosciences Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Corresponding author at: Indiana University School of Medicine, Neuroscience Research Building 400 D, 320 West 15th St, Indianapolis, IN 46202, USA.
| |
Collapse
|
11
|
Gutierrez A, Harvey EL, Creehan KM, Taffe MA. The long-term effects of repeated heroin vapor inhalation during adolescence on measures of nociception and anxiety-like behavior in adult Wistar rats. Psychopharmacology (Berl) 2022; 239:3939-3952. [PMID: 36287213 PMCID: PMC9672020 DOI: 10.1007/s00213-022-06267-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/17/2022] [Indexed: 11/25/2022]
Abstract
RATIONALE Adolescents represent a vulnerable group due to increased experimentation with illicit substances that is often associated with the adolescent period, and because adolescent drug use can result in long-term effects that differ from those caused by drug use initiated during adulthood. OBJECTIVES The purpose of the present study was to determine the effects of repeated heroin vapor inhalation during adolescence on measures of nociception, and anxiety-like behavior during adulthood in female and male Wistar rats. METHODS Rats were exposed twice daily to 30 min of heroin vapor from post-natal day (PND) 36 to PND 45. At 12 weeks of age, baseline thermal nociception was assessed across a range of temperatures with a warm-water tail-withdrawal assay. Anxiety-like behavior was assessed in an elevated plus-maze (EPM) and activity was measured in an open-field arena. Starting at 23 weeks of age, baseline thermal nociception was re-assessed, nociception was determined after acute heroin or naloxone injection, and anxiety-like behavior was redetermined in the EPM. RESULTS Adolescent heroin inhalation altered baseline thermal nociception in female rats at 12 weeks of age and in both female and male rats at ~ 23 weeks. Heroin-treated animals exhibited anxiety-like behavior when tested in the elevated plus-maze, showed blunted heroin-induced analgesia, but exhibited no effect on naloxone-induced hyperalgesia. CONCLUSIONS The present study demonstrates that heroin vapor inhalation during adolescence produces behavioral and physiological consequences in rats that persist well into adulthood.
Collapse
Affiliation(s)
- Arnold Gutierrez
- Department of Psychiatry, University of California, San Diego, Mail Code 0714, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Eric L Harvey
- Department of Psychiatry, University of California, San Diego, Mail Code 0714, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Kevin M Creehan
- Department of Psychiatry, University of California, San Diego, Mail Code 0714, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Michael A Taffe
- Department of Psychiatry, University of California, San Diego, Mail Code 0714, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
12
|
Khan MIH, Sawyer BJ, Akins NS, Le HV. A systematic review on the kappa opioid receptor and its ligands: New directions for the treatment of pain, anxiety, depression, and drug abuse. Eur J Med Chem 2022; 243:114785. [PMID: 36179400 DOI: 10.1016/j.ejmech.2022.114785] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022]
Abstract
Kappa opioid receptor (KOR) is a member of the opioid receptor system, the G protein-coupled receptors that are expressed throughout the peripheral and central nervous systems and play crucial roles in the modulation of antinociception and a variety of behavioral states like anxiety, depression, and drug abuse. KOR agonists are known to produce potent analgesic effects and have been used clinically for the treatment of pain, while KOR antagonists have shown efficacy in the treatment of anxiety and depression. This review summarizes the history, design strategy, discovery, and development of KOR ligands. KOR agonists are classified as non-biased, G protein-biased, and β-arrestin recruitment-biased, according to their degrees of bias. The mechanisms and associated effects of the G protein signaling pathway and β-arrestin recruitment signaling pathway are also discussed. Meanwhile, KOR antagonists are classified as long-acting and short-acting, based on their half-lives. In addition, we have special sections for mixed KOR agonists and selective peripheral KOR agonists. The mechanisms of action and pharmacokinetic, pharmacodynamic, and behavioral studies for each of these categories are also discussed in this review.
Collapse
Affiliation(s)
- Md Imdadul H Khan
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Benjamin J Sawyer
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Nicholas S Akins
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Hoang V Le
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
13
|
Dysfunctional Heteroreceptor Complexes as Novel Targets for the Treatment of Major Depressive and Anxiety Disorders. Cells 2022; 11:cells11111826. [PMID: 35681521 PMCID: PMC9180493 DOI: 10.3390/cells11111826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Among mental diseases, major depressive disorder (MDD) and anxiety deserve a special place due to their high prevalence and their negative impact both on society and patients suffering from these disorders. Consequently, the development of novel strategies designed to treat them quickly and efficiently, without or at least having limited side effects, is considered a highly important goal. Growing evidence indicates that emerging properties are developed on recognition, trafficking, and signaling of G-protein coupled receptors (GPCRs) upon their heteromerization with other types of GPCRs, receptor tyrosine kinases, and ionotropic receptors such as N-methyl-D-aspartate (NMDA) receptors. Therefore, to develop new treatments for MDD and anxiety, it will be important to identify the most vulnerable heteroreceptor complexes involved in MDD and anxiety. This review focuses on how GPCRs, especially serotonin, dopamine, galanin, and opioid heteroreceptor complexes, modulate synaptic and volume transmission in the limbic networks of the brain. We attempt to provide information showing how these emerging concepts can contribute to finding new ways to treat both MDD and anxiety disorders.
Collapse
|
14
|
Yakhnitsa V, Ji G, Hein M, Presto P, Griffin Z, Ponomareva O, Navratilova E, Porreca F, Neugebauer V. Kappa Opioid Receptor Blockade in the Amygdala Mitigates Pain Like-Behaviors by Inhibiting Corticotropin Releasing Factor Neurons in a Rat Model of Functional Pain. Front Pharmacol 2022; 13:903978. [PMID: 35694266 PMCID: PMC9177060 DOI: 10.3389/fphar.2022.903978] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 01/06/2023] Open
Abstract
Functional pain syndromes (FPS) occur in the absence of identifiable tissue injury or noxious events and include conditions such as migraine, fibromyalgia, and others. Stressors are very common triggers of pain attacks in various FPS conditions. It has been recently demonstrated that kappa opioid receptors (KOR) in the central nucleus of amygdala (CeA) contribute to FPS conditions, but underlying mechanisms remain unclear. The CeA is rich in KOR and encompasses major output pathways involving extra-amygdalar projections of corticotropin releasing factor (CRF) expressing neurons. Here we tested the hypothesis that KOR blockade in the CeA in a rat model of FPS reduces pain-like and nocifensive behaviors by restoring inhibition of CeA-CRF neurons. Intra-CeA administration of a KOR antagonist (nor-BNI) decreased mechanical hypersensitivity and affective and anxiety-like behaviors in a stress-induced FPS model. In systems electrophysiology experiments in anesthetized rats, intra-CeA application of nor-BNI reduced spontaneous firing and responsiveness of CeA neurons to peripheral stimulation. In brain slice whole-cell patch-clamp recordings, nor-BNI increased feedforward inhibitory transmission evoked by optogenetic and electrical stimulation of parabrachial afferents, but had no effect on monosynaptic excitatory transmission. Nor-BNI decreased frequency, but not amplitude, of spontaneous inhibitory synaptic currents, suggesting a presynaptic action. Blocking KOR receptors in stress-induced FPS conditions may therefore represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Vadim Yakhnitsa
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Matthew Hein
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Zack Griffin
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Olga Ponomareva
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Edita Navratilova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
15
|
Leconte C, Mongeau R, Noble F. Traumatic Stress-Induced Vulnerability to Addiction: Critical Role of the Dynorphin/Kappa Opioid Receptor System. Front Pharmacol 2022; 13:856672. [PMID: 35571111 PMCID: PMC9091501 DOI: 10.3389/fphar.2022.856672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Substance use disorders (SUD) may emerge from an individual’s attempt to limit negative affective states and symptoms linked to stress. Indeed, SUD is highly comorbid with chronic stress, traumatic stress, or post-traumatic stress disorder (PTSD), and treatments approved for each pathology individually often failed to have a therapeutic efficiency in such comorbid patients. The kappa-opioid receptor (KOR) and its endogenous ligand dynorphin (DYN), seem to play a key role in the occurrence of this comorbidity. The DYN/KOR function is increased either in traumatic stress or during drug use, dependence acquisition and DYN is released during stress. The behavioural effects of stress related to the DYN/KOR system include anxiety, dissociative and depressive symptoms, as well as increased conditioned fear response. Furthermore, the DYN/KOR system is implicated in negative reinforcement after the euphoric effects of a drug of abuse ends. During chronic drug consumption DYN/KOR functions increase and facilitate tolerance and dependence. The drug-seeking behaviour induced by KOR activation can be retrieved either during the development of an addictive behaviour, or during relapse after withdrawal. DYN is known to be one of the most powerful negative modulators of dopamine signalling, notably in brain structures implicated in both reward and fear circuitries. KOR are also acting as inhibitory heteroreceptors on serotonin neurons. Moreover, the DYN/KOR system cross-regulate with corticotropin-releasing factor in the brain. The sexual dimorphism of the DYN/KOR system could be the cause of the gender differences observed in patients with SUD or/and traumatic stress-related pathologies. This review underlies experimental and clinical results emphasizing the DYN/KOR system as common mechanisms shared by SUD or/and traumatic stress-related pathologies, and suggests KOR antagonist as a new pharmacological strategy to treat this comorbidity.
Collapse
|
16
|
Blockade of kappa opioid receptors reduces mechanical hyperalgesia and anxiety-like behavior in a rat model of trigeminal neuropathic pain. Behav Brain Res 2022; 417:113595. [PMID: 34592375 DOI: 10.1016/j.bbr.2021.113595] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/12/2021] [Accepted: 09/20/2021] [Indexed: 12/18/2022]
Abstract
It has been shown that kappa opioid receptor (KOR) antagonists, such as nor-binaltorphimine (nor-BNI), have antinociceptive effects in some pain models that affect the trigeminal system. Also, its anxiolytic-like effect has been extensively demonstrated in the literature. The present study aimed to investigate the systemic, local, and central effect of nor-BNI on trigeminal neuropathic pain using the infraorbital nerve constriction model (CCI-ION), as well as to evaluate its effect on anxiety-like behavior associated with this model. Animals received nor-BNI systemically; in the trigeminal ganglion (TG); in the subarachnoid space to target the spinal trigeminal nucleus caudalis (Sp5C) or in the central amygdala (CeA) 14 days after CCI-ION surgery. Systemic administration of nor-BNI caused a significant reduction of facial mechanical hyperalgesia and promoted an anxiolytic-like effect, which was detected in the elevated plus-maze and the light-dark transition tests. When administered in the TG or CeA, the KOR antagonist was able to reduce facial mechanical hyperalgesia induced by CCI-ION, but without changing the anxiety-like behavior. Moreover, no change was observed on nociception and anxiety-like behavior after nor-BNI injection into the Sp5C. The present study demonstrated antinociceptive and anxiolytic-like effects of nor-BNI in a model of trigeminal neuropathic pain. The antinociceptive effect seems to be dissociated from the anxiolytic-like effect, at both the sites involved and at the dose need to achieve the effect. In conclusion, the kappa opioid system may represent a promising target to be explored for the control of trigeminal pain and associated anxiety. However, further studies are necessary to better elucidate its functioning and modulatory role in chronic trigeminal pain states.
Collapse
|
17
|
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
|
18
|
Adult-Attention Deficit Hyperactive Disorder Symptoms Seem Not to Influence the Outcome of an Enhanced Agonist Opioid Treatment: A 30-Year Follow-Up. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182010997. [PMID: 34682744 PMCID: PMC8535915 DOI: 10.3390/ijerph182010997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 11/24/2022]
Abstract
The role of opioids and opioid medications in ADHD symptoms is still largely understudied. We tested the hypothesis that, in Heroin Use Disorder (HUD), when patients are treated with Agonist Opioid medications (AOT), treatment outcome is associated with the presence of Adult Attention-Deficit/Hyperactive Disorder (A-ADHD) symptomatology. A retrospective cohort study of 130 HUD patients in Castelfranco Veneto, Italy, covering 30 years, was divided into two groups according to the Adult ADHD Self-Report Scale (ASRS) score and compared them using demographic, clinical and pharmacological factors. Survival in treatment was studied by utilizing the available data for leaving treatment and relapsing into addictive behavior and for mortality during treatment as poor primary outcomes. Thirty-five HUD subjects (26.9%) were unlikely to have A-ADHD symptomatology, and 95 (73.1%) were likely to have it. Only current age and co-substance use at treatment entry differed significantly between groups. Censored patients were 29 (82.9%) for HUD patients and 70 (73.9%) for A-ADHD/HUD patients (Mantel-Cox test = 0.66 p = 0.415). There were no significant linear trends indicative of a poorer outcome with the presence of A-ADHD after adjustment for demographic, clinical and pharmacological factors. Conclusions: ADHD symptomatology does not seem to exert any influence on the retention in AOT of HUD patients.
Collapse
|
19
|
McCullough KM, Missig G, Robbie MA, Foilb AR, Wells AM, Hartmann J, Anderson KJ, Neve RL, Nestler EJ, Ressler KJ, Carlezon WA. Nucleus Accumbens Medium Spiny Neuron Subtypes Differentially Regulate Stress-Associated Alterations in Sleep Architecture. Biol Psychiatry 2021; 89:1138-1149. [PMID: 33715826 PMCID: PMC8178228 DOI: 10.1016/j.biopsych.2020.12.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/17/2020] [Accepted: 12/20/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Stress is implicated in the pathophysiology of major depression and posttraumatic stress disorder. These conditions share core features, including motivational deficits, heighted anxiety, and sleep dysregulation. Chronic stress produces these same features in rodents, with some individuals being susceptible or resilient, as seen in humans. While stress-induced neuroadaptations within the nucleus accumbens are implicated in susceptibility-related dysregulation of motivational and emotional behaviors, their effects on sleep are unclear. METHODS We used chemogenetics (DREADDs [designer receptors exclusively activated by designer drugs]) to examine the effects of selective alterations in activity of nucleus accumbens medium spiny neurons expressing dopamine D1 receptors (D1-MSNs) or dopamine D2 receptors (D2-MSNs) on sleep-related end points. Mice were implanted with wireless transmitters enabling continuous collection of data to quantify vigilance states over a 20-day test period. Parallel cohorts were examined in behavioral tests assessing stress susceptibility. RESULTS D1- and D2-MSNs play dissociable roles in sleep regulation. Stimulation of inhibitory or excitatory DREADDs expressed in D1-MSNs exclusively affects rapid eye movement sleep, whereas targeting D2-MSNs affects slow wave sleep. The combined effects of D1-MSN inhibition and D2-MSN activation on sleep resemble those of chronic social defeat stress. Alterations in D1-MSN function also affect stress susceptibility in social behavior tests. Elevation of CREB (cAMP response element-binding protein) within D1-MSNs is sufficient to produce stress-like effects on rapid eye movement sleep. CONCLUSIONS In addition to regulation of motivational and emotional behaviors, the nucleus accumbens also influences sleep, an end point with high translational relevance. These findings provide a neural basis for comorbidity in key features of stress-related illness.
Collapse
Affiliation(s)
- Kenneth M. McCullough
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Galen Missig
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Mykel A. Robbie
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Allison R. Foilb
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Audrey M. Wells
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Jakob Hartmann
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Kasey J. Anderson
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Rachael L. Neve
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai. New York, NY 10029, USA
| | - Kerry J. Ressler
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - William A. Carlezon
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA,Corresponding Author: William A. Carlezon, Jr., Ph.D., Department of Psychiatry, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA,
| |
Collapse
|
20
|
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
|
21
|
Lee GJ, Kim SA, Kim YJ, Oh SB. Naloxone-induced analgesia mediated by central kappa opioid system in chronic inflammatory pain. Brain Res 2021; 1762:147445. [PMID: 33766518 DOI: 10.1016/j.brainres.2021.147445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/07/2021] [Accepted: 03/17/2021] [Indexed: 12/22/2022]
Abstract
Opioids, which are widely used for the treatment of chronic pain, have an analgesic effect by mainly activating mu-opioid receptor (MOR). Paradoxically, a high dose of naloxone, non-selective opioid receptor antagonist, is also known to induce analgesia, but the underlying mechanism remains unclear. Since kappa-opioid receptor (KOR) and dynorphin (KOR ligand) have been implicated in the naloxone-induced analgesia, we aimed to elucidate its mechanism by focusing on the kappa-opioid system in the brain under inflammatory pain condition. Systemic administration of naloxone (10 mg/kg, i.p.) decreased spontaneous pain behaviors only in complete Freund's adjuvant (CFA)-induced chronic inflammatory pain model but not in the formalin-induced acute pain model. Immunohistochemistry analysis in the CFA model revealed both a significant decrease in MOR expression and an increase in prodynorphin density in the central nucleus of theamygdala (CeA) and nucleus accumbens (NAc) but not in other brain areas. Systemic administration of KOR antagonist (norbinaltorphimine, nor-BNI 10 mg/kg) also decreased spontaneous pain behaviors in the CFA model. Furthermore, microinjection of both naloxone and nor-BNI into NAc and CeA significantly reduced spontaneous chronic pain behavior. Taken together, our results suggest that naloxone-induced analgesia may be mediated by blocking facilitated kappa-opioid systems in the NAc and CeA.
Collapse
Affiliation(s)
- Grace J Lee
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Shin Ae Kim
- Dental Research Institute and Department of Neurobiology & Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Yea Jin Kim
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seog Bae Oh
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea; Dental Research Institute and Department of Neurobiology & Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Pajser A, Fisher H, Pickens CL. Pre-training naltrexone increases conditioned fear learning independent of adolescent alcohol consumption history. Physiol Behav 2021; 229:113212. [PMID: 33069685 DOI: 10.1016/j.physbeh.2020.113212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/26/2020] [Accepted: 10/14/2020] [Indexed: 11/27/2022]
Abstract
Our previous research has shown a relationship between low voluntary alcohol consumption and high conditioned fear in male Long Evans rats. Here, we examined whether differences in the endogenous opioid systems might be responsible for these differences. Rats received 6 weeks of chronic intermittent to 20% alcohol (v/v) or water-only from PND 26-66. Based on their consumption during the last 2 weeks of alcohol access, the alcohol-access rats were divided into high drinking (>2.5 g/kg/24-h) or low drinking (<2 g/kg/24-h). Rats were then given injections of the preferential mu opioid receptor antagonist naltrexone (1 mg/kg, s.c.) or the selective kappa opioid receptor antagonist LY2456302 (10 mg/kg, s.c.) prior to fear conditioning and were then tested for conditioned fear 2 days later. Pre-training naltrexone increased conditioned suppression of lever-pressing during training and testing, with no differences between high versus low alcohol drinkers or between water-only versus alcohol access groups (averaged across drinking levels). There was no effect of LY2456302 on conditioned fear in any comparison. We also found no differences between high and low alcohol drinkers and no reliable effect of prior alcohol access (averaged across drinking levels) on conditioned fear. Our experiment replicates and extends previous demonstrations that a preferential MOR antagonist can increase fear learning using conditioned suppression of lever-pressing as a fear measure. However, additional research is needed to determine the cause of the differences in conditioned fear that we previously observed (as they were not observed in the current experiments).
Collapse
Affiliation(s)
- Alisa Pajser
- Department of Psychological Sciences, Kansas State University, Manhattan, KS 66506, USA
| | - Hayley Fisher
- Department of Psychological Sciences, Kansas State University, Manhattan, KS 66506, USA
| | - Charles L Pickens
- Department of Psychological Sciences, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
23
|
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
|
24
|
Vázquez-León P, Arenas-Martínez U, Córdova-Maqueda D, Fregoso-Aguilar T, Juan ERS, Miranda-Páez A. Salvia divinorum increases alcohol intake and tonic immobility whilst decreasing food intake in Wistar rats. Acta Neurobiol Exp (Wars) 2021; 81:34-42. [PMID: 33949161 DOI: 10.21307/ane-2021-005] [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: 03/23/2020] [Accepted: 01/13/2021] [Indexed: 11/11/2022]
Abstract
The kappa-opioid system (KOP) is the key in drug abuse. Of all the compounds isolated from Salvia divinorum (S. divinorum), salvinorin-A (Sal-A) is predominant. Further, Sal-A is the only compound within S. divinorum which is reported to have psychoactive properties as a powerful kappa-opioid receptor (KOPr) agonist. Based on the key role of the KOP system in the consumption of drugs, S. divinorum extract (SDE) and Sal-A may modify the alcohol intake in Wistar rats. Assessing voluntary alcohol intake as a drug consummatory behavior, food intake as natural reward behavior and tonic immobility as indicative of anxiety-like behavior, the present study sought to identify the role of both SDE and Sal-A in the Wistar rat model. Forty-eight adult male rats were randomly divided into six groups: control, alcohol naive and vehicle, alcohol-naive and SDE, alcohol-naive and Sal-A, alcohol-consumption and vehicle, alcohol-consumption and SDE, and alcohol-consumption and Sal-A. Alcohol and food intake were assessed for two weeks. In the middle of these two weeks, vehicle, SDE (containing ~1 mg/kg of Sal-A) or Sal-A was injected intraperitoneally once a day for a week. Tonic immobility testing was performed once. The administration of SDE produced a significant increase in voluntary alcohol intake especially in rats with a history of forced alcohol consumption from a juvenile age, Sal-A elicited an increase in alcohol intake in animals with or without previous alcohol exposure, SDE and Sal-A prolonged the tonic immobility duration and decreased food intake. In conclusion, S. divinorum or Sal-A stimulated alcohol consumption in rats with a history of alcohol intake and independent of previous exposure respectively, also SDE or Sal-A elicited an anorexigenic effect, and increased tonic immobility as indicative of anxious-like behavior. The kappa-opioid system (KOP) is the key in drug abuse. Of all the compounds isolated from Salvia divinorum (S. divinorum), salvinorin-A (Sal-A) is predominant. Further, Sal-A is the only compound within S. divinorum which is reported to have psychoactive properties as a powerful kappa-opioid receptor (KOPr) agonist. Based on the key role of the KOP system in the consumption of drugs, S. divinorum extract (SDE) and Sal-A may modify the alcohol intake in Wistar rats. Assessing voluntary alcohol intake as a drug consummatory behavior, food intake as natural reward behavior and tonic immobility as indicative of anxiety-like behavior, the present study sought to identify the role of both SDE and Sal-A in the Wistar rat model. Forty-eight adult male rats were randomly divided into six groups: control, alcohol naive and vehicle, alcohol-naive and SDE, alcohol-naive and Sal-A, alcohol-consumption and vehicle, alcohol-consumption and SDE, and alcohol-consumption and Sal-A. Alcohol and food intake were assessed for two weeks. In the middle of these two weeks, vehicle, SDE (containing ~1 mg/kg of Sal-A) or Sal-A was injected intraperitoneally once a day for a week. Tonic immobility testing was performed once. The administration of SDE produced a significant increase in voluntary alcohol intake especially in rats with a history of forced alcohol consumption from a juvenile age, Sal-A elicited an increase in alcohol intake in animals with or without previous alcohol exposure, SDE and Sal-A prolonged the tonic immobility duration and decreased food intake. In conclusion, S. divinorum or Sal-A stimulated alcohol consumption in rats with a history of alcohol intake and independent of previous exposure respectively, also SDE or Sal-A elicited an anorexigenic effect, and increased tonic immobility as indicative of anxious-like behavior.
Collapse
Affiliation(s)
- Priscila Vázquez-León
- Departamento de Fisiología , Escuela Nacional de Ciencias Biológicas , Instituto Politécnico Nacional , Wilfrido Massieu esq. Manuel Stampa s/n , Col. Nueva Industrial Vallejo , Alcaldía Gustavo A. Madero , México City , Mexico ; Departamento de Fisiología y Farmacología , Centro de Ciencias Básicas , Universidad Autónoma de Aguascalientes , Ciudad Universitaria , Aguascalientes, Ags. , Mexico
| | - Ulises Arenas-Martínez
- Departamento de Fisiología , Escuela Nacional de Ciencias Biológicas , Instituto Politécnico Nacional , Wilfrido Massieu esq. Manuel Stampa s/n , Col. Nueva Industrial Vallejo , Alcaldía Gustavo A. Madero , México City , Mexico
| | - Dafne Córdova-Maqueda
- Laboratorio de Palinología , Departamento de Botánica , Escuela Nacional de Ciencias Biológicas , Instituto Politécnico Nacional , Plan de Ayala y Prolongación de Carpio s/n , Alcaldía Miguel Hidalgo , México City , Mexico
| | - Tomás Fregoso-Aguilar
- Departamento de Fisiología , Escuela Nacional de Ciencias Biológicas , Instituto Politécnico Nacional , Wilfrido Massieu esq. Manuel Stampa s/n , Col. Nueva Industrial Vallejo , Alcaldía Gustavo A. Madero , México City , Mexico
| | - Eduardo Ramírez-San Juan
- Departamento de Fisiología , Escuela Nacional de Ciencias Biológicas , Instituto Politécnico Nacional , Wilfrido Massieu esq. Manuel Stampa s/n , Col. Nueva Industrial Vallejo , Alcaldía Gustavo A. Madero , México City , Mexico
| | - Abraham Miranda-Páez
- Departamento de Fisiología , Escuela Nacional de Ciencias Biológicas , Instituto Politécnico Nacional , Wilfrido Massieu esq. Manuel Stampa s/n , Col. Nueva Industrial Vallejo , Alcaldía Gustavo A. Madero , México City , Mexico
| |
Collapse
|
25
|
Tejeda HA, Wang H, Flores RJ, Yarur HE. Dynorphin/Kappa-Opioid Receptor System Modulation of Cortical Circuitry. Handb Exp Pharmacol 2021; 271:223-253. [PMID: 33580392 DOI: 10.1007/164_2021_440] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cortical circuits control a plethora of behaviors, from sensation to cognition. The cortex is enriched with neuropeptides and receptors that play a role in information processing, including opioid peptides and their cognate receptors. The dynorphin (DYN)/kappa-opioid receptor (KOR) system has been implicated in the processing of sensory and motivationally-charged emotional information and is highly expressed in cortical circuits. This is important as dysregulation of DYN/KOR signaling in limbic and cortical circuits has been implicated in promoting negative affect and cognitive deficits in various neuropsychiatric disorders. However, research investigating the role of this system in controlling cortical circuits and computations therein is limited. Here, we review the (1) basic anatomy of cortical circuits, (2) anatomical architecture of the cortical DYN/KOR system, (3) functional regulation of cortical synaptic transmission and microcircuit function by the DYN/KOR system, (4) regulation of behavior by the cortical DYN/KOR system, (5) implications for the DYN/KOR system for human health and disease, and (6) future directions and unanswered questions for the field. Further work elucidating the role of the DYN/KOR system in controlling cortical information processing and associated behaviors will be of importance to increasing our understanding of principles underlying neuropeptide modulation of cortical circuits, mechanisms underlying sensation and perception, motivated and emotional behavior, and cognition. Increased emphasis in this area of study will also aid in the identification of novel ways to target the DYN/KOR system to treat neuropsychiatric disorders.
Collapse
Affiliation(s)
- Hugo A Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Huikun Wang
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Rodolfo J Flores
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Hector E Yarur
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Kappa Opioid Receptors in the Pathology and Treatment of Major Depressive Disorder. Handb Exp Pharmacol 2021; 271:493-524. [PMID: 33580854 DOI: 10.1007/164_2020_432] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The kappa opioid receptor (KOR) is thought to regulate neural systems associated with anhedonia and aversion and mediate negative affective states that are associated with a number of psychiatric disorders, but especially major depressive disorder (MDD). Largely because KOR antagonists mitigate the effects of stress in preclinical studies, KOR antagonists have been recommended as novel drugs for treating MDD. The purpose of this review is to examine the role of KORs and its endogenous ligand dynorphins (DYNs) in the pathology and treatment of MDD derived from different types of clinical studies. Evidence pertaining to the role of KOR and MDD will be reviewed from (1) post mortem mRNA expression patterns in MDD, (2) the utility of KOR neuroimaging agents and serum biomarkers in MDD, and (3) evidence from the recent Fast Fail clinical trial that established KOR antagonism as a potential therapeutic strategy for the alleviation of anhedonia, a core feature of MDD. These findings are compared with a focused evaluation of stress-induced alterations in OPRK and PDYN mRNA expression. Finally, the current status of the effects of KOR antagonists on behavioral phenotypes of stress in preclinical studies related to MDD is summarized.
Collapse
|
27
|
Zhang L, McCarthy DM, Eskow Jaunarajs KL, Biederman J, Spencer TJ, Bhide PG. Frontal Cortical Monoamine Release, Attention, and Working Memory in a Perinatal Nicotine Exposure Mouse Model Following Kappa Opioid Receptor Antagonism. Cereb Cortex 2021; 31:483-496. [PMID: 32869057 DOI: 10.1093/cercor/bhaa238] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 02/27/2024] Open
Abstract
Perinatal nicotine exposure (PNE) produces frontal cortical hypo-dopaminergic state and attention and working memory deficits consistent with neurodevelopmental disorders such as attention deficit hyperactivity disorder (ADHD). Methylphenidate alleviates ADHD symptoms by increasing extracellular dopamine and noradrenaline. Kappa opioid receptor (KOR) antagonism may be another mechanism to achieve the same results because KOR activation inhibits frontal cortical dopamine release. We administered the selective KOR antagonist norbinaltorphimine (norBNI) (20 mg/kg; intraperitoneal) or methylphenidate (0.75 mg/kg; intraperitoneal) to PNE mouse model and examined frontal cortical monoamine release, attention, and working memory. Both compounds increased dopamine and noradrenaline release but neither influenced serotonin release. Both compounds improved object-based attention and working memory in the PNE group, with norBNI's effects evident at 2.5 h and 5.5 h but absent at 24 h. Methylphenidate's effects were evident at 0.5 h but not at 2.5 h. norBNI's effects temporally coincided with frontal cortical c-Jun N-terminal kinase phosphorylation. norBNI did not alter tissue dopamine content in the nucleus accumbens, offering preliminary support for lack of reinforcement.
Collapse
Affiliation(s)
- Lin Zhang
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Deirdre M McCarthy
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | | | - Joseph Biederman
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Thomas J Spencer
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pradeep G Bhide
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
28
|
Lorente JD, Cuitavi J, Campos-Jurado Y, Hipólito L. Pain-induced alterations in the dynorphinergic system within the mesocorticolimbic pathway: Implication for alcohol addiction. J Neurosci Res 2020; 100:165-182. [PMID: 32770601 DOI: 10.1002/jnr.24703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/08/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022]
Abstract
Latest studies have revealed that pain negatively impacts on reward processing and motivation leading to negative affective states and stress. These states not only reduce quality of life of patients by increasing the appearance of psychiatric comorbidities, but also have an important impact on vulnerability to drug abuse, including alcohol. In fact, clinical, epidemiological but also preclinical studies have revealed that the presence of pain is closely related to alcohol use disorders (AUDs). All this evidence suggests that pain is a factor that increases the risk of suffering AUD, predicting heavy drinking behavior and relapse drinking in those patients with a previous history of AUD. The negative consequences of chronic pain and its impact on stress and AUD are likely mediated by alterations in the central nervous system, especially in the stress and reward systems. Therefore, pain and stress impact on dopaminergic mesolimbic pathway can lead to an increase in drug abuse liability. In this mini review we analyze the interaction between pain, stress, and alcohol addiction, and how dynamic changes in the kappa opioid system might play a crucial role in the development of compulsive alcohol drinking in chronic pain patients.
Collapse
Affiliation(s)
- Jesús David Lorente
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of València, Burjassot, Spain
| | - Javier Cuitavi
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of València, Burjassot, Spain
| | - Yolanda Campos-Jurado
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of València, Burjassot, Spain
| | - Lucía Hipólito
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of València, Burjassot, Spain
| |
Collapse
|
29
|
Horovitz O, Ardi Z, Ashkenazi SK, Ritov G, Anunu R, Richter-Levin G. Network Neuromodulation of Opioid and GABAergic Receptors Following a Combination of "Juvenile" and "Adult Stress" in Rats. Int J Mol Sci 2020; 21:ijms21155422. [PMID: 32751453 PMCID: PMC7432657 DOI: 10.3390/ijms21155422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Early life stress is suggested to alter behavioral responses during stressful challenges in adulthood and to exacerbate pathological symptoms that reminisce posttraumatic stress disorder (PTSD). These effects are often associated with changes in γ-Aminobutyric acid type A (GABAA) and κ opioid receptor expression and neuromodulation of the limbic system. Anxiety-like and stress coping behaviors were assessed in rats exposed to stress in adulthood on the background of previous exposure to stress in juvenility. Two weeks following behavioral assessment in adulthood, GABAAR α1 and α2 subunits and κ opioid receptor expression levels were measured in the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), amygdala, and periaqueductal gray (PAG). To illustrate changes at the network level, an integrated expression profile was constructed. We found that exposure to juvenile stress affected rats’ behavior during adult stress. The combination of juvenile and adult stress significantly affected rats’ long term anxious-like behavior. Probabilities predicting model integrating the expression of GABAA α1-α2 and κ opioid receptors in different brain regions yielded highly successful classification rates. This study emphasizes the ability of exposure to stress in juvenility to exacerbate the impact of coping with stress in adulthood. Moreover, the use of integrated receptor expression network profiling was found to effectively characterize the discussed affective styles and their behavioral manifestations.
Collapse
Affiliation(s)
- Omer Horovitz
- Psychology Department, Tel-Hai Academic College, Haifa 1220800, Israel;
| | - Ziv Ardi
- Department of Behavioral Sciences, Kinneret Academic College on the Sea of Galilee, Sea of Galilee 15132, Israel
- Correspondence: ; Tel.: +972-46653803
| | - Shiri Karni Ashkenazi
- Sagol Department of Neurobiology, University of Haifa, Mount Carmel 31095, Israel; (S.K.A.); (G.R.-L.)
| | - Gilad Ritov
- The Institute for the Study of Affective Neuroscience (ISAN), Mount Carmel 31095, Israel; (G.R.); (R.A.)
| | - Rachel Anunu
- The Institute for the Study of Affective Neuroscience (ISAN), Mount Carmel 31095, Israel; (G.R.); (R.A.)
| | - Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Mount Carmel 31095, Israel; (S.K.A.); (G.R.-L.)
- The Institute for the Study of Affective Neuroscience (ISAN), Mount Carmel 31095, Israel; (G.R.); (R.A.)
- Psychology Department, University of Haifa, Mount Carmel 31095, Israel
| |
Collapse
|
30
|
Haun HL, Griffin WC, Lopez MF, Becker HC. Kappa opioid receptors in the bed nucleus of the stria terminalis regulate binge-like alcohol consumption in male and female mice. Neuropharmacology 2020; 167:107984. [PMID: 32023486 PMCID: PMC7080606 DOI: 10.1016/j.neuropharm.2020.107984] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/20/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022]
Abstract
Binge drinking is the most common pattern of excessive alcohol consumption and is a significant contributor to the development of Alcohol Use Disorder and dependence. Previous studies demonstrated involvement of kappa opioid receptors (KOR) in binge-like drinking in mice using the Drinking-in-the-Dark model. The current studies examined the role of KOR specifically in the bed nucleus of the stria terminals (BNST) in binge-like alcohol consumption in male and female mice. Direct administration of the long lasting KOR antagonist, nor-BNI, into the BNST decreased binge-like alcohol consumption and blood alcohol concentrations in male and female C57BL/6J mice. Similarly, direct nor-BNI administration into the BNST modestly reduced sucrose consumption and the suppression of fluid intake was not related to reduced locomotor activity. To further determine the role of KOR within the BNST on binge-like alcohol consumption, the KOR agonist U50,488 was administered systemically which resulted in a robust increase in alcohol intake. Microinjection of nor-BNI into the BNST blocked the high level of alcohol intake after systemic U50,488 challenge reducing intake and resultant blood alcohol concentrations. Together, these data suggest that KOR activity in the BNST contributes to binge-like alcohol consumption in both male and female mice. This article is part of the special issue on 'Neuropeptides'.
Collapse
Affiliation(s)
- Harold L Haun
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Howard C Becker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; RHJ Department of Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
31
|
Abstract
BACKGROUND Depression and post-traumatic stress disorder (PTSD) are leading causes of disability and loss of life by suicide. Currently, there are less than satisfactory medical solutions to treat these mental disorders. Here, we explore recent preclinical and clinical studies demonstrating the potential of using buprenorphine to treat major depressive disorder, treatment-resistant depression, and PTSD. METHOD Bibliographic databases were searched to include preclinical and clinical studies demonstrating the therapeutic potential of buprenorphine and the involvement of the kappa opioid receptor (KOR) in mediating these effects. RESULTS Original clinical studies examining the effectiveness of buprenorphine to treat depression were mixed. The majority of participants in the PTSD studies were males and suffer from chronic pain and/or substance use disorders. Nonetheless, these recent studies and analyses established proof of concept warranting farther investigations. Additionally, KOR likely mediates the antidepressant and some of the anxiolytic effects of buprenorphine. Still, it appears that the full spectrum of buprenorphine's beneficial effects might be due to activity at other opioid receptors as well. CONCLUSIONS Pharmaceuticals' abilities to treat medical conditions directly relates to their ability to act upon the endogenous biological systems related to the conditions. Thus, these recent findings are likely a reflection of the central role that the endogenous opioid system has in these mental illnesses. Further studies are necessary to study the involvement of endogenous opioid systems, and specifically KOR, in mediating buprenorphine's beneficial effects and the ability to treat these medical conditions while minimizing risks for misuse and diversion.
Collapse
Affiliation(s)
- Caitlin A Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX77843, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX77843, USA
| |
Collapse
|
32
|
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
|
33
|
Affiliation(s)
- Marlene A Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Columbia VA Health Care System, Columbia, SC, United States
| | - Alexander J McDonald
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
34
|
Goldman A, Smalley JL, Mistry M, Krenzlin H, Zhang H, Dhawan A, Caldarone B, Moss SJ, Silbersweig DA, Lawler SE, Braun IM. A computationally inspired in-vivo approach identifies a link between amygdalar transcriptional heterogeneity, socialization and anxiety. Transl Psychiatry 2019; 9:336. [PMID: 31819040 PMCID: PMC6901550 DOI: 10.1038/s41398-019-0677-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/23/2019] [Accepted: 11/06/2019] [Indexed: 01/22/2023] Open
Abstract
Pharmaceutical breakthroughs for anxiety have been lackluster in the last half-century. Converging behavior and limbic molecular heterogeneity has the potential to revolutionize biomarker-driven interventions. However, current in vivo models too often deploy artificial systems including directed evolution, mutations and fear induction, which poorly mirror clinical manifestations. Here, we explore transcriptional heterogeneity of the amygdala in isogenic mice using an unbiased multi-dimensional computational approach that segregates intra-cohort reactions to moderate situational adversity and intersects it with high content molecular profiling. We show that while the computational approach stratifies known features of clinical anxiety including nitric oxide, opioid and corticotropin signaling, previously unrecognized druggable biomarkers emerge, such as calpain11 and scand1. Through ingenuity pathway analyses, we further describe a role for neurosteroid estradiol signaling, heat shock proteins, ubiquitin ligases and lipid metabolism. In addition, we report a remarkable behavioral pattern that maps to molecular features of anxiety in mice through counterphobic social attitudes, which manifest as increased, yet spatially distant socialization. These findings provide an unbiased approach for interrogating anxiolytics, and hint toward biomarkers underpinning behavioral and social patterns that merit further exploration.
Collapse
Affiliation(s)
- Aaron Goldman
- Harvard Medical School, Boston, USA. .,Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA.
| | - Joshua L. Smalley
- 0000 0000 8934 4045grid.67033.31Department of Neuroscience, Tufts University School of Medicine, Boston, USA
| | - Meeta Mistry
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, USA ,000000041936754Xgrid.38142.3cDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Harald Krenzlin
- 0000 0004 0378 8294grid.62560.37Harvey Cushing Neurooncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Boston, USA
| | - Hong Zhang
- 0000 0004 0378 8294grid.62560.37Harvey Cushing Neurooncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Boston, USA
| | - Andrew Dhawan
- 0000 0001 0675 4725grid.239578.2Neurological Institute, Cleveland Clinic, Cleveland, OH USA
| | - Barbara Caldarone
- 000000041936754Xgrid.38142.3cDepartment of Genetics, Harvard Medical School, Boston, USA
| | - Stephen J. Moss
- 0000 0000 8934 4045grid.67033.31Department of Neuroscience, Tufts University School of Medicine, Boston, USA ,0000000121901201grid.83440.3bDepartment of Neuroscience, Physiology and Pharmacology, University College, London, UK
| | - David A. Silbersweig
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, USA ,0000 0004 0378 8294grid.62560.37Department of Psychiatry, Brigham and Women’s Hospital, Boston, USA
| | - Sean E. Lawler
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, USA ,0000 0004 0378 8294grid.62560.37Harvey Cushing Neurooncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Boston, USA
| | - Ilana M. Braun
- 000000041936754Xgrid.38142.3cHarvard Medical School, Boston, USA ,0000 0004 0378 8294grid.62560.37Department of Psychiatry, Brigham and Women’s Hospital, Boston, USA ,0000 0001 2106 9910grid.65499.37Department of Psychosocial Oncology and Palliative Care, Dana Farber Cancer Institute, Boston, USA
| |
Collapse
|
35
|
Neumann AM, Blondell RD, Hoopsick RA, Homish GG. Randomized clinical trial comparing buprenorphine/naloxone and methadone for the treatment of patients with failed back surgery syndrome and opioid addiction. J Addict Dis 2019; 38:33-41. [PMID: 31774028 DOI: 10.1080/10550887.2019.1690929] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Opioid analgesic consumption has led to an unprecedented epidemic of overdose death and opioid addiction in the US history. The treatment of chronic pain in patients with opioid addiction who receive prescriptions for opioid medications presents a clinical dilemma. Continuing opioid medication could result in hyperalgesia rendering opioids ineffective and results in iatrogenic therapeutic damage as evidenced by the worsening of addiction. Discontinuing opioid medications could result in severe pain and cravings that often leads the patient to the illicit market. This study compared methadone and buprenorphine/naloxone in patients with failed back surgery syndrome and opioid addiction. Nineteen participants were randomly assigned to methadone or buprenorphine/naloxone and were followed for 6 months. In an intent-to-treat analysis analgesia, craving, functioning, drug use, depression, and treatment retention were assessed monthly. It was planned to enroll 66 patients with failed back surgery syndrome and opioid addiction; however, enrollment was closed early due to suspected abuse of medications. Patients in both treatment conditions exhibited significantly improved 24-hour pain severity with up to 20% reduction of pain severity at the last follow-up (p < .05). However, patients receiving methadone reported significantly reduced current pain severity, whereas patients receiving buprenorphine/naloxone did not. Patients reported significantly improved functioning, fewer cravings, less opioid use, and depression (p < .05) across the treatment conditions. When given a choice between methadone and buprenorphine/naloxone, buprenorphine/naloxone is recommended due to its superior safety profile. Treatment with either needs to be monitored closely.
Collapse
Affiliation(s)
- Anne M Neumann
- Primary Care Research Institute, Department of Family Medicine, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Richard D Blondell
- Primary Care Research Institute, Department of Family Medicine, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Rachel A Hoopsick
- Primary Care Research Institute, Department of Family Medicine, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Gregory G Homish
- Department of Community Health and Health Behavior, The State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
36
|
Addiction and stress: An allostatic view. Neurosci Biobehav Rev 2019; 106:245-262. [DOI: 10.1016/j.neubiorev.2018.09.008] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 08/30/2018] [Accepted: 09/08/2018] [Indexed: 01/24/2023]
|
37
|
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
|
38
|
Mavrikaki M, Pantano L, Potter D, Rogers-Grazado MA, Anastasiadou E, Slack FJ, Amr SS, Ressler KJ, Daskalakis NP, Chartoff E. Sex-Dependent Changes in miRNA Expression in the Bed Nucleus of the Stria Terminalis Following Stress. Front Mol Neurosci 2019; 12:236. [PMID: 31636537 PMCID: PMC6788329 DOI: 10.3389/fnmol.2019.00236] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/17/2019] [Indexed: 01/21/2023] Open
Abstract
Anxiety disorders disproportionately affect women compared to men, which may arise from sex differences in stress responses. MiRNAs are small non-coding RNAs known to regulate gene expression through actions on mRNAs. MiRNAs are regulated, in part, by factors such as stress and gonadal sex, and they have been implicated in the pathophysiology of multiple psychiatric disorders. Here, we assessed putative sex differences in miRNA expression in the bed nucleus of the stria terminalis (BNST) - a sexually dimorphic brain region implicated in anxiety - of adult male and female rats that had been exposed to social isolation (SI) stress throughout adolescence. To assess the translational utility of our results, we assessed if childhood trauma in humans resulted in changes in blood miRNA expression that are similar to those observed in rats. Male and female Sprague-Dawley rats underwent SI during adolescence or remained group housed (GH) and were tested for anxiety-like behavior in the elevated plus maze as adults. Small RNA sequencing was performed on tissue extracted from the BNST. Furthermore, we re-analyzed an already available small RNA sequencing data set from the Grady Trauma Project (GTP) from men and women to identify circulating miRNAs that are associated with childhood trauma exposure. Our results indicated that there were greater anxiogenic-like effects and changes in BNST miRNA expression in SI versus GH females compared to SI versus GH males. In addition, we found nine miRNAs that were regulated in both the BNST from SI compared to GH rats and in blood samples from humans exposed to childhood trauma. These studies emphasize the utility of rodent models in studying neurobiological mechanisms underlying psychiatric disorders and suggest that rodent models could be used to identify novel sex-specific pharmacotherapies for anxiety disorders.
Collapse
Affiliation(s)
- Maria Mavrikaki
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| | - Lorena Pantano
- Harvard Chan Bioinformatics Core, Harvard School of Public Health, Harvard University, Boston, MA, United States
| | - David Potter
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| | | | - Eleni Anastasiadou
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Frank J. Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Sami S. Amr
- Translational Genomics Core, Partners Healthcare Personalized Medicine, Cambridge, MA, United States
| | - Kerry J. Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| | - Nikolaos P. Daskalakis
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| | - Elena Chartoff
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| |
Collapse
|
39
|
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]
|
40
|
Hruby VJ. Multivalent peptide and peptidomimetic ligands for the treatment of pain without toxicities and addiction. Peptides 2019; 116:63-67. [PMID: 31014958 DOI: 10.1016/j.peptides.2019.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
The current opioid crisis has created a tragic problem in medicine and society. Pain is the most ubiquitous and costly disease in society and yet all of our "treatments" have toxicities, especially for prolonged use. However, there are several alternatives that have been discovered in the past fifteen years that have been demonstrated in animals to have none of the toxicities of current drugs. Many of the compounds are multivalent and have novel biological activity profiles. Unfortunately, none of these have been in clinical trials in humans, perhaps because they were discovered in academic laboratories. A review of these novel chemicals are given in this paper.
Collapse
MESH Headings
- Analgesics, Opioid/therapeutic use
- Animals
- Humans
- Ligands
- Opioid Peptides/chemistry
- Opioid Peptides/therapeutic use
- Pain/drug therapy
- Pain/pathology
- Pain Management
- Peptides/adverse effects
- Peptides/therapeutic use
- Peptidomimetics/adverse effects
- Peptidomimetics/therapeutic use
- Receptors, Opioid/chemistry
- Receptors, Opioid/therapeutic use
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, mu/chemistry
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- Victor J Hruby
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
41
|
da Cruz KR, Ianzer D, Turones LC, Reis LL, Camargo-Silva G, Mendonça MM, da Silva ES, Pedrino GR, de Castro CH, Costa EA, Xavier CH. Behavioral effects evoked by the beta globin-derived nonapeptide LVV-H6. Peptides 2019; 115:59-68. [PMID: 30890354 DOI: 10.1016/j.peptides.2019.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/04/2019] [Accepted: 03/12/2019] [Indexed: 02/05/2023]
Abstract
LVV-hemorphin-6 (LVV-h6) is bioactive peptide and is a product of the degradation of hemoglobin. Since LVV-h6 effects are possibly mediated by opioid or AT4/IRAP receptors, we hypothesized that LVV-h6 would modify behavior. We evaluated whether LVV-h6 affects: i) anxiety-like behavior and locomotion; ii) depression-like behavior; iii) cardiovascular and neuroendocrine reactivity to emotional stress. Male Wistar rats ( ± 300 g) received LVV-h6 (153 nmol/kg i.p.) or vehicle (NaCl 0.9% i.p.). We used: i) open field (OF) test for locomotion; ii) elevated plus maze (EPM) for anxiety-like behavior; iii) forced swimming test (FST) for depression-like behavior and iv) air jet for cardiovascular and neuroendocrine reactivity to stress. Diazepam (2 mg/kg i.p.) and imipramine (15 mg/kg i.p.) were used as positive control for EPM and FST, respectively. To evaluate the LVV-h6 mechanisms, we used: the antagonist of oxytocin (OT) receptors (atosiban - ATS 1 and 0.1 mg/kg i.p.); the inhibitor of tyrosine hydroxylase (Alpha-methyl-p-tyrosine - AMPT 200 mg/kg i.p.) to investigate the involvement of catecholaminergic paths; and the antagonist of opioid receptors (naltrexone - NTX 0.3 mg/kg s.c.). We found that LVV-h6: i) evoked anxiolytic-like effect; ii) evoked antidepressant-like effect in the FST; and iii) did not change the locomotion, neuroendocrine and cardiovascular responses to stress. The LVV-h6 anxiolytic-like effect was not reverted by ATS and AMPT. However, the antidepressant effects were reverted only by NTX. Hence, our findings demonstrate that LVV-h6 modulates anxiety-like behavior by routes that are not oxytocinergic, catecholaminergic or opioid. The antidepressant-like effects of LVV-h6 rely on opioid pathways.
Collapse
Affiliation(s)
- Kellen Rosa da Cruz
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Danielle Ianzer
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Larissa Córdova Turones
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Lilian Liz Reis
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Gabriel Camargo-Silva
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Michelle Mendanha Mendonça
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Elder Sales da Silva
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Gustavo Rodrigues Pedrino
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Carlos Henrique de Castro
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Elson Alves Costa
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Carlos H Xavier
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
42
|
Browne CA, Lucki I. Targeting opioid dysregulation in depression for the development of novel therapeutics. Pharmacol Ther 2019; 201:51-76. [PMID: 31051197 DOI: 10.1016/j.pharmthera.2019.04.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
Since the serendipitous discovery of the first class of modern antidepressants in the 1950's, all pharmacotherapies approved by the Food and Drug Administration for major depressive disorder (MDD) have shared a common mechanism of action, increased monoaminergic neurotransmission. Despite the widespread availability of antidepressants, as many as 50% of depressed patients are resistant to these conventional therapies. The significant length of time required to produce meaningful symptom relief with these medications, 4-6 weeks, indicates that other mechanisms are likely involved in the pathophysiology of depression which may yield more viable targets for drug development. For decades, no viable candidate target with a different mechanism of action to that of conventional therapies proved successful in clinical studies. Now several exciting avenues for drug development are under intense investigation. One of these emerging targets is modulation of endogenous opioid tone. This review will evaluate preclinical and clinical evidence pertaining to opioid dysregulation in depression, focusing on the role of the endogenous ligands endorphin, enkephalin, dynorphin, and nociceptin/orphanin FQ (N/OFQ) and their respective receptors, mu (MOR), delta (DOR), kappa (KOR), and the N/OFQ receptor (NOP) in mediating behaviors relevant to depression and anxiety. Finally, putative opioid based antidepressants that are under investigation in clinical trials, ALKS5461, JNJ-67953964 (formerly LY2456302 and CERC-501) and BTRX-246040 (formerly LY-2940094) will be discussed. This review will illustrate the potential therapeutic value of targeting opioid dysregulation in developing novel therapies for MDD.
Collapse
Affiliation(s)
- Caroline A Browne
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America
| | - Irwin Lucki
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| |
Collapse
|
43
|
Turner CA, Hagenauer MH, Aurbach EL, Maras PM, Fournier CL, Blandino P, Chauhan RB, Panksepp J, Watson SJ, Akil H. Effects of early-life FGF2 on ultrasonic vocalizations (USVs) and the mu-opioid receptor in male Sprague-Dawley rats selectively-bred for differences in their response to novelty. Brain Res 2019; 1715:106-114. [PMID: 30880118 DOI: 10.1016/j.brainres.2019.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 01/03/2023]
Abstract
In previous studies, early-life fibroblast growth factor-2 (FGF2) administration conferred resilience to developing anxiety-like behavior in vulnerable animals in adulthood. To follow up on this work, we administered FGF2 the day after birth to animals that differ in emotional behavior and further explored its long-term effects on affective behavior and circuitry. Selectively-bred "high responder" rats (bHRs) exhibit low levels of anxiety-like and depression-like behavior, whereas selectively-bred "low responders" (bLRs) display high levels of anxiety-like and depression-like behavior. We found that early-life administration of FGF2 decreased negative affect in bLRs during the early post-natal period, as indexed by 40 kHz ultrasonic vocalizations (USVs) in response to a brief maternal separation on PND11. FGF2 also increased positive affect during the juvenile period, as measured by 50 kHz USVs in response to heterospecific hand play ("tickling") after weaning. In general, we found that bHRs produced more 50 kHz USVs than bLRs. In adulthood, we measured opioid ligand and receptor expression in brain regions implicated in USV production and affect regulation by mRNA in situ hybridization. Within multiple affective brain regions, bHRs had greater expression of the mu opioid receptor than bLRs. FGF2 increased mu opioid expression in bLRs. The bLRs had more kappa and less delta receptor expression than bHRs, and FGF2 increased prodynorphin in bLRs. Our results provide support for further investigations into the role of growth factors and endogenous opioids in the treatment of disorders characterized by altered affect, such as anxiety and depression.
Collapse
Affiliation(s)
- Cortney A Turner
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Megan H Hagenauer
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Elyse L Aurbach
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Pamela M Maras
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Chelsea L Fournier
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Peter Blandino
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Rikav B Chauhan
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jaak Panksepp
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, USA
| | - Stanley J Watson
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Huda Akil
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
44
|
Burns JA, Kroll DS, Feldman DE, Kure Liu C, Manza P, Wiers CE, Volkow ND, Wang GJ. Molecular Imaging of Opioid and Dopamine Systems: Insights Into the Pharmacogenetics of Opioid Use Disorders. Front Psychiatry 2019; 10:626. [PMID: 31620026 PMCID: PMC6759955 DOI: 10.3389/fpsyt.2019.00626] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/05/2019] [Indexed: 12/21/2022] Open
Abstract
Opioid use in the United States has steadily risen since the 1990s, along with staggering increases in addiction and overdose fatalities. With this surge in prescription and illicit opioid abuse, it is paramount to understand the genetic risk factors and neuropsychological effects of opioid use disorder (OUD). Polymorphisms disrupting the opioid and dopamine systems have been associated with increased risk for developing substance use disorders. Molecular imaging studies have revealed how these polymorphisms impact the brain and contribute to cognitive and behavioral differences across individuals. Here, we review the current molecular imaging literature to assess how genetic variations in the opioid and dopamine systems affect function in the brain's reward, cognition, and stress pathways, potentially resulting in vulnerabilities to OUD. Continued research of the functional consequences of genetic variants and corresponding alterations in neural mechanisms will inform prevention and treatment of OUD.
Collapse
Affiliation(s)
- Jamie A Burns
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Danielle S Kroll
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Dana E Feldman
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | | | - Peter Manza
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States.,National Institute on Drug Abuse, Bethesda, MD, United States
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| |
Collapse
|
45
|
Becker JB, Chartoff E. Sex differences in neural mechanisms mediating reward and addiction. Neuropsychopharmacology 2019; 44:166-183. [PMID: 29946108 PMCID: PMC6235836 DOI: 10.1038/s41386-018-0125-6] [Citation(s) in RCA: 304] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/27/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022]
Abstract
There is increasing evidence in humans and laboratory animals for biologically based sex differences in every phase of drug addiction: acute reinforcing effects, transition from occasional to compulsive use, withdrawal-associated negative affective states, craving, and relapse. There is also evidence that many qualitative aspects of the addiction phases do not differ significantly between males and females, but one sex may be more likely to exhibit a trait than the other, resulting in population differences. The conceptual framework of this review is to focus on hormonal, chromosomal, and epigenetic organizational and contingent, sex-dependent mechanisms of four neural systems that are known-primarily in males-to be key players in addiction: dopamine, mu-opioid receptors (MOR), kappa opioid receptors (KOR), and brain-derived neurotrophic factor (BDNF). We highlight data demonstrating sex differences in development, expression, and function of these neural systems as they relate-directly or indirectly-to processes of reward and addictive behavior, with a focus on psychostimulants and opioids. We identify gaps in knowledge about how these neural systems interact with sex to influence addictive behavior, emphasizing throughout that the impact of sex can be highly nuanced and male/female data should be reported regardless of the outcome.
Collapse
Affiliation(s)
- Jill B Becker
- Department of Psychology and the Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Elena Chartoff
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
46
|
Azocar VH, Sepúlveda G, Ruiz C, Aguilera C, Andrés ME, Fuentealba JA. The blocking of kappa‐opioid receptor reverses the changes in dorsolateral striatum dopamine dynamics during the amphetamine sensitization. J Neurochem 2018; 148:348-358. [DOI: 10.1111/jnc.14612] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/25/2018] [Accepted: 10/05/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Victor Hugo Azocar
- Department of Pharmacy and Interdisciplinary Center of Neuroscience Pontificia Universidad Catolica de Chile Santiago Chile
| | - Gladys Sepúlveda
- Department of Pharmacy and Interdisciplinary Center of Neuroscience Pontificia Universidad Catolica de Chile Santiago Chile
| | - Catalina Ruiz
- Department of Pharmacy and Interdisciplinary Center of Neuroscience Pontificia Universidad Catolica de Chile Santiago Chile
| | - Consuelo Aguilera
- Department of Pharmacy and Interdisciplinary Center of Neuroscience Pontificia Universidad Catolica de Chile Santiago Chile
| | - Maria Estela Andrés
- Department of Cellular and Molecular Biology Faculty of Biological Science Pontificia Universidad Católica de Chile Santiago Chile
| | - José Antonio Fuentealba
- Department of Pharmacy and Interdisciplinary Center of Neuroscience Pontificia Universidad Catolica de Chile Santiago Chile
| |
Collapse
|
47
|
Age as a factor in stress and alcohol interactions: A critical role for the kappa opioid system. Alcohol 2018; 72:9-18. [PMID: 30322483 DOI: 10.1016/j.alcohol.2017.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/27/2017] [Accepted: 10/06/2017] [Indexed: 12/18/2022]
Abstract
The endogenous kappa opioid system has primarily been shown to be involved with a state of dysphoria and aversion. Stress and exposure to drugs of abuse, particularly alcohol, can produce similar states of unease and anxiety, implicating the kappa opioid system as a target of stress and alcohol. Numerous behavioral studies have demonstrated reduced sensitivity to manipulations of the kappa opioid system in early life relative to adulthood, and recent reports have shown that the kappa opioid system is functionally different across ontogeny. Given the global rise in early-life stress and alcohol consumption, understanding how the kappa opioid system responds and adapts to stress and/or alcohol exposure differently in early life and adulthood is imperative. Therefore, the objective of this review is to highlight and discuss studies examining the impact of early-life stress and/or alcohol on the kappa opioid system, with focus on the documented neuroadaptations that may contribute to future vulnerability to stress and/or increase the risk of relapse. We first provide a brief summary of the importance of studying the effects of stress and alcohol during early life (prenatal, neonatal/juvenile, and adolescence). We then discuss the literature on the effects of stress or alcohol during early life and adulthood on the kappa opioid system. Finally, we discuss the few studies that have shown interactions between stress and alcohol on the kappa opioid system and provide some discussion about the need for studies investigating the development of the kappa opioid system.
Collapse
|
48
|
Williams AV, Laman-Maharg A, Armstrong CV, Ramos-Maciel S, Minie VA, Trainor BC. Acute inhibition of kappa opioid receptors before stress blocks depression-like behaviors in California mice. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:166-174. [PMID: 29879438 PMCID: PMC6054130 DOI: 10.1016/j.pnpbp.2018.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/02/2018] [Accepted: 06/02/2018] [Indexed: 11/30/2022]
Abstract
Kappa opioid receptors (KOR) are considered to be a promising therapeutic target for stress-induced psychiatric disorders such as anxiety and depression. Preclinical data show that KOR antagonists have greater efficacy if administered before stressful experiences as opposed to afterwards. However, almost all of these studies use long-acting antagonists, leaving it unclear whether inhibition of KOR after stress is required for efficacy. Here we show that administration of the short-acting KOR antagonist AZ-MTAB before episodes of social defeat stress block the induction of anhedonia (both males and females) and social avoidance responses (females) that persist two weeks after stress. In both males and females pre-stress AZ-MTAB treatment also blunted anticipatory autogrooming behavior immediately prior to the third episode of defeat. In contrast when AZ-MTAB was administered two weeks after defeat (immediately before behavior testing) in female California mice, it was ineffective at reversing anhedonia and social avoidance. These results suggest that short-acting KOR antagonists may have greater therapeutic potential if administered before exposure to psychosocial stressors.
Collapse
Affiliation(s)
- Alexia V Williams
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Abigail Laman-Maharg
- Neuroscience Graduate Group, University of California, Davis, Davis, CA, United States
| | - Crystal V Armstrong
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | | | - Vanessa A Minie
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Brian C Trainor
- Department of Psychology, University of California, Davis, Davis, CA, United States; Neuroscience Graduate Group, University of California, Davis, Davis, CA, United States.
| |
Collapse
|
49
|
Ondachi PW, Kormos CM, Runyon SP, Thomas JB, Mascarella SW, Decker AM, Navarro HA, Fennell TR, Snyder RW, Carroll FI. Potent and Selective Tetrahydroisoquinoline Kappa Opioid Receptor Antagonists of Lead Compound (3 R)-7-Hydroxy- N-[(1 S)-2-methyl-1-(piperidin-1-ylmethyl)propyl]-1,2,3,4-tetrahydroisoquinoline-3-carboxamide (PDTic). J Med Chem 2018; 61:7525-7545. [PMID: 30117738 DOI: 10.1021/acs.jmedchem.8b00673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Past studies have shown that it has been difficult to discover and develop potent and selective κ opioid receptor antagonists, particularly compounds having potential for clinical development. In this study, we present a structure-activity relationship (SAR) study of a recently discovered new class of tetrahydroisoquinoline κ opioid receptor antagonists which led to (3 R)-7-hydroxy- N-{(1 S)-2-methyl-1-[(-4-methylpiperidine-1-yl)methyl]propyl}-1,2,3,4-tetrahydroisoquinoline-3-carboxamide (12) (4-Me-PDTic). Compound 12 had a Ke = 0.37 nM in a [35S]GTPγS binding assay and was 645- and >8100-fold selective for the κ relative to the μ and δ opioid receptors, respectively. Calculated log BB and CNS (central nervous system) multiparameter optimization (MPO) and low molecular weight values all predict that 12 will penetrate the brain, and pharmacokinetic studies in rats show that 12 does indeed penetrate the brain.
Collapse
Affiliation(s)
- Pauline W Ondachi
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Chad M Kormos
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Scott P Runyon
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - James B Thomas
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - S Wayne Mascarella
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Ann M Decker
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Hernán A Navarro
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Timothy R Fennell
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Rodney W Snyder
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - F Ivy Carroll
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| |
Collapse
|
50
|
Kormos CM, Ondachi PW, Runyon SP, Thomas JB, Mascarella SW, Decker AM, Navarro HA, Fennell TR, Snyder RW, Carroll FI. Potent and Selective Tetrahydroisoquinoline Kappa Opioid Receptor Antagonists of Lead Compound (3 R)- N-[1 R)-1-(Cyclohexylmethyl)-2-methylpropyl]-7-hydroxy-1,2,3,4-tetrahydroisoquinoline-3-carboxamide (CDTic). J Med Chem 2018; 61:7546-7559. [PMID: 30032602 DOI: 10.1021/acs.jmedchem.8b00674] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Animal pharmacological studies suggest that potent and selective κ opioid receptor antagonists have potential as pharmacotherapies targeting depression, anxiety, and substance abuse (opiates, alcohol, nicotine, cocaine). We recently reported lead compound 1 as a new class of κ opioid receptor antagonists with only one basic amine group. Analogues were synthesized and evaluated for their in vitro opioid receptor antagonist properties using a [35S]GTPγS binding assay. All analogues were pure opioid receptor antagonists with no agonist activity. Compounds 1, 8, 9, 13, and 14 ( Ke values 0.058-0.64 nM) are highly potent and highly selective for the κ relative to the μ and δ opioid receptors. Favorable calculated physiochemical properties were confirmed in rat PK studies, demonstrating brain penetration for selected compounds 1, 9, and 13. High κ opioid receptor potency and selectivity and highly favorable calculated physiochemical and PK properties for brain penetration suggest these compounds should be considered for further development.
Collapse
Affiliation(s)
- Chad M Kormos
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Pauline W Ondachi
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Scott P Runyon
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - James B Thomas
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - S Wayne Mascarella
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Ann M Decker
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Hernán A Navarro
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Timothy R Fennell
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - Rodney W Snyder
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| | - F Ivy Carroll
- Research Triangle Institute , P.O. Box 12194, Research Triangle Park , North Carolina 27709-2194 , United States
| |
Collapse
|