1
|
Mayer FP, Stewart A, Varman DR, Moritz AE, Foster JD, Owens AW, Areal LB, Gowrishankar R, Velez M, Wickham K, Phelps H, Katamish R, Rabil M, Jayanthi LD, Vaughan RA, Daws LC, Blakely RD, Ramamoorthy S. Kappa Opioid Receptor Antagonism Restores Phosphorylation, Trafficking and Behavior induced by a Disease Associated Dopamine Transporter Variant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.03.539310. [PMID: 37205452 PMCID: PMC10187322 DOI: 10.1101/2023.05.03.539310] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Aberrant dopamine (DA) signaling is implicated in schizophrenia, bipolar disorder (BPD), autism spectrum disorder (ASD), substance use disorder, and attention-deficit/hyperactivity disorder (ADHD). Treatment of these disorders remains inadequate, as exemplified by the therapeutic use of d-amphetamine and methylphenidate for the treatment of ADHD, agents with high abuse liability. In search for an improved and non-addictive therapeutic approach for the treatment of DA-linked disorders, we utilized a preclinical mouse model expressing the human DA transporter (DAT) coding variant DAT Val559, previously identified in individuals with ADHD, ASD, or BPD. DAT Val559, like several other disease-associated variants of DAT, exhibits anomalous DA efflux (ADE) that can be blocked by d-amphetamine and methylphenidate. Kappa opioid receptors (KORs) are expressed by DA neurons and modulate DA release and clearance, suggesting that targeting KORs might also provide an alternative approach to normalizing DA-signaling disrupted by perturbed DAT function. Here we demonstrate that KOR stimulation leads to enhanced surface trafficking and phosphorylation of Thr53 in wildtype DAT, effects achieved constitutively by the Val559 mutant. Moreover, these effects can be rescued by KOR antagonism of DAT Val559 in ex vivo preparations. Importantly, KOR antagonism also corrected in vivo DA release as well as sex-dependent behavioral abnormalities observed in DAT Val559 mice. Given their low abuse liability, our studies with a construct valid model of human DA associated disorders reinforce considerations of KOR antagonism as a pharmacological strategy to treat DA associated brain disorders.
Collapse
Affiliation(s)
- Felix P. Mayer
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy E. Moritz
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - James D. Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Anthony W. Owens
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA
| | - Lorena B. Areal
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Raajaram Gowrishankar
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Michelle Velez
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Kyria Wickham
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Hannah Phelps
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Rania Katamish
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Maximilian Rabil
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Lankupalle D. Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Roxanne A. Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Lynette C. Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
2
|
Estave PM, Albertson SE, Karkhanis AN, Jones SR. Co-targeting the kappa opioid receptor and dopamine transporter reduces motivation to self-administer cocaine and partially reverses dopamine system dysregulation. Sci Rep 2024; 14:6509. [PMID: 38499566 PMCID: PMC10948819 DOI: 10.1038/s41598-024-53463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/31/2024] [Indexed: 03/20/2024] Open
Abstract
Cocaine disrupts dopamine (DA) and kappa opioid receptor (KOR) system activity, with long-term exposure reducing inhibiton of DA uptake by cocaine and increasing KOR system function. Single treatment therapies have not been successful for cocaine use disorder; therefore, this study focuses on a combination therapy targeting the dopamine transporter (DAT) and KOR. Sprague Dawley rats self-administered 5 days of cocaine (1.5 mg/kg/inf, max 40 inf/day, FR1), followed by 14 days on a progressive ratio (PR) schedule (0.19 mg/kg/infusion). Behavioral effects of individual and combined administration of phenmetrazine and nBNI were then examined using PR. Additionally, ex vivo fast scan cyclic voltammetry was then used to assess alterations in DA and KOR system activity in the nucleus accumbens before and after treatments. Chronic administration of phenmetrazine as well as the combination of phenmetrazine and nBNI-but not nBNI alone-significantly reduced PR breakpoints. In addition, the combination of phenmetrazine and nBNI partially reversed cocaine-induced neurodysregulations of the KOR and DA systems, indicating therapeutic benefits of targeting the DA and KOR systems in tandem. These data highlight the potential benefits of the DAT and KOR as dual-cellular targets to reduce motivation to administer cocaine and reverse cocaine-induced alterations of the DA system.
Collapse
Affiliation(s)
- Paige M Estave
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA
| | - Steven E Albertson
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA
| | - Anushree N Karkhanis
- Department of Psychology, Binghamton University - State University of New York, Binghamton, NY, 13902, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA.
| |
Collapse
|
3
|
Farahbakhsh ZZ, Holleran KM, Sens JP, Fordahl SC, Mauterer MI, López AJ, Cuzon Carlson VC, Kiraly DD, Grant KA, Jones SR, Siciliano CA. Synchrony between midbrain gene transcription and dopamine terminal regulation is modulated by chronic alcohol drinking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.584711. [PMID: 38559169 PMCID: PMC10979957 DOI: 10.1101/2024.03.15.584711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Alcohol use disorder is marked by disrupted behavioral and emotional states which persist into abstinence. The enduring synaptic alterations that remain despite the absence of alcohol are of interest for interventions to prevent relapse. Here, 28 male rhesus macaques underwent over 20 months of alcohol drinking interspersed with three 30-day forced abstinence periods. After the last abstinence period, we paired direct sub-second dopamine monitoring via ex vivo voltammetry in nucleus accumbens slices with RNA-sequencing of the ventral tegmental area. We found persistent augmentation of dopamine transporter function, kappa opioid receptor sensitivity, and dynorphin release - all inhibitory regulators which act to decrease extracellular dopamine. Surprisingly, though transcript expression was not altered, the relationship between gene expression and functional readouts of these encoded proteins was highly dynamic and altered by drinking history. These results outline the long-lasting synaptic impact of alcohol use and suggest that assessment of transcript-function relationships is critical for the rational design of precision therapeutics.
Collapse
|
4
|
Gordon-Fennell L, Farero R, Burgeno L, Murray N, Abraham A, Soden M, Stuber G, Chavkin C, Zweifel L, Phillips P. Kappa Opioid Receptors in Mesolimbic Terminals Mediate Escalation of Cocaine Consumption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572842. [PMID: 38187718 PMCID: PMC10769440 DOI: 10.1101/2023.12.21.572842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Increases in drug consumption over time, also known as escalation, is a key behavioral component of substance use disorder (SUD) that is related to potential harm to users, such as overdose. Studying escalation also allows researchers to investigate the transition from casual drug use to more SUD-like drug use. Understanding the neurobiological systems that drive this transition will inform therapeutic treatments in the aim to prevent increases in drug use and the development of SUD. The kappa opioid receptor (KOR) system is typically known for its role in negative affect, which is commonly found in SUD as well. Furthermore, the KOR system has also been implicated in drug use and importantly, modulating the negative effects of drug use. However, the specific neuronal subpopulation expressing KOR involved has not been identified. Here, we first demonstrated that pharmacologically inhibiting KOR in the nucleus accumbens core (NAcC), as a whole, blocks cocaine escalation under long-access self-administration conditions. We then demonstrated that KOR expressed on ventral tegmental area (VTA) neurons but not NAcC neurons is sufficient for blocking cocaine escalation by utilizing a novel virally-mediated CRISPR-SaCas9 knock-out of the oprk1 gene. Together, this suggests that activation of KOR on VTA terminals in the NAcC drives the transition to the SUD-like phenotype of escalation of cocaine consumption.
Collapse
Affiliation(s)
- L. Gordon-Fennell
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Psychiatry & Behavioral Science, University of Washington, Seattle, WA 98195
| | - R.D. Farero
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Psychiatry & Behavioral Science, University of Washington, Seattle, WA 98195
| | - L.M. Burgeno
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Psychiatry & Behavioral Science, University of Washington, Seattle, WA 98195
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - N.L. Murray
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Psychiatry & Behavioral Science, University of Washington, Seattle, WA 98195
| | - A.D. Abraham
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - M.E. Soden
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - G.D. Stuber
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Psychiatry & Behavioral Science, University of Washington, Seattle, WA 98195
- Department of Pharmacology, University of Washington, Seattle, WA 98195
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA 98195
| | - C. Chavkin
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - L.S. Zweifel
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Psychiatry & Behavioral Science, University of Washington, Seattle, WA 98195
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - P.E.M. Phillips
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195
- Center for Neurobiology of Addiction, Pain & Emotion, University of Washington, Seattle, WA 98195
- Department of Psychiatry & Behavioral Science, University of Washington, Seattle, WA 98195
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| |
Collapse
|
5
|
Karkhanis AN, West AM, Jones SR. Kappa opioid receptor agonist U50,488 inhibits dopamine more in caudal than rostral nucleus accumbens core. Basic Clin Pharmacol Toxicol 2023; 133:526-534. [PMID: 37539456 DOI: 10.1111/bcpt.13929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/07/2023] [Accepted: 07/14/2023] [Indexed: 08/05/2023]
Abstract
The nucleus accumbens (NAc) core is involved in regulating stress and shaping reward seeking behaviours. Multiple neuromodulators, including dynorphin/kappa opioid receptor (KOR) and dopamine systems, converge in this area to influence behavioural outcomes. KOR activation acutely inhibits dopamine release and chronically depresses overall dopamine transmission. Recently, studies in the NAc shell have revealed that the impact of KOR activation on behaviour is regionally specific, and these rostro-caudal differences are likely driven by greater control of KORs over dopamine inhibition in the caudal compared with rostral subregion. Given the importance of NAc core, particularly the interaction between KORs and dopamine in regulating reward seeking behaviours, we examined the impact of KOR activation on dopamine release and uptake along the rostro-caudal axis in the NAc core of male and female mice. Using ex vivo fast scan cyclic voltammetry, we observed that KOR mediated inhibition of dopamine release was significantly greater in caudal compared with rostral NAc core with no significant sex differences observed. These data suggest that KORs regulate dopamine release differentially along the rostro-caudal axis, providing a new axis on which to examine the process by which the KOR/dopamine system controls reward encoding.
Collapse
Affiliation(s)
| | - Alyssa M West
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
6
|
Ide S, Hirai T, Muto T, Yamakawa T, Ikeda K. Effects of the novel selective κ-opioid receptor agonist NP-5497-KA on morphine-induced reward-related behaviors. Sci Rep 2023; 13:18164. [PMID: 37875567 PMCID: PMC10598265 DOI: 10.1038/s41598-023-45584-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/21/2023] [Indexed: 10/26/2023] Open
Abstract
Opioid addiction and the opioid overdose epidemic are becoming more serious, and the development of therapeutic agents is essential for the pharmacological treatment of substance use disorders. The κ-opioid receptor (KOP) is a member of the opioid receptor system that has been gaining attention as a promising molecular target for the treatment of numerous human disorders, including pain, depression, anxiety, and drug addiction. Here, we biologically and pharmacologically evaluated a novel azepane-derived ligand, NP-5497-KA, as a selective KOP agonist. NP-5497-KA had 1000-fold higher selectivity for the KOP over the μ-opioid receptor (MOP), which was higher than nalfurafine (KOP/MOP: 65-fold), and acted as a selective KOP full agonist in the 3',5'-cyclic adenosine monophosphate assay. The oral administration of NP-5497-KA (1-10 mg/kg) dose-dependently suppressed morphine-induced conditioned place preference in C57BL/6 J mice, and its effects were comparable to an intraperitoneal injection of nalfurafine (1-10 μg/kg). Nalfurafine (10 μg/kg) significantly inhibited rotarod performance, whereas NP-5497-KA (10 mg/kg) exerted no effect on rotarod performance. These results indicate that NP-5497-KA may be a novel option for the treatment of opioid use disorder with fewer side effects.
Collapse
Affiliation(s)
- Soichiro Ide
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
| | - Toshitake Hirai
- Discovery Research Laboratories, Nippon Chemiphar Co., Ltd., 1-22 Hikokawado, Misato, Saitama, 341-0005, Japan
| | - Takafumi Muto
- Discovery Research Laboratories, Nippon Chemiphar Co., Ltd., 1-22 Hikokawado, Misato, Saitama, 341-0005, Japan
| | - Tomio Yamakawa
- Discovery Research Laboratories, Nippon Chemiphar Co., Ltd., 1-22 Hikokawado, Misato, Saitama, 341-0005, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| |
Collapse
|
7
|
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
|
8
|
Yang R, Tuan RRL, Hwang FJ, Bloodgood DW, Kong D, Ding JB. Dichotomous regulation of striatal plasticity by dynorphin. Mol Psychiatry 2023; 28:434-447. [PMID: 36460726 PMCID: PMC10188294 DOI: 10.1038/s41380-022-01885-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022]
Abstract
Modulation of corticostriatal plasticity alters the information flow throughout basal ganglia circuits and represents a fundamental mechanism for motor learning, action selection, and reward. Synaptic plasticity in the striatal direct- and indirect-pathway spiny projection neurons (dSPNs and iSPNs) is regulated by two distinct networks of GPCR signaling cascades. While it is well-known that dopamine D2 and adenosine A2a receptors bi-directionally regulate iSPN plasticity, it remains unclear how D1 signaling modulation of synaptic plasticity is counteracted by dSPN-specific Gi signaling. Here, we show that striatal dynorphin selectively suppresses long-term potentiation (LTP) through Kappa Opioid Receptor (KOR) signaling in dSPNs. Both KOR antagonism and conditional deletion of dynorphin in dSPNs enhance LTP counterbalancing with different levels of D1 receptor activation. Behaviorally, mice lacking dynorphin in D1 neurons show comparable motor behavior and reward-based learning, but enhanced flexibility during reversal learning. These findings support a model in which D1R and KOR signaling bi-directionally modulate synaptic plasticity and behavior in the direct pathway.
Collapse
Affiliation(s)
- Renzhi Yang
- Biology Graduate Program, Stanford University, Stanford, CA, USA
| | - Rupa R Lalchandani Tuan
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
| | - Fuu-Jiun Hwang
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | | | - Dong Kong
- Division of Endocrinology, Department of Pediatrics, F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Stanford Bio-X, Stanford University, Stanford, CA, USA.
| |
Collapse
|
9
|
Pomrenze MB, Cardozo Pinto DF, Neumann PA, Llorach P, Tucciarone JM, Morishita W, Eshel N, Heifets BD, Malenka RC. Modulation of 5-HT release by dynorphin mediates social deficits during opioid withdrawal. Neuron 2022; 110:4125-4143.e6. [PMID: 36202097 PMCID: PMC9789200 DOI: 10.1016/j.neuron.2022.09.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/25/2022] [Accepted: 09/19/2022] [Indexed: 12/24/2022]
Abstract
Social isolation during opioid withdrawal is a major contributor to the current opioid addiction crisis. We find that sociability deficits during protracted opioid withdrawal in mice require activation of kappa opioid receptors (KORs) in the nucleus accumbens (NAc) medial shell. Blockade of release from dynorphin (Pdyn)-expressing dorsal raphe neurons (DRPdyn), but not from NAcPdyn neurons, prevents these deficits in prosocial behaviors. Conversely, optogenetic activation of DRPdyn neurons reproduced NAc KOR-dependent decreases in sociability. Deletion of KORs from serotonin (5-HT) neurons, but not from NAc neurons or dopamine (DA) neurons, prevented sociability deficits during withdrawal. Finally, measurements with the genetically encoded GRAB5-HT sensor revealed that during withdrawal KORs block the NAc 5-HT release that normally occurs during social interactions. These results define a neuromodulatory mechanism that is engaged during protracted opioid withdrawal to induce maladaptive deficits in prosocial behaviors, which in humans contribute to relapse.
Collapse
Affiliation(s)
- Matthew B Pomrenze
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Daniel F Cardozo Pinto
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Peter A Neumann
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Pierre Llorach
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jason M Tucciarone
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Wade Morishita
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Neir Eshel
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
10
|
Escobar AP, Meza R, Gonzalez M, Henny P, Andrés ME. Immunolocalization of kappa opioid receptors in the axon initial segment of a group of embryonic mesencephalic dopamine neurons. IBRO Neurosci Rep 2022; 12:411-418. [PMID: 35746971 PMCID: PMC9210487 DOI: 10.1016/j.ibneur.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 11/13/2022] Open
Abstract
The dopamine mesolimbic system is a major circuit involved in controlling goal-directed behaviors. Dopamine D2 receptors (D2R) and kappa opioid receptors (KOR) are abundant Gi protein-coupled receptors in the mesolimbic system. D2R and KOR share several functions in dopamine mesencephalic neurons, such as regulation of dopamine release and uptake, and firing of dopamine neurons. In addition, KOR and D2R modulate each other functioning. This evidence indicates that both receptors functionally interact, however, their colocalization in the mesostriatal system has not been addressed. Immunofluorescent assays were performed in cultured dopamine neurons and adult mice’s brain tissue to answer this question. We observed that KOR and D2R are present in similar density in dendrites and soma of cultured dopamine neurons, but in a segregated manner. Interestingly, KOR immunolabelling was observed in the first part of the axon, colocalizing with Ankyrin in 20% of cultured dopamine neurons, indicative that KOR is present in the axon initial segment (AIS) of a group of dopaminergic neurons. In the adult brain, KOR and D2R are also segregated in striatal tissue. While the KOR label is in fiber tracts such as the striatal streaks, corpus callosum, and anterior commissure, D2R is located mainly within the striatum and nucleus accumbens, surrounding fiber tracts. D2R is also localized in some fibers that are mostly different from those positives for KOR. In conclusion, KOR and D2R are present in the soma and dendrites of mesencephalic dopaminergic neurons, but KOR is also found in the AIS of a subpopulation of these neurons. KOR and D2R localize in cell bodies of primary cultured TH neurons. In primary cultured TH neurons KOR localizes in axon initial segment. KOR and D2R co-localize in cell bodies of the CPu and NAc.
Collapse
|
11
|
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
|
12
|
Shokri-Kojori E, Naganawa M, Ramchandani VA, Wong DF, Wang GJ, Volkow ND. Brain opioid segments and striatal patterns of dopamine release induced by naloxone and morphine. Hum Brain Mapp 2021; 43:1419-1430. [PMID: 34873784 PMCID: PMC8837588 DOI: 10.1002/hbm.25733] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/10/2021] [Accepted: 11/19/2021] [Indexed: 11/25/2022] Open
Abstract
Opioid receptors are expressed throughout the brain and play a major role in regulating striatal dopamine (DA) release. Clinical studies have shown that naloxone (NAL, a nonspecific opioid antagonist) in individuals with opioid use disorder and morphine (MRP, a nonspecific opioid agonist) in healthy controls, resulted in DA release in the dorsal and ventral striatum, respectively. It is not known whether the underlying patterns of striatal DA release are associated with the striatal distribution of opioid receptors. We leveraged previously published PET datasets (collected in independent cohorts) to study the brain‐wide distribution of opioid receptors and to compare striatal opioid receptor availability with striatal DA release patterns. We identified three major gray matter segments based on availability maps of DA and opioid receptors: striatum, and primary and secondary opioid segments with high and intermediate opioid receptor availability, respectively. Patterns of DA release induced by NAL and MRP were inversely associated and correlated with kappa (NAL: r(68) = −0.81, MRP: r(68) = 0.54), and mu (NAL: r(68) = −0.62, MRP: r(68) = 0.46) opioid receptor availability. Kappa opioid receptor availability accounted for a unique part of variance in NAL‐ and MRP‐DA release patterns (ΔR2 >0.14, p <.0001). In sum, distributions of opioid receptors distinguished major cortical and subcortical regions. Patterns of NAL‐ and MRP‐induced DA release had inverse associations with striatal opioid receptor availability. Our approach provides a pattern‐based characterization of drug‐induced DA targets and is relevant for modeling the role of opioid receptors in modulating striatal DA release.
Collapse
Affiliation(s)
- Ehsan Shokri-Kojori
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Mika Naganawa
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Vijay A Ramchandani
- Human Psychopharmacology Laboratory, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Dean F Wong
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Nora D Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Coleman BC, Manz KM, Grueter BA. Kappa opioid receptor modulation of excitatory drive onto nucleus accumbens fast-spiking interneurons. Neuropsychopharmacology 2021; 46:2340-2349. [PMID: 34400782 PMCID: PMC8581025 DOI: 10.1038/s41386-021-01146-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023]
Abstract
The dynorphin/kappa opioid receptor (KOR) system within the nucleus accumbens (NAc) contributes to affective states. Parvalbumin fast-spiking interneurons (PV-FSIs), a key component of feedforward inhibition, participate in integration of excitatory inputs to the NAc by robustly inhibiting select populations of medium spiny output neurons, therefore greatly influencing NAc dependent behavior. How the dynorphin/KOR system regulates feedforward inhibition in the NAc remains unknown. Here, we elucidate the molecular mechanisms of KOR inhibition of excitatory transmission onto NAc PV-FSIs using a combination of whole-cell patch-clamp electrophysiology, optogenetics, pharmacology, and a parvalbumin reporter mouse. We find that postsynaptic KOR stimulation induces long-term depression (LTD) of excitatory synapses onto PV-FSI by stimulating the endocytosis of AMPARs via a PKA and calcineurin-dependent mechanism. Furthermore, KOR regulation of PV-FSI synapses are input specific, inhibiting thalamic but not cortical inputs. Finally, following acute stress, a protocol known to elevate dynorphin/KOR signaling in the NAc, KOR agonists no longer inhibit excitatory transmission onto PV-FSI. In conclusion, we delineate pathway-specific mechanisms mediating KOR control of feedforward inhibitory circuits in the NAc and provide evidence for the recruitment of this system in response to stress.
Collapse
Affiliation(s)
| | - Kevin M Manz
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Brad A Grueter
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
14
|
Covey DP, Yocky AG. Endocannabinoid Modulation of Nucleus Accumbens Microcircuitry and Terminal Dopamine Release. Front Synaptic Neurosci 2021; 13:734975. [PMID: 34497503 PMCID: PMC8419321 DOI: 10.3389/fnsyn.2021.734975] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/05/2021] [Indexed: 12/20/2022] Open
Abstract
The nucleus accumbens (NAc) is located in the ventromedial portion of the striatum and is vital to valence-based predictions and motivated action. The neural architecture of the NAc allows for complex interactions between various cell types that filter incoming and outgoing information. Dopamine (DA) input serves a crucial role in modulating NAc function, but the mechanisms that control terminal DA release and its effect on NAc neurons continues to be elucidated. The endocannabinoid (eCB) system has emerged as an important filter of neural circuitry within the NAc that locally shapes terminal DA release through various cell type- and site-specific actions. Here, we will discuss how eCB signaling modulates terminal DA release by shaping the activity patterns of NAc neurons and their afferent inputs. We then discuss recent technological advancements that are capable of dissecting how distinct cell types, their afferent projections, and local neuromodulators influence valence-based actions.
Collapse
Affiliation(s)
- Dan P Covey
- Department of Neuroscience, Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Alyssa G Yocky
- Department of Neuroscience, Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| |
Collapse
|
15
|
Replacing a Palatable High-Fat Diet with a Low-Fat Alternative Heightens κ-Opioid Receptor Control over Nucleus Accumbens Dopamine. Nutrients 2021; 13:nu13072341. [PMID: 34371851 PMCID: PMC8308677 DOI: 10.3390/nu13072341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/22/2021] [Accepted: 07/06/2021] [Indexed: 12/04/2022] Open
Abstract
Diet-induced obesity reduces dopaminergic neurotransmission in the nucleus accumbens (NAc), and stressful weight loss interventions could promote cravings for palatable foods high in fat and sugar that stimulate dopamine. Activation of κ-opioid receptors (KORs) reduces synaptic dopamine, but contribution of KORs to lower dopamine tone after dietary changes is unknown. Therefore, the purpose of this study was to determine the function of KORs in C57BL/6 mice that consumed a 60% high-fat diet (HFD) for six weeks followed by replacement of HFD with a control 10% fat diet for one day or one week. HFD replacement induced voluntary caloric restriction and weight loss. However, fast-scan cyclic voltammetry revealed no differences in baseline dopamine parameters, whereas sex effects were revealed during KOR stimulation. NAc core dopamine release was reduced by KOR agonism after one day of HFD replacement in females but after one week of HFD replacement in males. Further, elevated plus-maze testing revealed no diet effects during HFD replacement on overt anxiety. These results suggest that KORs reduce NAc dopamine tone and increase food-related anxiety during dietary weight loss interventions that could subsequently promote palatable food cravings and inhibit weight loss.
Collapse
|
16
|
Obesity and dietary fat influence dopamine neurotransmission: exploring the convergence of metabolic state, physiological stress, and inflammation on dopaminergic control of food intake. Nutr Res Rev 2021; 35:236-251. [PMID: 34184629 DOI: 10.1017/s0954422421000196] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aim of this review is to explore how metabolic changes induced by diets high in saturated fat (HFD) affect nucleus accumbens (NAc) dopamine neurotransmission and food intake, and to explore how stress and inflammation influence this process. Recent evidence linked diet-induced obesity and HFD with reduced dopamine release and reuptake. Altered dopamine neurotransmission could disrupt satiety circuits between NAc dopamine terminals and projections to the hypothalamus. The NAc directs learning and motivated behaviours based on homeostatic needs and psychological states. Therefore, impaired dopaminergic responses to palatable food could contribute to weight gain by disrupting responses to food cues or stress, which impacts type and quantity of food consumed. Specifically, saturated fat promotes neuronal resistance to anorectic hormones and activation of immune cells that release proinflammatory cytokines. Insulin has been shown to regulate dopamine neurotransmission by enhancing satiety, but less is known about effects of diet-induced stress. Therefore, changes to dopamine signalling due to HFD warrant further examination to characterise crosstalk of cytokines with endocrine and neurotransmitter signals. A HFD promotes a proinflammatory environment that may disrupt neuronal endocrine function and dopamine signalling that could be exacerbated by the hypothalamic-pituitary-adrenal and κ-opioid receptor stress systems. Together, these adaptive changes may dysregulate eating by changing NAc dopamine during hedonic versus homeostatic food intake. This could drive palatable food cravings during energy restriction and hinder weight loss. Understanding links between HFD and dopamine neurotransmission will inform treatment strategies for diet-induced obesity and identify molecular candidates for targeted therapeutics.
Collapse
|
17
|
Kappa Opioid Receptor Mediated Differential Regulation of Serotonin and Dopamine Transporters in Mood and Substance Use Disorder. Handb Exp Pharmacol 2021; 271:97-112. [PMID: 34136961 DOI: 10.1007/164_2021_499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Dynorphin (DYN) is an endogenous neurosecretory peptide which exerts its activity by binding to the family of G protein-coupled receptors, namely the kappa opioid receptor (KOR). Opioids are associated with pain, analgesia, and drug abuse, which play a central role in mood disorders with monoamine neurotransmitter interactions. Growing evidence demonstrates the cellular signaling cascades linked to KOR-mediated monoamine transporters regulation in cell models and native brain tissues. This chapter will review DYN/KOR role in mood and addiction in relevance to dopaminergic and serotonergic neurotransmissions. Also, we discuss the recent findings on KOR-mediated differential regulation of serotonin and dopamine transporters (SERT and DAT). These findings led to a better understanding of the role of DYN/KOR system in aminergic neurotransmission via its modulatory effect on both amine release and clearance. Detailed knowledge of these processes at the molecular level enables designing novel pharmacological reagents to target transporter motifs to treat mood and addiction and reduce unwanted side effects such as aversion, dysphoria, sedation, and psychomimesis.
Collapse
|
18
|
Bidirectional role of acupuncture in the treatment of drug addiction. Neurosci Biobehav Rev 2021; 126:382-397. [PMID: 33839169 DOI: 10.1016/j.neubiorev.2021.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
Drug addiction is a chronically relapsing disorder, affecting people from all walks of life. Studies of acupuncture effects on drug addiction are intriguing in light of the fact that acupuncture can be used as a convenient therapeutic intervention for treating drug addiction by direct activation of brain pathway. The current review aims to discuss the neurobiological mechanisms underlying acupuncture's effectiveness in the treatment of drug addiction, on the basis of two different theories (the incentive sensitization theory and the opponent process theory) that have seemingly opposite view on the role of the mesolimbic reward pathways in mediating compulsive drug-seeking behavior. This review provides evidence that acupuncture may reduce relapse to drug-seeking behavior by regulating neurotransmitters involved in drug craving modulation via somatosensory afferent mechanisms. Also, acupuncture normalizes hyper-reactivity or hypoactivity of the mesolimbic dopamine system in these opposed processes in drug addiction, suggesting bidirectional role of acupuncture in regulation of drug addiction. This proposes that acupuncture may reduce drug craving by correcting both dysfunctions of the mesolimbic dopamine pathway.
Collapse
|
19
|
Zachry JE, Nolan SO, Brady LJ, Kelly SJ, Siciliano CA, Calipari ES. Sex differences in dopamine release regulation in the striatum. Neuropsychopharmacology 2021; 46:491-499. [PMID: 33318634 PMCID: PMC8027008 DOI: 10.1038/s41386-020-00915-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 01/05/2023]
Abstract
The mesolimbic dopamine system-which originates in the ventral tegmental area and projects to the striatum-has been shown to be involved in the expression of sex-specific behavior and is thought to be a critical mediator of many psychiatric diseases. While substantial work has focused on sex differences in the anatomy of dopamine neurons and relative dopamine levels between males and females, an important characteristic of dopamine release from axon terminals in the striatum is that it is rapidly modulated by local regulatory mechanisms independent of somatic activity. These processes can occur via homosynaptic mechanisms-such as presynaptic dopamine autoreceptors and dopamine transporters-as well as heterosynaptic mechanisms, such as retrograde signaling from postsynaptic cholinergic and GABAergic systems, among others. These regulators serve as potential targets for the expression of sex differences in dopamine regulation in both ovarian hormone-dependent and independent fashions. This review describes how sex differences in microcircuit regulatory mechanisms can alter dopamine dynamics between males and females. We then describe what is known about the hormonal mechanisms controlling/regulating these processes. Finally, we highlight the missing gaps in our knowledge of these systems in females. Together, a more comprehensive and mechanistic understanding of how sex differences in dopamine function manifest will be particularly important in developing evidence-based therapeutics that target this system and show efficacy in both sexes.
Collapse
Affiliation(s)
- Jennifer E. Zachry
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
| | - Suzanne O. Nolan
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
| | - Lillian J. Brady
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
| | - Shannon J. Kelly
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA
| | - Cody A. Siciliano
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232 USA
| | - Erin S. Calipari
- grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN 37232 USA
| |
Collapse
|
20
|
Carlson HN, Weiner JL. The neural, behavioral, and epidemiological underpinnings of comorbid alcohol use disorder and post-traumatic stress disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:69-142. [PMID: 33648676 DOI: 10.1016/bs.irn.2020.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alcohol use disorder (AUD) and (PTSD) frequently co-occur and individuals suffering from this dual diagnosis often exhibit increased symptom severity and poorer treatment outcomes than those with only one of these diseases. Although there have been significant advances in our understanding of the neurobiological mechanisms underlying each of these disorders, the neural underpinnings of the comorbid condition remain poorly understood. This chapter summarizes recent epidemiological findings on comorbid AUD and PTSD, with a focus on vulnerable populations, the temporal relationship between these disorders, and the clinical consequences associated with the dual diagnosis. We then review animal models of the comorbid condition and emerging human and non-human animal research that is beginning to identify maladaptive neural changes common to both disorders, primarily involving functional changes in brain reward and stress networks. We end by proposing a neural framework, based on the emerging field of affective valence encoding, that may better explain the epidemiological and neural findings on AUD and PTSD.
Collapse
Affiliation(s)
- Hannah N Carlson
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jeff L Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
21
|
Holleran KM, Rose JH, Fordahl SC, Benton KC, Rohr KE, Gasser PJ, Jones SR. Organic cation transporter 3 and the dopamine transporter differentially regulate catecholamine uptake in the basolateral amygdala and nucleus accumbens. Eur J Neurosci 2020; 52:4546-4562. [PMID: 32725894 DOI: 10.1111/ejn.14927] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 03/28/2020] [Accepted: 04/24/2020] [Indexed: 12/18/2022]
Abstract
Regional alterations in kinetics of catecholamine uptake are due in part to variations in clearance mechanisms. The rate of clearance is a critical determinant of the strength of catecholamine signaling. Catecholamine transmission in the nucleus accumbens core (NAcc) and basolateral amygdala (BLA) is of particular interest due to involvement of these regions in cognition and motivation. Previous work has shown that catecholamine clearance in the NAcc is largely mediated by the dopamine transporter (DAT), but clearance in the BLA is less DAT-dependent. A growing body of literature suggests that organic cation transporter 3 (OCT3) also contributes to catecholamine clearance in both regions. Consistent with different clearance mechanisms between regions, catecholamine clearance is more rapid in the NAcc than in the BLA, though mechanisms underlying this have not been resolved. We compared the expression of DAT and OCT3 and their contributions to catecholamine clearance in the NAcc and BLA. We found DAT protein levels were ~ 4-fold higher in the NAcc than in the BLA, while OCT3 protein expression was similar between the two regions. Immunofluorescent labeling of the two transporters in brain sections confirmed these findings. Ex vivo voltammetry demonstrated that the magnitude of catecholamine release was greater, and the clearance rate was faster in the NAcc than in the BLA. Additionally, catecholamine clearance in the BLA was more sensitive to the OCT3 inhibitor corticosterone, while clearance in the NAcc was more cocaine sensitive. These distinctions in catecholamine clearance may underlie differential effects of catecholamines on behavioral outputs mediated by these regions.
Collapse
Affiliation(s)
- Katherine M Holleran
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jamie H Rose
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Steven C Fordahl
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kelsey C Benton
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Kayla E Rohr
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Paul J Gasser
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
22
|
Adolescent Intermittent Ethanol Exposure Effects on Kappa Opioid Receptor Mediated Dopamine Transmission: Sex and Age of Exposure Matter. Brain Sci 2020; 10:brainsci10080472. [PMID: 32717830 PMCID: PMC7463732 DOI: 10.3390/brainsci10080472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 12/28/2022] Open
Abstract
Underage alcohol drinking increases the risk of developing alcohol use disorder (AUD). In rodents, adolescent ethanol exposure augments ethanol consumption and anxiety-like behavior while reducing social interaction. However, the underlying mechanisms driving these adaptations are unclear. The dopamine and kappa opioid receptor (KOR) systems in the nucleus accumbens (NAc) are implicated in affective disorders, including AUD, with studies showing augmented KOR function and reduced dopamine transmission in ethanol-dependent adult animals. Thus, here we examine the impact of adolescent intermittent ethanol (AIE) exposure on dopamine transmission and KOR function in the NAc. Rats were exposed to water or ethanol (4 g/kg, intragastrically) every other day during early (postnatal day (PD) 25–45) or late (PD 45–65) adolescence. While AIE exposure during early adolescence (early-AIE) did not alter dopamine release in male and female rats, AIE exposure during late adolescence (late-AIE) resulted in greater dopamine release in males and lower dopamine release in females. To determine the impact of AIE on KOR function, we measured the effect of KOR activation using U50,488 (0.01–1.00 µM) on dopamine release. Early-AIE exposure potentiated KOR-mediated inhibition of dopamine release in females, while late-AIE exposure attenuated this effect in males. Interestingly, no differences in KOR function were observed in early-AIE exposed males and late-AIE exposed females. Together, these data suggest that AIE exposure impact on neural processes is dependent on sex and exposure timing. These differences likely arise from differential developmental timing in males and females. This is the first study to show changes in KOR function following AIE exposure.
Collapse
|
23
|
Nolan SO, Zachry JE, Johnson AR, Brady LJ, Siciliano CA, Calipari ES. Direct dopamine terminal regulation by local striatal microcircuitry. J Neurochem 2020; 155:475-493. [PMID: 32356315 DOI: 10.1111/jnc.15034] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023]
Abstract
Regulation of axonal dopamine release by local microcircuitry is at the hub of several biological processes that govern the timing and magnitude of signaling events in reward-related brain regions. An important characteristic of dopamine release from axon terminals in the striatum is that it is rapidly modulated by local regulatory mechanisms. These processes can occur via homosynaptic mechanisms-such as presynaptic dopamine autoreceptors and dopamine transporters - as well heterosynaptic mechanisms such as retrograde signaling from postsynaptic cholinergic and dynorphin systems, among others. Additionally, modulation of dopamine release via diffusible messengers, such as nitric oxide and hydrogen peroxide, allows for various metabolic factors to quickly and efficiently regulate dopamine release and subsequent signaling. Here we review how these mechanisms work in concert to influence the timing and magnitude of striatal dopamine signaling, independent of action potential activity at the level of dopaminergic cell bodies in the midbrain, thereby providing a parallel pathway by which dopamine can be modulated. Understanding the complexities of local regulation of dopamine signaling is required for building comprehensive frameworks of how activity throughout the dopamine system is integrated to drive signaling and control behavior.
Collapse
Affiliation(s)
- Suzanne O Nolan
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jennifer E Zachry
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Amy R Johnson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Lillian J Brady
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN TN, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.,Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
24
|
Qu N, He Y, Wang C, Xu P, Yang Y, Cai X, Liu H, Yu K, Pei Z, Hyseni I, Sun Z, Fukuda M, Li Y, Tian Q, Xu Y. A POMC-originated circuit regulates stress-induced hypophagia, depression, and anhedonia. Mol Psychiatry 2020; 25:1006-1021. [PMID: 31485012 PMCID: PMC7056580 DOI: 10.1038/s41380-019-0506-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 06/01/2019] [Accepted: 07/17/2019] [Indexed: 01/27/2023]
Abstract
Chronic stress causes dysregulations of mood and energy homeostasis, but the neurocircuitry underlying these alterations remain to be fully elucidated. Here we demonstrate that chronic restraint stress in mice results in hyperactivity of pro-opiomelanocortin neurons in the arcuate nucleus of the hypothalamus (POMCARH neurons) associated with decreased neural activities of dopamine neurons in the ventral tegmental area (DAVTA neurons). We further revealed that POMCARH neurons project to the VTA and provide an inhibitory tone to DAVTA neurons via both direct and indirect neurotransmissions. Finally, we show that photoinhibition of the POMCARH→VTA circuit in mice increases body weight and food intake, and reduces depression-like behaviors and anhedonia in mice exposed to chronic restraint stress. Thus, our results identified a novel neurocircuitry regulating feeding and mood in response to stress.
Collapse
Affiliation(s)
- Na Qu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, 430012, Wuhan, China
| | - Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Pingwen Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Xing Cai
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Hesong Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Kaifan Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Zhou Pei
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Ilirjana Hyseni
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Zheng Sun
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Makoto Fukuda
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yi Li
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, 430012, Wuhan, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, 430074, Wuhan, China
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Institute for Brain Research, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Karkhanis AN, Al-Hasani R. Dynorphin and its role in alcohol use disorder. Brain Res 2020; 1735:146742. [PMID: 32114059 DOI: 10.1016/j.brainres.2020.146742] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
The dynorphin / kappa opioid receptor (KOR) system has been implicated in many aspects that influence neuropsychiatric disorders. Namely, this system modulates neural circuits that primarily regulate reward seeking, motivation processing, stress responsivity, and pain sensitivity, thus affecting the development of substance and alcohol use disorder (AUD). The effects of this system are often bidirectional and depend on projection targets. To date, a majority of the studies focusing on this system have examined the KOR function using agonists and antagonists. Indeed, there are studies that have examined prodynorphin and dynorphin levels by measuring mRNA and tissue content levels; however, static levels of the neuropeptide and its precursor do not explain complete and online function of the peptide as would be explained by measuring dynorphin transmission in real time. New and exciting methods using optogenetics, chemogenetics, genetic sensors, fast scan cyclic voltammetry are now being developed to detect various neuropeptides with a focus on opioid peptides, including dynorphin. In this review we discuss studies that examine dynorphin projections in areas involved in AUD, its functional involvement in AUD and vulnerability to develop AUD at various ages. Moreover, we discuss dynorphin's role in promoting AUD by dysregulation motivation circuits and how advancements in opioid peptide detection will further our understanding.
Collapse
Affiliation(s)
- Anushree N Karkhanis
- Department of Psychology, Developmental Exposure Alcohol Research Center, Center for Developmental and Behavioral Neuroscience, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY 13902, USA.
| | - Ream Al-Hasani
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, Department of Anesthesiology Washington University in St. Louis, Center for Clinical Pharmacology, Washington University School of Medicine & St. Louis College of Pharmacy 660 S.Euclid, Box 8054, St. Louis, MO 63110, USA.
| |
Collapse
|
26
|
Escobar ADP, Casanova JP, Andrés ME, Fuentealba JA. Crosstalk Between Kappa Opioid and Dopamine Systems in Compulsive Behaviors. Front Pharmacol 2020; 11:57. [PMID: 32132923 PMCID: PMC7040183 DOI: 10.3389/fphar.2020.00057] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/22/2020] [Indexed: 12/02/2022] Open
Abstract
The strength of goal-oriented behaviors is regulated by midbrain dopamine neurons. Dysfunctions of dopaminergic circuits are observed in drug addiction and obsessive-compulsive disorder. Compulsive behavior is a feature that both disorders share, which is associated to a heightened dopamine neurotransmission. The activity of midbrain dopamine neurons is principally regulated by the homeostatic action of dopamine through D2 receptors (D2R) that decrease the firing of neurons as well as dopamine synthesis and release. Dopamine transmission is also regulated by heterologous neurotransmitter systems such as the kappa opioid system, among others. Much of our current knowledge of the kappa opioid system and its influence on dopamine transmission comes from preclinical animal models of brain diseases. In 1988, using cerebral microdialysis, it was shown that the acute activation of the Kappa Opioid Receptors (KOR) decreases synaptic levels of dopamine in the striatum. This inhibitory effect of KOR opposes to the facilitating influence of drugs of abuse on dopamine release, leading to the proposition of the use of KOR agonists as pharmacological therapy for compulsive drug intake. Surprisingly, 30 years later, KOR antagonists are instead proposed to treat drug addiction. What may have happened during these years that generated this drastic change of paradigm? The collected evidence suggested that the effect of KOR on synaptic dopamine levels is complex, depending on the frequency of KOR activation and timing with other incoming stimuli to dopamine neurons, as well as sex and species differences. Conversely to its acute effect, chronic KOR activation seems to facilitate dopamine neurotransmission and dopamine-mediated behaviors. The opposing actions exerted by acute versus chronic KOR activation have been associated with an initial aversive and a delayed rewarding effect, during the exposure to drugs of abuse. Compulsive behaviors induced by repeated activation of D2R are also potentiated by the sustained co-activation of KOR, which correlates with decreased synaptic levels of dopamine and sensitized D2R. Thus, the time-dependent activation of KOR impacts directly on dopamine levels affecting the tuning of motivated behaviors. This review analyzes the contribution of the kappa opioid system to the dopaminergic correlates of compulsive behaviors.
Collapse
Affiliation(s)
- Angélica Del Pilar Escobar
- Centro Interdisciplinario de Neurociencias de Valparaíso, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| | - José Patricio Casanova
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Núcleo Milenio NUMIND Biology of Neuropsychiatric Disorders, Universidad de Valparaíso, Valparaíso, Chile
| | - María Estela Andrés
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José Antonio Fuentealba
- Department of Pharmacy and Interdisciplinary Center of Neuroscience, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
27
|
Puryear CB, Brooks J, Tan L, Smith K, Li Y, Cunningham J, Todtenkopf MS, Dean RL, Sanchez C. Opioid receptor modulation of neural circuits in depression: What can be learned from preclinical data? Neurosci Biobehav Rev 2020; 108:658-678. [DOI: 10.1016/j.neubiorev.2019.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
|
28
|
Abijo T, Blum K, Gondré-Lewis MC. Neuropharmacological and Neurogenetic Correlates of Opioid Use Disorder (OUD) As a Function of Ethnicity: Relevance to Precision Addiction Medicine. Curr Neuropharmacol 2020; 18:578-595. [PMID: 31744450 PMCID: PMC7457418 DOI: 10.2174/1570159x17666191118125702] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/31/2019] [Accepted: 11/16/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Over 100 people die daily from opioid overdose and $78.5B per year is spent on treatment efforts, however, the real societal cost is multifold greater. Alternative strategies to eradicate/manage drug misuse and addiction need consideration. The perception of opioid addiction as a social/criminal problem has evolved to evidence-based considerations of them as clinical disorders with a genetic basis. We present evaluations of the genetics of addiction with ancestryspecific risk profiles for consideration. OBJECTIVE Studies of gene variants associated with predisposition to substance use disorders (SUDs) are monolithic, and exclude many ethnic groups, especially Hispanics and African Americans. We evaluate gene polymorphisms that impact brain reward and predispose individuals to opioid addictions, with a focus on the disparity of research which includes individuals of African and Hispanic descent. METHODOLOGY PubMed and Google Scholar were searched for: Opioid Use Disorder (OUD), Genome- wide association studies (GWAS); genetic variants; polymorphisms, restriction fragment length polymorphisms (RFLP); genomics, epigenetics, race, ethnic group, ethnicity, ancestry, Caucasian/ White, African American/Black, Hispanic, Asian, addictive behaviors, reward deficiency syndrome (RDS), mutation, insertion/deletion, and promotor region. RESULTS Many studies exclude non-White individuals. Studies that include diverse populations report ethnicity-specific frequencies of risk genes, with certain polymorphisms specifically associated with Caucasian and not African-American or Hispanic susceptibility to OUD or SUDs, and vice versa. CONCLUSION To adapt precision medicine-based addiction management in a blended society, we propose that ethnicity/ancestry-informed genetic variations must be analyzed to provide real precision- guided therapeutics with the intent to attenuate this uncontrollable fatal epidemic.
Collapse
Affiliation(s)
| | | | - Marjorie C. Gondré-Lewis
- Address correspondence to this author at the Department of Anatomy, Howard University College of Medicine, 520 W St NW, Washington DC 20059 USA; Tel/Fax: +1-202-806-5274; E-mail:
| |
Collapse
|
29
|
The selective κ-opioid receptor antagonist JDTic attenuates the alcohol deprivation effect in rats. Eur Neuropsychopharmacol 2019; 29:1386-1396. [PMID: 31679889 DOI: 10.1016/j.euroneuro.2019.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/22/2019] [Accepted: 10/10/2019] [Indexed: 11/20/2022]
Abstract
The mechanisms behind relapse to ethanol intake in recovering alcoholics are still unclear. The negative reinforcing effects contributing to ethanol addiction, including relapse, are considered to be partly driven by the κ-opioidergic system. As the κ-opioidergic system interacts with the mesolimbic reward pathway, the aim of the study was to clarify the role of nucleus accumbens shell κ-opioidergic mechanisms in relapse to ethanol intake by using the alcohol deprivation effect (ADE) paradigm. The ADE is defined as a transient increase in voluntary ethanol intake after a forced period of abstinence. Male Long-Evans rats were trained to voluntarily consume 10% (v/v) ethanol solution. Ethanol access and deprivation cycles were initiated after stable ethanol intake baselines had been reached and bilateral guide cannulas had been implanted above the nucleus accumbens shell. One cycle consisted of 10 days of 90 min access to ethanol followed by 6 days of ethanol deprivation. The ADE was measured in the beginning of a new cycle. Rats received JDTic, a selective κ-antagonist, either subcutaneously (10 mg/kg) or intra-accumbally (15 µg/site) or, as a reference substance, systemic naltrexone (0.3 mg/kg) before ethanol re-access, and the effects on the ADE were evaluated. Systemic and intra-accumbal JDTic significantly attenuated the ADE on the first day of ethanol re-access, as did systemic naltrexone. Additionally, naltrexone decreased ethanol intake levels. These results suggest that nucleus accumbens shell κ-opioidergic mechanisms may have a role in mediating relapse to ethanol intake. Additionally, κ-antagonism could be a valuable adjunct in ethanol relapse prevention.
Collapse
|
30
|
Clark SD, Abi-Dargham A. The Role of Dynorphin and the Kappa Opioid Receptor in the Symptomatology of Schizophrenia: A Review of the Evidence. Biol Psychiatry 2019; 86:502-511. [PMID: 31376930 DOI: 10.1016/j.biopsych.2019.05.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/25/2019] [Accepted: 05/05/2019] [Indexed: 01/17/2023]
Abstract
Schizophrenia is a debilitating mental illness that affects approximately 1% of the world's population. Despite much research in its neurobiology to aid in developing new treatments, little progress has been made. One system that has not received adequate attention is the kappa opioid system and its potential role in the emergence of symptoms, as well as its therapeutic potential. Here we present an overview of the kappa system and review various lines of evidence derived from clinical studies for dynorphin and kappa opioid receptor involvement in the pathology of both the positive and negative symptoms of schizophrenia. This overview includes evidence for the psychotomimetic effects of kappa opioid receptor agonists in healthy volunteers and their reversal by the pan-opioid antagonists naloxone and naltrexone and evidence for a therapeutic benefit in schizophrenia for 4 pan-opioid antagonists. We describe the interactions between kappa opioid receptors and the dopaminergic pathways that are disrupted in schizophrenia and the histologic evidence suggesting abnormal kappa opioid receptor signaling in schizophrenia. We conclude by discussing future directions.
Collapse
Affiliation(s)
- Samuel David Clark
- Columbia University Medical Center, New York; Terran Biosciences Inc., New York.
| | - Anissa Abi-Dargham
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| |
Collapse
|
31
|
Social status and demographic effects of the kappa opioid receptor: a PET imaging study with a novel agonist radiotracer in healthy volunteers. Neuropsychopharmacology 2019; 44:1714-1719. [PMID: 30928993 PMCID: PMC6785144 DOI: 10.1038/s41386-019-0379-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/26/2019] [Accepted: 03/22/2019] [Indexed: 12/21/2022]
Abstract
Kappa opioid receptors (KORs) have been characterized as an aversive system in the brain and implicated in social behavior in preclinical models. This work investigated the effect of social status on the KOR system in humans using positron emission tomography (PET) imaging with the novel KOR agonist radiotracer [11C]EKAP. Eighteen healthy participants (mean age 41.2 ± 9.3) completed the Barratt Simplified Measure of Social Status (BSMSS), an MRI and an [11C]EKAP PET scan on the High Resolution Research Tomograph. Arterial blood sampling and metabolite analysis were conducted to obtain the input function. Regions of interest were based upon an MR template and included the reward/aversion areas of the brain. The multilinear analysis-1 (MA1) method was applied to the regional time-activity curves (TACs) to calculate [11C]EKAP regional volume of distribution (VT). Mixed models and Pearson correlation coefficients were used for body mass index (BMI), gender and age, with age being dropped in subsequent analyses because of nonsignificance. An overall effect of primary ROIs (F7, 112 7.43, p < 0.0001), BSMSS score (F1, 13 7.45, p = 0.02), BMI (F1, 13 23.5, p < 0.001), and gender (F1, 13 23.75, p < 0.001), but not age (F1, 13 1.12, p = 0.35) was observed. Regional [11C]EKAP VT and BSMSS were found to be negatively correlated in the amygdala (r = -0.69, p < 0.01), anterior cingulate cortex (r = -0.56, p = 0.02), caudate (r = -0.66, p < 0.01), frontal cortex (r = -0.52, p = 0.04), hippocampus (r = -0.60, p = 0.01), pallidum (r = -0.59, p = 0.02), putamen (r = -0.62, p = 0.01), and ventral striatum (r = -0.66, p < 0.01). In secondary (non-reward) regions, correlations of [11C]EKAP VT and BSMSS were nonsignificant with the exception of the insula. There was an inverse correlation between social status and KOR levels that was largely specific to the reward/aversion (e.g., saliency) areas of the brain. This finding suggests the KOR system may act as a mediator for the negative effects of social behaviors in humans.
Collapse
|
32
|
Beck TC, Hapstack MA, Beck KR, Dix TA. Therapeutic Potential of Kappa Opioid Agonists. Pharmaceuticals (Basel) 2019; 12:ph12020095. [PMID: 31226764 PMCID: PMC6631266 DOI: 10.3390/ph12020095] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 06/12/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022] Open
Abstract
Many original research articles have been published that describe findings and outline areas for the development of kappa-opioid agonists (KOAs) as novel drugs; however, a single review article that summarizes the broad potential for KOAs in drug development does not exist. It is well-established that KOAs demonstrate efficacy in pain attenuation; however, KOAs also have proven to be beneficial in treating a variety of novel but often overlapping conditions including cardiovascular disease, pruritus, nausea, inflammatory diseases, spinal anesthesia, stroke, hypoxic pulmonary hypertension, multiple sclerosis, addiction, and post-traumatic cartilage degeneration. This article summarizes key findings of KOAs and discusses the untapped therapeutic potential of KOAs in the treatment of many human diseases.
Collapse
Affiliation(s)
- Tyler C Beck
- Drug Discovery & Biomedical Sciences, Medical University of South Carolina, 280 Calhoun Street, QF204, Charleston, SC 29424-2303, USA.
- College of Medicine, 173 Ashley Ave., Charleston, SC 29424-2303, USA.
| | | | - Kyle R Beck
- College of Pharmacy, The Ohio State University, 500 W 12th Ave, Columbus, OH 43210-9998, USA.
| | - Thomas A Dix
- Drug Discovery & Biomedical Sciences, Medical University of South Carolina, 280 Calhoun Street, QF204, Charleston, SC 29424-2303, USA.
- JT Pharmaceuticals, Inc., 300 West Coleman Blvd., Suite 203, Mount Pleasant, SC 29464-2303, USA.
| |
Collapse
|
33
|
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]
|
34
|
Castro DC, Bruchas MR. A Motivational and Neuropeptidergic Hub: Anatomical and Functional Diversity within the Nucleus Accumbens Shell. Neuron 2019; 102:529-552. [PMID: 31071288 PMCID: PMC6528838 DOI: 10.1016/j.neuron.2019.03.003] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 01/14/2023]
Abstract
The mesocorticolimbic pathway is canonically known as the "reward pathway." Embedded within the center of this circuit is the striatum, a massive and complex network hub that synthesizes motivation, affect, learning, cognition, stress, and sensorimotor information. Although striatal subregions collectively share many anatomical and functional similarities, it has become increasingly clear that it is an extraordinarily heterogeneous region. In particular, the nucleus accumbens (NAc) medial shell has repeatedly demonstrated that the rules dictated by more dorsal aspects of the striatum do not apply or are even reversed in functional logic. These discrepancies are perhaps most easily captured when isolating the functions of various neuromodulatory peptide systems within the striatum. Endogenous peptides are thought to play a critical role in modulating striatal signals to either amplify or dampen evoked behaviors. Here we describe the anatomical-functional backdrop upon which several neuropeptides act within the NAc to modulate behavior, with a specific emphasis on nucleus accumbens medial shell and stress responsivity. Additionally, we propose that, as the field continues to dissect fast neurotransmitter systems within the NAc, we must also provide considerable contextual weight to the roles local peptides play in modulating these circuits to more comprehensively understand how this important subregion gates motivated behaviors.
Collapse
Affiliation(s)
- Daniel C Castro
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Michael R Bruchas
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
35
|
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
|
36
|
de Oliveira PG, Ramos MLS, Amaro AJ, Dias RA, Vieira SI. G i/o-Protein Coupled Receptors in the Aging Brain. Front Aging Neurosci 2019; 11:89. [PMID: 31105551 PMCID: PMC6492497 DOI: 10.3389/fnagi.2019.00089] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/03/2019] [Indexed: 12/18/2022] Open
Abstract
Cells translate extracellular signals to regulate processes such as differentiation, metabolism and proliferation, via transmembranar receptors. G protein-coupled receptors (GPCRs) belong to the largest family of transmembrane receptors, with over 800 members in the human species. Given the variety of key physiological functions regulated by GPCRs, these are main targets of existing drugs. During normal aging, alterations in the expression and activity of GPCRs have been observed. The central nervous system (CNS) is particularly affected by these alterations, which results in decreased brain functions, impaired neuroregeneration, and increased vulnerability to neuropathologies, such as Alzheimer's and Parkinson diseases. GPCRs signal via heterotrimeric G proteins, such as Go, the most abundant heterotrimeric G protein in CNS. We here review age-induced effects of GPCR signaling via the Gi/o subfamily at the CNS. During the aging process, a reduction in protein density is observed for almost half of the Gi/o-coupled GPCRs, particularly in age-vulnerable regions such as the frontal cortex, hippocampus, substantia nigra and striatum. Gi/o levels also tend to decrease with aging, particularly in regions such as the frontal cortex. Alterations in the expression and activity of GPCRs and coupled G proteins result from altered proteostasis, peroxidation of membranar lipids and age-associated neuronal degeneration and death, and have impact on aging hallmarks and age-related neuropathologies. Further, due to oligomerization of GPCRs at the membrane and their cooperative signaling, down-regulation of a specific Gi/o-coupled GPCR may affect signaling and drug targeting of other types/subtypes of GPCRs with which it dimerizes. Gi/o-coupled GPCRs receptorsomes are thus the focus of more effective therapeutic drugs aiming to prevent or revert the decline in brain functions and increased risk of neuropathologies at advanced ages.
Collapse
Affiliation(s)
- Patrícia G de Oliveira
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - Marta L S Ramos
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - António J Amaro
- School of Health Sciences (ESSUA), Universidade de Aveiro, Aveiro, Portugal
| | - Roberto A Dias
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - Sandra I Vieira
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| |
Collapse
|
37
|
Abstract
Chronic pain is a frequent condition that affects an estimated 20% of people worldwide, accounting for 15%-20% of doctors' appointments (Treede et al., 2015). It lacks the acute warning function of physiologic nociception, and instead involves the activation of multiple neurophysiologic mechanisms in the somatosensory system, a complex neuronal network under the control of powerful autoregulatory loops and able to undergo rapid neuroplastic alteration (Verdu et al., 2008). There is a growing body of research suggesting that some such pathways are shared by major psychologic disorders such as depression and anxiety, opening new avenues in co-treatment strategies. In particular, besides anticonvulsants, which are today used as analgesics, other psychopharmaceuticals, such as the tricyclic antidepressants, are displaying efficacy in the treatment of neuropathic and nociceptive chronic pain. The state of the art regarding the mechanisms of nociception and the pharmacology of both the neurotransmitters involved and the wide range of psychoactive compounds that may be useful in the treatment of chronic pain are discussed.
Collapse
|
38
|
Abstract
Research concerning psychiatric issues relating to opioid drugs currently focuses primarily on their role in reinforcing addictive behaviors, given the recent proliferation of lethal abuse of illicit opiates in the United States and around the world. In contrast, this article will review the mechanism of action of opioids in affective disorders and the available evidence and potential for their use, especially in the treatment of resistant major depression. Buprenorphine is the opioid derivative of special interest; we review this and other opioid derivatives, highlighting the growing role of opioids in treating depressive illnesses and other related psychopathologies.
Collapse
Affiliation(s)
- Parnika P Saxena
- Brockton Neighborhood Health Center, 63 Main St, Brockton, MA, 02301, USA.
| | - J Alexander Bodkin
- McLean Hospital, 115 Mill St, North Belknap, Belmont, MA, 02478, USA.,Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
| |
Collapse
|
39
|
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
|
40
|
Uhari-Väänänen J, Raasmaja A, Bäckström P, Oinio V, Carroll FI, Airavaara M, Kiianmaa K, Piepponen P. The κ-opioid receptor antagonist JDTic decreases ethanol intake in alcohol-preferring AA rats. Psychopharmacology (Berl) 2018; 235:1581-1591. [PMID: 29492614 DOI: 10.1007/s00213-018-4868-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 02/20/2018] [Indexed: 11/30/2022]
Abstract
RATIONALE Studies suggest that the κ-opioidergic system becomes overactivated as ethanol use disorders develop. Nalmefene, a currently approved treatment for ethanol use disorders, may also elicit some of its main effects via the κ-opioidergic system. However, the exact role of κ-opioid receptors on regulating ethanol intake and contribution to the development of ethanol addiction remains to be elucidated. OBJECTIVES The aim of the present study was to clarify the role of accumbal κ-opioid receptors in controlling ethanol intake in alcohol-preferring Alko Alcohol (AA) rats. METHODS Microinfusions of the long-acting and selective κ-opioid receptor antagonist JDTic (1-15 μg/site) were administered bilaterally into the nucleus accumbens shell of AA rats voluntarily consuming 10% ethanol solution in the intermittent, time-restricted two-bottle choice access paradigm. JDTic (10 mg/kg) was also administered subcutaneously. Both the acute and long-term effects of the treatment on ethanol intake were examined. As a reference, nor-BNI (3 μg/site) was administered intra-accumbally. RESULTS Systemically administered JDTic decreased ethanol intake significantly 2 days and showed a similar trend 4 days after administration. Furthermore, intra-accumbally administered JDTic showed a weak decreasing effect on ethanol intake long-term but had no acute effects. Intra-accumbal administration of nor-BNI tended to decrease ethanol intake. CONCLUSIONS The results provide further evidence that κ-opioid receptors play a role in controlling ethanol intake and that accumbal κ-opioid receptors participate in the modulation of the reinforcing effects of ethanol. Furthermore, the results suggest that κ-opioid receptor antagonists may be a valuable adjunct in the pharmacotherapy of ethanol use disorders.
Collapse
Affiliation(s)
- Johanna Uhari-Väänänen
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland. .,Department of Health, National Institute for Health and Welfare, P.O. Box 30, 00271, Helsinki, Finland.
| | - Atso Raasmaja
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Pia Bäckström
- Department of Health, National Institute for Health and Welfare, P.O. Box 30, 00271, Helsinki, Finland
| | - Ville Oinio
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.,Department of Health, National Institute for Health and Welfare, P.O. Box 30, 00271, Helsinki, Finland
| | - F Ivy Carroll
- RTI International, P.O. Box 12194, Research Triangle Park, NC, USA
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Kalervo Kiianmaa
- Department of Health, National Institute for Health and Welfare, P.O. Box 30, 00271, Helsinki, Finland
| | - Petteri Piepponen
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| |
Collapse
|
41
|
Siciliano CA, Karkhanis AN, Holleran KM, Melchior JR, Jones SR. Cross-Species Alterations in Synaptic Dopamine Regulation After Chronic Alcohol Exposure. Handb Exp Pharmacol 2018; 248:213-238. [PMID: 29675581 PMCID: PMC6195853 DOI: 10.1007/164_2018_106] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alcohol use disorders are a leading public health concern, engendering enormous costs in terms of both economic loss and human suffering. These disorders are characterized by compulsive and excessive alcohol use, as well as negative affect and alcohol craving during abstinence. Extensive research has implicated the dopamine system in both the acute pharmacological effects of alcohol and the symptomology of alcohol use disorders that develop after extended alcohol use. Preclinical research has shed light on many mechanisms by which chronic alcohol exposure dysregulates the dopamine system. However, many of the findings are inconsistent across experimental parameters such as alcohol exposure length, route of administration, and model organism. We propose that the dopaminergic alterations driving the core symptomology of alcohol use disorders are likely to be relatively stable across experimental settings. Recent work has been aimed at using multiple model organisms (mouse, rat, monkey) across various alcohol exposure procedures to search for commonalities. Here, we review recent advances in our understanding of the effects of chronic alcohol use on the dopamine system by highlighting findings that are consistent across experimental setting and species.
Collapse
Affiliation(s)
- Cody A Siciliano
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Anushree N Karkhanis
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Katherine M Holleran
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James R Melchior
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
42
|
Karkhanis A, Holleran KM, Jones SR. Dynorphin/Kappa Opioid Receptor Signaling in Preclinical Models of Alcohol, Drug, and Food Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:53-88. [PMID: 29056156 DOI: 10.1016/bs.irn.2017.08.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The dynorphin/kappa opioid receptor (KOR) system is implicated in the "dark side" of addiction, in which stress exacerbates maladaptive responses to drug and alcohol exposure. For example, acute stress and acute ethanol exposure result in an elevation in dynorphin, the KOR endogenous ligand. Activation of KORs results in modulation of several neurotransmitters; however, this chapter will focus on its regulatory effects on dopamine in mesolimbic areas. Specifically, KOR activation has an inhibitory effect on dopamine release, thereby influencing reward processing. Repeated stimulation of KORs, for example, via chronic drug and/or stress exposure, results in increased function of the dynorphin/KOR system. This augmentation in KOR function shifts the homeostatic balance in favor of an overall reduction in dopamine signaling via either by reducing dopamine release or by increasing dopamine transporter function. This chapter examines the effects of chronic ethanol exposure on KOR function and the downstream effects on dopamine transmission. Additionally, the impact of chronic cocaine exposure and its effects on KOR function will be explored. Further, KORs may also be involved in driving excessive consumption of food, contributing to the risk of developing obesity. While some studies have shown that KOR agonists reduce drug intake, other studies have shown that antagonists reduce addiction-like behaviors, demonstrating therapeutic potential. For example, KOR inhibition reduces ethanol intake in dependent animals, motivation to self-administer cocaine in chronic stress-exposed animals, and food consumption in obese animals. This chapter will delve into the mechanisms by which modulation of the dynorphin/KOR system may be therapeutic.
Collapse
Affiliation(s)
| | | | - Sara R Jones
- Wake Forest School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
43
|
Escobar AP, González MP, Meza RC, Noches V, Henny P, Gysling K, España RA, Fuentealba JA, Andrés ME. Mechanisms of Kappa Opioid Receptor Potentiation of Dopamine D2 Receptor Function in Quinpirole-Induced Locomotor Sensitization in Rats. Int J Neuropsychopharmacol 2017; 20:660-669. [PMID: 28531297 PMCID: PMC5569963 DOI: 10.1093/ijnp/pyx042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/15/2017] [Accepted: 05/19/2017] [Indexed: 12/11/2022] Open
Abstract
Background Increased locomotor activity in response to the same stimulus is an index of behavioral sensitization observed in preclinical models of drug addiction and compulsive behaviors. Repeated administration of quinpirole, a D2/D3 dopamine agonist, induces locomotor sensitization. This effect is potentiated and accelerated by co-administration of U69593, a kappa opioid receptor agonist. The mechanism underlying kappa opioid receptor potentiation of quinpirole-induced locomotor sensitization remains to be elucidated. Methods Immunofluorescence anatomical studies were undertaken in mice brain slices and rat presynaptic synaptosomes to reveal kappa opioid receptor and D2R pre- and postsynaptic colocalization in the nucleus accumbens. Tonic and phasic dopamine release in the nucleus accumbens of rats repeatedly treated with U69593 and quinpirole was assessed by microdialysis and fast scan cyclic voltammetry. Results Anatomical data show that kappa opioid receptor and D2R colocalize postsynaptically in medium spiny neurons of the nucleus accumbens and the highest presynaptic colocalization occurs on the same dopamine terminals. Significantly reduced dopamine levels were observed in quinpirole, and U69593-quinpirole treated rats, explaining sensitization of D2R. Presynaptic inhibition induced by kappa opioid receptor and D2R of electrically evoked dopamine release was faster in U69593-quinpirole compared with quinpirole-repeatedly treated rats. Conclusions Pre- and postsynaptic colocalization of kappa opioid receptor and D2R supports a role for kappa opioid receptor potentiating both the D2R inhibitory autoreceptor function and the inhibitory action of D2R on efferent medium spiny neurons. Kappa opioid receptor co-activation accelerates D2R sensitization by contributing to decrease dopamine release in the nucleus accumbens.
Collapse
Affiliation(s)
- Angélica P Escobar
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| | - Marcela P González
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| | - Rodrigo C Meza
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| | - Verónica Noches
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| | - Pablo Henny
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| | - Katia Gysling
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| | - Rodrigo A España
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| | - José A Fuentealba
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| | - María E Andrés
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Escobar, Ms González, and Drs Noches, Gysling, and Andrés); Laboratory of Neuroanatomy, Department of Anatomy and Interdisciplinary Center of Neuroscience, NeuroUC, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile (Mr Meza and Dr Henny); Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania (Dr España); Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile (Dr Fuentealba)
| |
Collapse
|
44
|
The Link between Depression and Chronic Pain: Neural Mechanisms in the Brain. Neural Plast 2017; 2017:9724371. [PMID: 28706741 PMCID: PMC5494581 DOI: 10.1155/2017/9724371] [Citation(s) in RCA: 414] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 05/04/2017] [Accepted: 05/24/2017] [Indexed: 02/08/2023] Open
Abstract
Chronic pain, as a stress state, is one of the critical factors for determining depression, and their coexistence tends to further aggravate the severity of both disorders. Unfortunately, their association remains unclear, which creates a bottleneck problem for managing chronic pain-induced depression. In recent years, studies have found considerable overlaps between pain- and depression-induced neuroplasticity changes and neurobiological mechanism changes. Such overlaps are vital to facilitating the occurrence and development of chronic pain and chronic pain-induced depression. In this review, we summarized the role of neuroplasticity in the occurrence and development of the two disorders in question and explored individualized application strategies of analgesic drugs and antidepressants that have different pharmacological effects in the treatment of chronic pain-induced depression. Therefore, this review may provide new insights into the understanding of association between chronic pain and depression.
Collapse
|
45
|
Anderson RI, Becker HC. Role of the Dynorphin/Kappa Opioid Receptor System in the Motivational Effects of Ethanol. Alcohol Clin Exp Res 2017; 41:1402-1418. [PMID: 28425121 DOI: 10.1111/acer.13406] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 04/13/2017] [Indexed: 12/20/2022]
Abstract
Evidence has demonstrated that dynorphin (DYN) and the kappa opioid receptor (KOR) system contribute to various psychiatric disorders, including anxiety, depression, and addiction. More recently, this endogenous opioid system has received increased attention as a potential therapeutic target for treating alcohol use disorders. In this review, we provide an overview and synthesis of preclinical studies examining the influence of alcohol (ethanol [EtOH]) exposure on DYN/KOR expression and function, as well as studies examining the effects of DYN/KOR manipulation on EtOH's rewarding and aversive properties. We then describe work that has characterized effects of KOR activation and blockade on EtOH self-administration and EtOH dependence/withdrawal-related behaviors. Finally, we address how the DYN/KOR system may contribute to stress-EtOH interactions. Despite an apparent role for the DYN/KOR system in motivational effects of EtOH, support comes from relatively few studies. Nevertheless, review of this literature reveals several common themes: (i) rodent strains genetically predisposed to consume more EtOH generally appear to have reduced DYN/KOR tone in brain reward circuitry; (ii) acute and chronic EtOH exposure typically up-regulate the DYN/KOR system; (iii) KOR antagonists reduce behavioral indices of negative affect associated with stress and chronic EtOH exposure/withdrawal; and (iv) KOR antagonists are effective in reducing EtOH consumption, but are often more efficacious under conditions that engender high levels of consumption, such as dependence or stress exposure. These results support the contention that the DYN/KOR system plays a significant role in contributing to dependence- and stress-induced elevation in EtOH consumption. Overall, more comprehensive analyses (on both behavioral and mechanistic levels) are needed to provide additional insight into how the DYN/KOR system is engaged and adapts to influence the motivation effects of EtOH. This information will be critical for the development of new pharmacological agents targeting KORs as promising novel therapeutics for alcohol use disorders and comorbid affective disorders.
Collapse
Affiliation(s)
- Rachel I Anderson
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina.,Department of Neuroscience , Medical University of South Carolina, Charleston, South Carolina.,RHJ Department of Veterans Affairs Medical Center , Charleston, South Carolina
| |
Collapse
|
46
|
Ugur M, Kaya E, Gozen O, Koylu EO, Kanit L, Keser A, Balkan B. Chronic nicotine-induced changes in gene expression of delta and kappa-opioid receptors and their endogenous ligands in the mesocorticolimbic system of the rat. Synapse 2017; 71. [PMID: 28509375 DOI: 10.1002/syn.21985] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022]
Abstract
Delta and kappa opioid receptors (DOR and KOR, respectively) and their endogenous ligands, proenkephalin (PENK) and prodynorphin (PDYN)-derived opioid peptides are proposed as important mediators of nicotine reward. This study investigated the regulatory effect of chronic nicotine treatment on the gene expression of DOR, KOR, PENK and PDYN in the mesocorticolimbic system. Three groups of rats were injected subcutaneously with nicotine at doses of 0.2, 0.4, or 0.6 mg/kg/day for 6 days. Rats were decapitated 1 hr after the last dose on day six, as this timing coincides with increased dopamine release in the mesocorticolimbic system. mRNA levels in the ventral tegmental area (VTA), lateral hypothalamic area (LHA), amygdala (AMG), dorsal striatum (DST), nucleus accumbens, and medial prefrontal cortex were measured by quantitative real-time PCR. Our results showed that nicotine upregulated DOR mRNA in the VTA at all of the doses employed, in the AMG at the 0.4 and 0.6 mg/kg doses, and in the DST at the 0.4 mg/kg dose. Conversely, PDYN mRNA was reduced in the LHA with 0.6 mg/kg nicotine and in the AMG with 0.4 mg/kg nicotine. KOR mRNA was also decreased in the DST with 0.6 mg/kg nicotine. Nicotine did not regulate PENK mRNA in any brain region studied.
Collapse
Affiliation(s)
- Muzeyyen Ugur
- Department of Physiology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Egemen Kaya
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Oguz Gozen
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Ersin O Koylu
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Lutfiye Kanit
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Aysegul Keser
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Burcu Balkan
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| |
Collapse
|
47
|
Carlezon WA, Krystal AD. Kappa-Opioid Antagonists for Psychiatric Disorders: From Bench to Clinical Trials. Depress Anxiety 2016; 33:895-906. [PMID: 27699938 PMCID: PMC5288841 DOI: 10.1002/da.22500] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 12/15/2022] Open
Abstract
Kappa-opioid receptor (KOR) antagonists are currently being considered for the treatment of a variety of neuropsychiatric conditions, including depressive, anxiety, and substance abuse disorders. A general ability to mitigate the effects of stress, which can trigger or exacerbate these conditions, may explain their putative efficacy across such a broad array of conditions. The discovery of their potentially therapeutic effects evolved from preclinical research designed to characterize the molecular mechanisms by which experience causes neuroadaptations in the nucleus accumbens (NAc), a key element of brain reward circuitry. This research established that exposure to drugs of abuse or stress increases the activity of the transcription factor CREB (cAMP response element binding protein) in the NAc, which leads to elevated expression of the opioid peptide dynorphin that in turn causes core signs of depressive- and anxiety-related disorders. Disruption of KORs-the endogenous receptors for dynorphin-produces antidepressant- and anxiolytic-like actions in screening procedures that identify standard drugs of these classes, and reduces stress effects in tests used to study addiction and stress-related disorders. Although interest in this target is high, prototypical KOR antagonists have extraordinarily persistent pharmacodynamic effects that complicate clinical trials. The development of shorter acting KOR antagonists together with more rapid designs for clinical trials may soon provide insight on whether these drugs are efficacious as would be predicted by preclinical work. If successful, KOR antagonists would represent a unique example in psychiatry where the therapeutic mechanism of a drug class is understood before it is shown to be efficacious in humans.
Collapse
Affiliation(s)
- William A. Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA
| | - Andrew D. Krystal
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC
| |
Collapse
|
48
|
Sulzer D, Cragg SJ, Rice ME. Striatal dopamine neurotransmission: regulation of release and uptake. ACTA ACUST UNITED AC 2016; 6:123-148. [PMID: 27141430 DOI: 10.1016/j.baga.2016.02.001] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dopamine (DA) transmission is governed by processes that regulate release from axonal boutons in the forebrain and the somatodendritic compartment in midbrain, and by clearance by the DA transporter, diffusion, and extracellular metabolism. We review how axonal DA release is regulated by neuronal activity and by autoreceptors and heteroreceptors, and address how quantal release events are regulated in size and frequency. In brain regions densely innervated by DA axons, DA clearance is due predominantly to uptake by the DA transporter, whereas in cortex, midbrain, and other regions with relatively sparse DA inputs, the norepinephrine transporter and diffusion are involved. We discuss the role of DA uptake in restricting the sphere of influence of DA and in temporal accumulation of extracellular DA levels upon successive action potentials. The tonic discharge activity of DA neurons may be translated into a tonic extracellular DA level, whereas their bursting activity can generate discrete extracellular DA transients.
Collapse
Affiliation(s)
- David Sulzer
- Depts of Psychiatry, Neurology, & Pharmacology, NY State Psychiatric Institute, Columbia University, New York, NY, USA
| | - Stephanie J Cragg
- Dept Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Margaret E Rice
- Depts of Neurosurgery & Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
49
|
Karkhanis AN, Rose JH, Weiner JL, Jones SR. Early-Life Social Isolation Stress Increases Kappa Opioid Receptor Responsiveness and Downregulates the Dopamine System. Neuropsychopharmacology 2016; 41:2263-74. [PMID: 26860203 PMCID: PMC4946054 DOI: 10.1038/npp.2016.21] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/15/2016] [Accepted: 02/05/2016] [Indexed: 12/30/2022]
Abstract
Chronic early-life stress increases vulnerability to alcoholism and anxiety disorders during adulthood. Similarly, rats reared in social isolation (SI) during adolescence exhibit augmented ethanol intake and anxiety-like behaviors compared with group housed (GH) rats. Prior studies suggest that disruption of dopamine (DA) signaling contributes to SI-associated behaviors, although the mechanisms underlying these alterations are not fully understood. Kappa opioid receptors (KORs) have an important role in regulating mesolimbic DA signaling, and other kinds of stressors have been shown to augment KOR function. Therefore, we tested the hypothesis that SI-induced increases in KOR function contribute to the dysregulation of NAc DA and the escalation in ethanol intake associated with SI. Our ex vivo voltammetry experiments showed that the inhibitory effects of the kappa agonist U50,488 on DA release were significantly enhanced in the NAc core and shell of SI rats. Dynorphin levels in NAc tissue were observed to be lower in SI rats. Microdialysis in freely moving rats revealed that SI was also associated with reduced baseline DA levels, and pretreatment with the KOR antagonist nor-binaltorphimine (nor-BNI) increased DA levels selectively in SI subjects. Acute ethanol elevated DA in SI and GH rats and nor-BNI pretreatment augmented this effect in SI subjects, while having no effect on ethanol-stimulated DA release in GH rats. Together, these data suggest that KORs may have increased responsiveness following SI, which could lead to hypodopaminergia and contribute to an increased drive to consume ethanol. Indeed, SI rats exhibited greater ethanol intake and preference and KOR blockade selectively attenuated ethanol intake in SI rats. Collectively, the findings that nor-BNI reversed SI-mediated hypodopaminergic state and escalated ethanol intake suggest that KOR antagonists may represent a promising therapeutic strategy for the treatment of alcohol use disorders, particularly in cases linked to chronic early-life stress.
Collapse
Affiliation(s)
- Anushree N Karkhanis
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA,Translational Center for the Neurobehavioral Study of Alcohol, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jamie H Rose
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jeffrey L Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA,Translational Center for the Neurobehavioral Study of Alcohol, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA,Translational Center for the Neurobehavioral Study of Alcohol, Wake Forest School of Medicine, Winston-Salem, NC, USA,Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA, Tel: +1 336 716 8533, Fax: +1 336 716 8501, E-mail:
| |
Collapse
|
50
|
Karkhanis AN, Huggins KN, Rose JH, Jones SR. Switch from excitatory to inhibitory actions of ethanol on dopamine levels after chronic exposure: Role of kappa opioid receptors. Neuropharmacology 2016; 110:190-197. [PMID: 27450094 DOI: 10.1016/j.neuropharm.2016.07.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/04/2016] [Accepted: 07/18/2016] [Indexed: 12/17/2022]
Abstract
Acute ethanol exposure is known to stimulate the dopamine system; however, chronic exposure has been shown to downregulate the dopamine system. In rodents, chronic intermittent exposure (CIE) to ethanol also increases negative affect during withdrawal, such as, increases in anxiety- and depressive-like behavior. Moreover, CIE exposure results in increased ethanol drinking and preference during withdrawal. Previous literature documents reductions in CIE-induced anxiety-, depressive-like behaviors and ethanol intake in response to kappa opioid receptor (KOR) blockade. KORs are located on presynaptic dopamine terminals in the nucleus accumbens (NAc) and inhibit release, an effect which has been linked to negative affective behaviors. Previous reports show an upregulation in KOR function following extended CIE exposure; however it is not clear whether there is a direct link between KOR upregulation and dopamine downregulation during withdrawal from CIE. This study aimed to examine the effects of KOR modulation on dopamine responses to ethanol of behaving mice exposed to air or ethanol vapor in a repeated intermittent pattern. First, we showed that KORs have a greater response to an agonist after moderate CIE compared to air exposed mice using ex vivo fast scan cyclic voltammetry. Second, using in vivo microdialysis, we showed that, in contrast to the expected increase in extracellular levels of dopamine following an acute ethanol challenge in air exposed mice, CIE exposed mice exhibited a robust decrease in dopamine levels. Third, we showed that blockade of KORs reversed the aberrant inhibitory dopamine response to ethanol in CIE exposed mice while not affecting the air exposed mice demonstrating that inhibition of KORs "rescued" dopamine responses in CIE exposed mice. Taken together, these findings indicate that augmentation of dynorphin/KOR system activity drives the reduction in stimulated (electrical and ethanol) dopamine release in the NAc. Thus, blockade of KORs is a promising avenue for developing pharmacotherapies for alcoholism.
Collapse
Affiliation(s)
- Anushree N Karkhanis
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA; Translational Center for the Neurobehavioral Study of Alcohol, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kimberly N Huggins
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jamie H Rose
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA; Translational Center for the Neurobehavioral Study of Alcohol, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|