1
|
Tollefson S, Stoughton C, Himes ML, McKinney KE, Mason S, Ciccocioppo R, Narendran R. Imaging Nociceptin Opioid Peptide Receptors in Alcohol Use Disorder With [ 11C]NOP-1A and Positron Emission Tomography: Findings From a Second Cohort. Biol Psychiatry 2023; 94:416-423. [PMID: 36906499 PMCID: PMC10319913 DOI: 10.1016/j.biopsych.2022.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/14/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Nociceptin, which binds to the nociceptin opioid peptide receptor (NOP), regulates stress and reward in addiction. In a previous [11C]NOP-1A positron emission tomography (PET) study, we found no differences in NOP in non-treatment-seeking individuals with alcohol use disorder (AUD) relative to healthy control subjects Here, we evaluated NOP in treatment-seeking individuals with AUD to document its relationship with relapse to alcohol. METHODS [11C]NOP-1A distribution volume (VT) was measured in recently abstinent individuals with AUD and healthy control subjects (n = 27/group) using an arterial input function-based kinetic analysis in brain regions that regulate reward and stress behaviors. Recent heavy drinking before PET was quantified using hair ethyl glucuronide (≥30 pg/mg was defined as heavy drinking). To document relapse, 22 subjects with AUD were followed with urine ethyl glucoronide tests (3/week) for 12 weeks after PET, where they were incentivized with money to abstain. RESULTS There were no differences in [11C]NOP-1A VT between individuals with AUD and healthy control subjects. Individuals with AUD who drank heavily before the study had significantly lower VT than those with no recent heavy drinking history. Significant negative correlations between VT and the number of drinking days and the number of drinks consumed per drinking day in the 30 days before enrollment were also present. Individuals with AUD who relapsed (and dropped out) had significantly lower VT than those who abstained for 12 weeks. CONCLUSIONS Lower NOP VT in heavy drinking AUD predicted relapse to alcohol during a 12-week follow-up period. The results of this PET study support the need to investigate medications that act at NOP to prevent relapse in individuals with AUD.
Collapse
Affiliation(s)
- Savannah Tollefson
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Clara Stoughton
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael L Himes
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kaylynn E McKinney
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Scott Mason
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
2
|
Rahman MM, Islam MR, Supti FA, Dhar PS, Shohag S, Ferdous J, Shuvo SK, Akter A, Hossain MS, Sharma R. Exploring the Therapeutic Effect of Neurotrophins and Neuropeptides in Neurodegenerative Diseases: at a Glance. Mol Neurobiol 2023:10.1007/s12035-023-03328-5. [PMID: 37052791 DOI: 10.1007/s12035-023-03328-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/22/2023] [Indexed: 04/14/2023]
Abstract
Neurotrophins and neuropeptides are the essential regulators of peripheral nociceptive nerves that help to induce, sensitize, and maintain pain. Neuropeptide has a neuroprotective impact as it increases trophic support, regulates calcium homeostasis, and reduces excitotoxicity and neuroinflammation. In contrast, neurotrophins target neurons afflicted by ischemia, epilepsy, depression, and eating disorders, among other neuropsychiatric conditions. Neurotrophins are reported to inhibit neuronal death. Strategies maintained for "brain-derived neurotrophic factor (BDNF) therapies" are to upregulate BDNF levels using the delivery of protein and genes or compounds that target BDNF production and boosting BDNF signals by expanding with BDNF mimetics. This review discusses the mechanisms of neurotrophins and neuropeptides against acute neural damage as well as highlighting neuropeptides as a potential therapeutic agent against Parkinson's disease (PD), Huntington's disease (HD), Alzheimer's disease (AD), and Machado-Joseph disease (MJD), the signaling pathways affected by neurotrophins and their receptors in both standard and diseased CNS systems, and future perspectives that can lead to the potent application of neurotrophins and neuropeptides in neurodegenerative diseases (NDs).
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Fatema Akter Supti
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Puja Sutro Dhar
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Sheikh Shohag
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Mirpur 12, Dhaka, 1216, Bangladesh
| | - Jannatul Ferdous
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Shakil Khan Shuvo
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Aklima Akter
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Sarowar Hossain
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Rohit Sharma
- Department of Rasa Shastra & Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
3
|
Caminski ES, Antunes FTT, Souza IA, Dallegrave E, Zamponi GW. Regulation of N-type calcium channels by nociceptin receptors and its possible role in neurological disorders. Mol Brain 2022; 15:95. [PMID: 36434658 PMCID: PMC9700961 DOI: 10.1186/s13041-022-00982-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Activation of nociceptin opioid peptide receptors (NOP, a.k.a. opioid-like receptor-1, ORL-1) by the ligand nociceptin/orphanin FQ, leads to G protein-dependent regulation of Cav2.2 (N-type) voltage-gated calcium channels (VGCCs). This typically causes a reduction in calcium currents, triggering changes in presynaptic calcium levels and thus neurotransmission. Because of the widespread expression patterns of NOP and VGCCs across multiple brain regions, the dorsal horn of the spinal cord, and the dorsal root ganglia, this results in the alteration of numerous neurophysiological features. Here we review the regulation of N-type calcium channels by the NOP-nociceptin system in the context of neurological conditions such as anxiety, addiction, and pain.
Collapse
Affiliation(s)
- Emanuelle Sistherenn Caminski
- grid.412344.40000 0004 0444 6202Graduate Program in Health Sciences, Laboratory of Research in Toxicology (LAPETOX), Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS Brazil
| | - Flavia Tasmin Techera Antunes
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | - Ivana Assis Souza
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | - Eliane Dallegrave
- grid.412344.40000 0004 0444 6202Graduate Program in Health Sciences, Laboratory of Research in Toxicology (LAPETOX), Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS Brazil
| | - Gerald W. Zamponi
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| |
Collapse
|
4
|
Narendra S, Klengel C, Hamzeh B, Patel D, Otten J, Lardenoije R, Newman EL, Miczek KA, Klengel T, Ressler KJ, Suh J. Genome-wide transcriptomics of the amygdala reveals similar oligodendrocyte-related responses to acute and chronic alcohol drinking in female mice. Transl Psychiatry 2022; 12:476. [PMID: 36371333 PMCID: PMC9653459 DOI: 10.1038/s41398-022-02231-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/13/2022] Open
Abstract
Repeated excessive alcohol consumption is a risk factor for alcohol use disorder (AUD). Although AUD has been more common in men than women, women develop more severe behavioral and physical impairments. However, relatively few new therapeutics targeting development of AUD, particularly in women, have been validated. To gain a better understanding of molecular mechanisms underlying alcohol intake, we conducted a genome-wide RNA-sequencing analysis in female mice exposed to different modes (acute vs chronic) of ethanol drinking. We focused on transcriptional profiles in the amygdala including the central and basolateral subnuclei, brain areas previously implicated in alcohol drinking and seeking. Surprisingly, we found that both drinking modes triggered similar changes in gene expression and canonical pathways, including upregulation of ribosome-related/translational pathways and myelination pathways, and downregulation of chromatin binding and histone modification. In addition, analyses of hub genes and upstream regulatory pathways revealed that voluntary ethanol consumption affects epigenetic changes via histone deacetylation pathways, oligodendrocyte and myelin function, and the oligodendrocyte-related transcription factor, Sox17. Furthermore, a viral vector-assisted knockdown of Sox17 gene expression in the amygdala prevented a gradual increase in alcohol consumption during repeated accesses. Overall, these results suggest that the expression of oligodendrocyte-related genes in the amygdala is sensitive to voluntary alcohol drinking in female mice. These findings suggest potential molecular targets for future therapeutic approaches to prevent the development of AUD, due to repeated excessive alcohol consumption, particularly in women.
Collapse
Affiliation(s)
- Sharvari Narendra
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
- Department of Bioinformatics, Northeastern University, Boston, MA, 02115, USA
| | - Claudia Klengel
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
| | - Bilal Hamzeh
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
| | - Drasti Patel
- Department of Bioinformatics, Northeastern University, Boston, MA, 02115, USA
| | - Joy Otten
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Roy Lardenoije
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Emily L Newman
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
| | - Klaus A Miczek
- Psychology and Neuroscience Departments, Tufts University, Medford, MA, 02155, USA
| | - Torsten Klengel
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Kerry J Ressler
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA.
| | - Junghyup Suh
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
5
|
Melkumyan M, Silberman Y. Subregional Differences in Alcohol Modulation of Central Amygdala Neurocircuitry. Front Mol Neurosci 2022; 15:888345. [PMID: 35866156 PMCID: PMC9294740 DOI: 10.3389/fnmol.2022.888345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Alcohol use disorder is a highly significant medical condition characterized by an impaired ability to stop or control alcohol use, compulsive alcohol seeking behavior, and withdrawal symptoms in the absence of alcohol. Understanding how alcohol modulates neurocircuitry critical for long term and binge-like alcohol use, such as the central amygdala (CeA), may lead to the development of novel therapeutic strategies to treat alcohol use disorder. In clinical studies, reduction in the volume of the amygdala has been linked with susceptibility to relapse to alcohol use. Preclinical studies have shown the involvement of the CeA in the effects of alcohol use, with lesions of the amygdala showing a reduction in alcohol drinking, and manipulations of cells in the CeA altering alcohol drinking. A great deal of work has shown that acute alcohol, as well as chronic alcohol exposure via intake or dependence models, alters glutamatergic and GABAergic transmission in the CeA. The CeA, however, contains heterogeneous cell populations and distinct subregional differences in neurocircuit architecture which may influence the mechanism by which alcohol modulates CeA function overall. The current review aimed to parse out the differences in alcohol effects on the medial and lateral subregions of the CeA, and what role neuroinflammatory cells and markers, the endocannabinoid system, and the most commonly studied neuropeptide systems play in mediating these effects. A better understanding of alcohol effects on CeA subregional cell type and neurocircuit function may lead to development of more selective pharmacological interventions for alcohol use disorder.
Collapse
Affiliation(s)
- Mariam Melkumyan
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
6
|
Domi A, Lunerti V, Petrella M, Domi E, Borruto AM, Ubaldi M, Weiss F, Ciccocioppo R. Genetic deletion or pharmacological blockade of nociceptin/orphanin FQ receptors in the ventral tegmental area attenuates nicotine-motivated behaviour. Br J Pharmacol 2022; 179:2647-2658. [PMID: 34854073 PMCID: PMC9081114 DOI: 10.1111/bph.15762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The nociceptin/orphanin FQ (N/OFQ)-nociceptin opioid-like peptide (NOP) receptor system is widely distributed in the brain and pharmacological activation of this system revealed therapeutic potential in animal models of substance use disorder. Studies also showed that genetic deletion or pharmacological blockade of NOP receptors confer resistance to the development of alcohol abuse. Here, we have used a genetic and pharmacological approach to evaluate the therapeutic potential of NOP antagonism in smoking cessation. EXPERIMENTAL APPROACH Constitutive NOP receptor knockout rats (NOP-/- ) and their wild-type counterparts (NOP+/+ ) were tested over a range of behaviours to characterize their motivation for nicotine. We next explored the effects of systemic administration of the NOP receptor antagonist LY2817412 (1.0 & 3.0 mg·kg-1 ) on nicotine self-administration. NOP receptor blockade was further evaluated at the brain circuitry level, by microinjecting LY2817412 (3.0 & 6.0 μg·μl-1 ) into the ventral tegmental area (VTA), nucleus accumbens (NAc) and central amygdala (CeA). KEY RESULTS Genetic NOP receptor deletion resulted in decreased nicotine intake, decreased motivation to self-administer and attenuation of cue-induced nicotine reinstatement. LY2817412 reduced nicotine intake in NOP+/+ but not in NOP-/- rats, confirming that its effect is mediated by inhibition of NOP transmission. Finally, injection of LY2817412 into the VTA but not into the NAc or CeA decreased nicotine self-administration. CONCLUSIONS AND IMPLICATIONS These findings indicate that inhibition of NOP transmission attenuates the motivation for nicotine through mechanisms involving the VTA and suggest that NOP receptor antagonism may represent a potential treatment for smoking cessation.
Collapse
Affiliation(s)
- Ana Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Veronica Lunerti
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Michele Petrella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Esi Domi
- Center for Social and Affective Neuroscience, Institute for Clinical and Experimental Medicine, Linkoping University, Linkoping 58183, Sweden
| | - Anna Maria Borruto
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
7
|
Wang Y, Qin D, Guo Z, Shi F, Cannella N, Ciccocioppo R, Li H. Research progress on the potential novel analgesic BU08028. Eur J Pharmacol 2022; 914:174678. [PMID: 34875275 DOI: 10.1016/j.ejphar.2021.174678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/03/2022]
Abstract
Pain is a common symptom accompanying several clinical conditions and causes serious distress to patients. Addressing pain management is an important aspect of disease treatment, including cancer therapy. Opioid analgesics used to manage pain in human and veterinary medicine have been associated with substance dependence and other adverse effects, thereby limiting their application. Thus, the development of opioid analgesics with good safety profiles with minimal adverse effects and no addictive effects, is presently the focus of pain research. As a new potential analgesic, (2S)-2-[(5R,6R,7R,14S)-N-cyclopropylmethyl-4,5-epoxy-6,14-ethano-3-hydroxy-6-methoxymorphinan-7-yl]-3,3-dimethylpentan-2-ol (BU08028) has fewer adverse effects than other analgesics and is expected to be a safer alternative. In this review, we discuss the development of the opioid analog BU08028 and summarize its analgesic effects and biological characteristics, including efficiency, safety, and tolerance. Furthermore, we elaborate on studies showing the bifunctional effect of BU08028, which targets both mu opioid peptide and nociceptin-orphanin FQ peptide receptors, as well as the unique advantages of using BU08028 over single-target opioid agonists. Previous studies have suggested that BU08028 can not only weaken the reward and abuse effects of opioids and other drugs, but also enhance the anti-nociceptive effect of the mu opioid peptide receptors, making it a potent analgesic. Besides, we describe studies suggesting that BU08028 inhibits the effects of alcohol, making it a candidate drug for the management of alcohol addiction. Our review suggests that BU08028 is a potential novel medicine for managing pain and addiction.
Collapse
Affiliation(s)
- Ya Wang
- School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, China
| | - Di Qin
- China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Zhihua Guo
- School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, China
| | - Fuqiang Shi
- School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, China
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, Camerino, 62032, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, Camerino, 62032, Italy
| | - Hongwu Li
- School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, China.
| |
Collapse
|
8
|
Ubaldi M, Cannella N, Borruto AM, Petrella M, Micioni Di Bonaventura MV, Soverchia L, Stopponi S, Weiss F, Cifani C, Ciccocioppo R. Role of Nociceptin/Orphanin FQ-NOP Receptor System in the Regulation of Stress-Related Disorders. Int J Mol Sci 2021; 22:12956. [PMID: 34884757 PMCID: PMC8657682 DOI: 10.3390/ijms222312956] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022] Open
Abstract
Nociceptin/orphanin FQ (N/OFQ) is a 17-residue neuropeptide that binds the nociceptin opioid-like receptor (NOP). N/OFQ exhibits nucleotidic and aminoacidics sequence homology with the precursors of other opioid neuropeptides but it does not activate either MOP, KOP or DOP receptors. Furthermore, opioid neuropeptides do not activate the NOP receptor. Generally, activation of N/OFQ system exerts anti-opioids effects, for instance toward opioid-induced reward and analgesia. The NOP receptor is widely expressed throughout the brain, whereas N/OFQ localization is confined to brain nuclei that are involved in stress response such as amygdala, BNST and hypothalamus. Decades of studies have delineated the biological role of this system demonstrating its involvement in significant physiological processes such as pain, learning and memory, anxiety, depression, feeding, drug and alcohol dependence. This review discusses the role of this peptidergic system in the modulation of stress and stress-associated psychiatric disorders in particular drug addiction, mood, anxiety and food-related associated-disorders. Emerging preclinical evidence suggests that both NOP agonists and antagonists may represent a effective therapeutic approaches for substances use disorder. Moreover, the current literature suggests that NOP antagonists can be useful to treat depression and feeding-related diseases, such as obesity and binge eating behavior, whereas the activation of NOP receptor by agonists could be a promising tool for anxiety.
Collapse
Affiliation(s)
- Massimo Ubaldi
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Nazzareno Cannella
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Anna Maria Borruto
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Michele Petrella
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Maria Vittoria Micioni Di Bonaventura
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Laura Soverchia
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Serena Stopponi
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA;
| | - Carlo Cifani
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Roberto Ciccocioppo
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| |
Collapse
|
9
|
Borruto AM, Fotio Y, Stopponi S, Petrella M, De Carlo S, Domi A, Ubaldi M, Weiss F, Ciccocioppo R. NOP receptor antagonism attenuates reinstatement of alcohol-seeking through modulation of the mesolimbic circuitry in male and female alcohol-preferring rats. Neuropsychopharmacology 2021; 46:2121-2131. [PMID: 34285372 PMCID: PMC8505627 DOI: 10.1038/s41386-021-01096-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023]
Abstract
In patients suffering from alcohol use disorder (AUD), stress and environmental stimuli associated with alcohol availability are important triggers of relapse. Activation of the nociceptin opioid peptide (NOP) receptor by its endogenous ligand Nociceptin/Orphanin FQ (N/OFQ) attenuates alcohol drinking and relapse in rodents, suggesting that NOP agonists may be efficacious in treating AUD. Intriguingly, recent data demonstrated that also blockade of NOP receptor reduced alcohol drinking in rodents. To explore further the potential of NOP antagonism, we investigated its effects on the reinstatement of alcohol-seeking elicited by administration of the α2 antagonist yohimbine (1.25 mg/kg, i.p.) or by environmental conditioning factors in male and female genetically selected alcohol-preferring Marchigian Sardinian (msP) rats. The selective NOP receptor antagonist LY2817412 (0.0, 3.0, 10.0, and 30.0 mg/kg) was first tested following oral (p.o.) administration. We then investigated the effects of LY2817412 (1.0, 3.0, 6.0 μg/μl/rat) microinjected into three candidate mesolimbic brain regions: the ventral tegmental area (VTA), the central nucleus of the amygdala (CeA), and the nucleus accumbens (NAc). We found that relapse to alcohol seeking was generally stronger in female than in male rats and oral administration of LY2817412 reduced yohimbine- and cue-induced reinstatement in both sexes. Following site-specific microinjections, LY2817412 reduced yohimbine-induced reinstatement of alcohol-seeking when administered into the VTA and the CeA, but not in the NAc. Cue-induced reinstatement was suppressed only when LY2817412 was microinjected into the VTA. Infusions of LY2817412 into the VTA and the CeA did not alter saccharin self-administration. These results demonstrate that NOP receptor blockade prevents the reinstatement of alcohol-seeking through modulation of mesolimbic system circuitry, providing further evidence of the therapeutic potential of NOP receptor antagonism in AUD.
Collapse
Affiliation(s)
- Anna Maria Borruto
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Yannick Fotio
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Michele Petrella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Sara De Carlo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Ana Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy.
| |
Collapse
|
10
|
Effects of datumetine on hippocampal NMDAR activity. Toxicol Rep 2021; 8:1131-1142. [PMID: 34150523 PMCID: PMC8190477 DOI: 10.1016/j.toxrep.2021.05.009] [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: 10/15/2020] [Revised: 03/16/2021] [Accepted: 05/21/2021] [Indexed: 11/20/2022] Open
Abstract
The usage (abuse) of Datura metel is becoming increasingly worrisome among the Nigerian populace especially among the youth considering its side effects such as hallucination. This work was designed to identify the phytochemicals in datura plant that potentially interact with NMDAR as it affects the electrical and memory activities of the brain. Ligand-protein interaction was assessed using autodock vina to identify phytochemicals that can interact with NMDAR. Datumetine was found to have the best interaction fit with NMDAR at both allosteric and orthosteric binding sites. Furthermore, using electrophysiological, behavioural and western blotting techniques, it was observed that the administration of datumetine positively modulates the NMDAR current by prolonging burst duration and interspike interval, induces seizures in C57BL/6 mice. Acute exposure leads to memory deficit on NOR and Y-maze test while immunoblotting results showed increased expression of GluN1 and CamKIIα while pCamKIIα-T286, CREB and BDNF were downregulated. The results showed that the memory deficit seen in datura intoxication is possibly the effects of datumetine on NMDAR.
Collapse
|
11
|
Walker LC. A balancing act: the role of pro- and anti-stress peptides within the central amygdala in anxiety and alcohol use disorders. J Neurochem 2021; 157:1615-1643. [PMID: 33450069 DOI: 10.1111/jnc.15301] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/18/2020] [Accepted: 01/06/2021] [Indexed: 12/21/2022]
Abstract
The central nucleus of the amygdala (CeA) is widely implicated as a structure that integrates both appetitive and aversive stimuli. While intrinsic CeA microcircuits primarily consist of GABAergic neurons that regulate amygdala output, a notable feature of the CeA is the heterogeneity of neuropeptides and neuropeptide/neuromodulator receptors that it expresses. There is growing interest in the role of the CeA in mediating psychopathologies, including stress and anxiety states and their interactions with alcohol use disorders. Within the CeA, neuropeptides and neuromodulators often exert pro- or anti- stress actions, which can influence anxiety and alcohol associated behaviours. In turn, alcohol use can cause adaptions within the CeA, which may render an individual more vulnerable to stress which is a major trigger of relapse to alcohol seeking. This review examines the neurocircuitry, neurochemical phenotypes and how pro- and anti-stress peptide systems act within the CeA to regulate anxiety and alcohol seeking, focusing on preclinical observations from animal models. Furthermore, literature exploring the targeting of genetically defined populations or neuronal ensembles and the role of the CeA in mediating sex differences in stress x alcohol interactions are explored.
Collapse
Affiliation(s)
- Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic, Australia
| |
Collapse
|
12
|
Abstract
Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug-seeking and drug-taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central nucleus of the amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptive mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid (GABA)ergic transmission in the CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the CeA, whereas chronic alcohol up-regulates NMDA receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and antistress (e.g., nociceptin/orphanin FQ, oxytocin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.
Collapse
Affiliation(s)
- Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Dean Kirson
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Sophia Khom
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
13
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
14
|
Hedges DM, Yorgason JT, Brundage JN, Wadsworth HA, Williams B, Steffensen SC, Roberto M. Corticotropin releasing factor, but not alcohol, modulates norepinephrine release in the rat central nucleus of the amygdala. Neuropharmacology 2020; 179:108293. [PMID: 32871155 DOI: 10.1016/j.neuropharm.2020.108293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/20/2022]
Abstract
Alcohol misuse and dependence is a widespread health problem. The central nucleus of the amygdala (CeA) plays important roles in both the anxiety associated with alcohol (ethanol) dependence and the increased alcohol intake that is observed during withdrawal in dependent animals. We and others have shown the essential involvement of the corticotropin releasing factor (CRF) system in alcohol's synaptic effects on the CeA and in the development of ethanol dependence. Another system that has been shown to be critically involved in the molecular underpinnings of alcohol dependence is the norepinephrine (NE) system originating in the locus coeruleus. Both the CRF and NE systems act in concert to facilitate a stress response: central amygdalar afferents release CRF in the locus coeruleus promoting widespread release of NE. In this study, we are the first to use fast-scan cyclic voltammetry to classify local electrically-evoked NE release in the CeA and to determine if acute alcohol and CRF modulate it. Evoked NE release is action potential dependent, is abolished after depletion of monoaminergic vesicles, differs pharmacologically from dopamine release, is insensitive to acute alcohol, and decreases in response to locally applied CRF. Taken together, these results indicate that NE release in the CeA is released canonically in a vesicular-dependent manner, and that while acute alcohol does not directly alter NE release, CRF decreases it. Our results suggest that CRF acts locally on NE terminals as negative feedback and potentially prevents hyperactivation of the CRF-norepinephrine stress pathway.
Collapse
Affiliation(s)
- David M Hedges
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA.
| | - Jordan T Yorgason
- Neuroscience Program, Brigham Young University, Provo, UT, 84602, USA.
| | - James N Brundage
- Neuroscience Program, Brigham Young University, Provo, UT, 84602, USA
| | | | - Benjamin Williams
- Neuroscience Program, Brigham Young University, Provo, UT, 84602, USA
| | | | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
15
|
Acute Alcohol Effects on Response Inhibition Depend on Response Automatization, but not on GABA or Glutamate Levels in the ACC and Striatum. J Clin Med 2020; 9:jcm9020481. [PMID: 32050509 PMCID: PMC7073826 DOI: 10.3390/jcm9020481] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/01/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
Alcohol increases GABAergic signaling and decreases glutamatergic signaling in the brain. Variations in these neurotransmitter levels may modulate/predict executive functioning. Matching this, strong impairments of response inhibition are one of the most consistently reported cognitive/behavioral effects of acute alcohol intoxication. However, it has never been investigated whether baseline differences in these neurotransmitters allow to predict how much alcohol intoxication impairs response inhibition, and whether this is reflected in neurophysiological measures of cognitive control. We used MR spectroscopy to assess baseline (i.e., sober) GABA and glutamate levels in the anterior cingulate cortex (ACC) and striatum in n = 30 healthy young males, who were subsequently tested once sober and once intoxicated (1.01 permille). Inhibition was assessed with the sustained attention to response task (SART). This paradigm also allows to examine the effect of different degrees of response automatization, which is a known modulator for response inhibition, but does not seem to be substantially impaired during acute intoxication. As a neurophysiological correlate of response inhibition and control, we quantified EEG-derived theta band power and located its source using beamforming analyses. We found that alcohol-induced response inhibition deficits only occurred in the case of response automatization. This was reflected by decreased theta band activity in the left supplementary motor area (SMA), which may reflect modulations in the encoding of a surprise signal in response to inhibition cues. However, we did not find that differences in baseline (i.e., sober) GABA or glutamate levels significantly modulated differences in the size of alcohol-induced inhibition deficits.
Collapse
|
16
|
Borruto AM, Fotio Y, Stopponi S, Brunori G, Petrella M, Caputi FF, Romualdi P, Candeletti S, Narendran R, Rorick-Kehn LM, Ubaldi M, Weiss F, Ciccocioppo R. NOP receptor antagonism reduces alcohol drinking in male and female rats through mechanisms involving the central amygdala and ventral tegmental area. Br J Pharmacol 2020; 177:1525-1537. [PMID: 31713848 DOI: 10.1111/bph.14915] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Nociceptin/orphanin FQ (N/OFQ) peptide and its cognate receptor (NOP) are widely expressed in mesolimbic brain regions where they play an important role in modulating reward and motivation. Early evidence suggested that NOP receptor activation attenuates the rewarding effects of drugs of abuse, including alcohol. However, emerging data indicate that NOP receptor blockade also effectively attenuates alcohol drinking and relapse. To advance our understanding of the role of the N/OFQ-NOP receptor system in alcohol abuse, we examined the effect of NOP receptor blockade on voluntary alcohol drinking at the neurocircuitry level. EXPERIMENTAL APPROACH Using male and female genetically selected alcohol-preferring Marchigian Sardinian (msP) rats, we initially evaluated the effects of the selective NOP receptor antagonist LY2817412 (3, 10, and 30 mg·kg-1 , p.o.) on alcohol consumption in a two-bottle free-choice paradigm. We then microinjected LY2817412 (3 and 6 μg·μl-1 per rat) in the central nucleus of the amygdala (CeA), ventral tegmental area (VTA), and nucleus accumbens (NAc). KEY RESULTS Peripheral LY2817412 administration dose-dependently and selectively reduced voluntary alcohol intake in male and female msP rats. Central injections of LY2817412 markedly attenuated voluntary alcohol intake in both sexes following administration in the CeA and VTA but not in the NAc. CONCLUSION AND IMPLICATIONS The present results revealed that the CeA and VTA are neuroanatomical substrates that mediate the effects of NOP receptor antagonism on alcohol consumption. Overall, our findings support the potential of NOP receptor antagonism as a treatment strategy to attenuate alcohol use and addiction.
Collapse
Affiliation(s)
| | - Yannick Fotio
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Gloria Brunori
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy.,Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Michele Petrella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Francesca Felicia Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Linda M Rorick-Kehn
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
17
|
Kirson D, Oleata CS, Roberto M. Taurine Suppression of Central Amygdala GABAergic Inhibitory Signaling via Glycine Receptors Is Disrupted in Alcohol Dependence. Alcohol Clin Exp Res 2019; 44:445-454. [PMID: 31782155 DOI: 10.1111/acer.14252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/18/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) increases brain stress systems while suppressing reward system functioning. One expression of stress system recruitment is elevated GABAergic activity in the central amygdala (CeA), which is involved in the excessive drinking seen with AUD. The sulfonic amino acid taurine, a glycine receptor partial agonist, modulates GABAergic activity in the rewarding effects of alcohol. Despite taurine abundance in the amygdala, its role in the dysregulation of GABAergic activity associated with AUD has not been studied. Thus, here, we evaluated the effects of taurine on locally stimulated GABAergic neurotransmission in the CeA of naïve- and alcohol-dependent rats. METHODS We recorded intracellularly from CeA neurons of naïve- and alcohol-dependent rats, quantifying locally evoked GABAA receptor-mediated inhibitory postsynaptic potentials (eIPSP). We examined the effects of taurine and alcohol on CeA eIPSP to characterize potential alcohol dependence-induced changes in the effects of taurine. RESULTS We found that taurine decreased amplitudes of eIPSP in CeA neurons of naïve rats, without affecting the acute alcohol-induced facilitation of GABAergic responses. In CeA neurons from dependent rats, taurine no longer had an effect on eIPSP, but now blocked the ethanol (EtOH)-induced increase in eIPSP amplitude normally seen. Additionally, preapplication of the glycine receptor-specific antagonist strychnine blocked the EtOH-induced increase in eIPSP amplitude in neurons from naïve rats. CONCLUSIONS These data suggest taurine may act to oppose the effects of acute alcohol via the glycine receptor in the CeA of naïve rats, and this modulatory system is altered in the CeA of dependent rats.
Collapse
Affiliation(s)
- Dean Kirson
- Department of Molecular Medicine, and Alcohol Research Center, The Scripps Research Institute, La Jolla, California
| | - Christopher S Oleata
- Department of Molecular Medicine, and Alcohol Research Center, The Scripps Research Institute, La Jolla, California
| | - Marisa Roberto
- Department of Molecular Medicine, and Alcohol Research Center, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
18
|
Centanni SW, Bedse G, Patel S, Winder DG. Driving the Downward Spiral: Alcohol-Induced Dysregulation of Extended Amygdala Circuits and Negative Affect. Alcohol Clin Exp Res 2019; 43:2000-2013. [PMID: 31403699 PMCID: PMC6779502 DOI: 10.1111/acer.14178] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
Alcohol use disorder (AUD) afflicts a large number of individuals, families, and communities globally. Affective disturbances, including stress, depression, and anxiety, are highly comorbid with AUD, contributing in some cases to initial alcohol use and continued use. Negative affect has a particularly strong influence on the withdrawal/abstinence stage of addiction as individuals with AUD frequently report stressful events, depression, and anxiety as key factors for relapse. Treatment options for negative affect associated with AUD are limited and often ineffective, highlighting the pressing need for preclinical studies examining the underlying neural circuitry driving AUD-associated negative affect. The extended amygdala (EA) is a set of brain areas collectively involved in generating and regulating affect, and extensive research has defined a critical role for the EA in all facets of substance use disorder. Here, we review the expansive historical literature examining the effects of ethanol exposure on the EA, with an emphasis on the complex EA neural circuitry driving negative affect in all phases of the alcohol addiction cycle. Specifically, this review focuses on the effects of alcohol exposure on the neural circuitry in 2 key components of the EA, the central nucleus of the amygdala and the bed nucleus of the stria terminalis. Additionally, future directions are proposed to advance our understanding of the relationship between AUD-associated negative affect and neural circuitry in the EA, with the long-term goal of developing better diagnostic tools and new pharmacological targets aimed at treating negative affect in AUD. The concepts detailed here will serve as the foundation for a companion review focusing on the potential for the endogenous cannabinoid system in the EA as a novel target for treating the stress, anxiety, and negative emotional state driving AUD.
Collapse
Affiliation(s)
- Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
| | - Gaurav Bedse
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Sachin Patel
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| |
Collapse
|
19
|
Bensmann W, Kayali ÖF, Beste C, Stock AK. Young frequent binge drinkers show no behavioral deficits in inhibitory control and cognitive flexibility. Prog Neuropsychopharmacol Biol Psychiatry 2019; 93:93-101. [PMID: 30946938 DOI: 10.1016/j.pnpbp.2019.03.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/15/2019] [Accepted: 03/29/2019] [Indexed: 11/30/2022]
Abstract
Alcohol intoxication and abuse are well-known to cause impairments in executive functioning and control. Still, we know surprisingly little about individuals engaging in frequent binge drinking, even though they have an increased risk to develop an alcohol use disorder (AUD) later in life. As this risk has been suggested to be linked to (premorbid) executive deficits, we assessed changes in cognitive flexibility and inhibition with the help of a switching task and a stop-change task. Both paradigms had previously been shown to be modulated by alcohol, as well as by functional variations in dopaminergic and GABAergic neurotransmission. We employed an extreme group approach, where we compared pre-selected samples of frequent binge drinkers and non-frequent binge drinkers, all of which had stably pursued their respective consumption pattern for at least 3 years. In combination with Bayes analyses, our results showed that individuals engaging in frequent binge drinking showed no impairments of cognitive flexibility or inhibition, as compared to non-frequent binge drinkers. These observations suggest that frequent binge drinking alone is not associated with the cognitive control deficits commonly observed in AUD. Importantly, the investigated executive functions are known to be altered both during binge drinking and in individuals with AUD. It could hence be speculated that their intermittent consumption pattern prevents non-AUD frequent binge drinkers from the homeostatic counter-regulations of alcohol- and control-associated neurotransmitter systems that may be observed in AUD patients. Yet, this hypothesis still needs to be tested in future research, including studies that combine MR and molecular imaging.
Collapse
Affiliation(s)
- Wiebke Bensmann
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Özlem Feray Kayali
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Ann-Kathrin Stock
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany.
| |
Collapse
|
20
|
Abstract
Ethanol produces intoxication through actions on numerous molecular and cellular targets. Adaptations involving these and other targets contribute to chronic drug actions that underlie continued and problematic drinking. Among the mechanisms involved in these ethanol actions are alterations in presynaptic mechanisms of synaptic transmission, including presynaptic protein function and excitation-secretion coupling. At synapses in the central nervous system (CNS), excitation-secretion coupling involves ion channel activation followed by vesicle fusion and neurotransmitter release. These mechanisms are altered by presynaptic neurotransmitter receptors and prominently by G protein-coupled receptors (GPCRs). Studies over the last 20-25 years have revealed that acute ethanol exposure alters neurotransmitter secretion, with especially robust effects on synapses that use the neurotransmitter gamma-aminobutyric acid (GABA). Intracellular signaling pathways involving second messengers such as cyclic AMP and calcium are implicated in these acute ethanol actions. Ethanol-induced release of neuropeptides and small molecule neurotransmitters that act on presynaptic GPCRs also contribute to presynaptic potentiation at synapses in the amygdala and hippocampus and inhibition of GABA release in the striatum. Prolonged exposure to ethanol alters neurotransmitter release at many CNS GABAergic and glutamatergic synapses, and changes in GPCR function are implicated in many of these neuroadaptations. These presynaptic neuroadaptations appear to involve compensation for acute drug effects at some synapses, but "allostatic" effects that result in long-term resetting of synaptic efficacy occur at others. Current investigations are determining how presynaptic neuroadaptations contribute to behavioral changes at different stages of alcohol drinking, with increasing focus on circuit adaptations underlying these behaviors. This chapter will discuss the acute and chronic presynaptic effects of ethanol in the CNS, as well as some of the consequences of these effects in amygdala and corticostriatal circuits that are related to excessive seeking/drinking and ethanol abuse.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| |
Collapse
|
21
|
Li H, Hu B, Zhang HP, Boyle CA, Lei S. Roles of K + and cation channels in ORL-1 receptor-mediated depression of neuronal excitability and epileptic activities in the medial entorhinal cortex. Neuropharmacology 2019; 151:144-158. [PMID: 30998945 PMCID: PMC6500758 DOI: 10.1016/j.neuropharm.2019.04.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/24/2019] [Accepted: 04/13/2019] [Indexed: 02/05/2023]
Abstract
Nociceptin (NOP) is an endogenous opioid-like peptide that selectively activates the opioid receptor-like (ORL-1) receptors. The entorhinal cortex (EC) is closely related to temporal lobe epilepsy and expresses high densities of ORL-1 receptors. However, the functions of NOP in the EC, especially in modulating the epileptiform activity in the EC, have not been determined. We demonstrated that activation of ORL-1 receptors remarkably inhibited the epileptiform activity in entorhinal slices induced by application of picrotoxin or by deprivation of extracellular Mg2+. NOP-mediated depression of epileptiform activity was independent of synaptic transmission in the EC, but mediated by inhibition of neuronal excitability in the EC. NOP hyperpolarized entorhinal neurons via activation of K+ channels and inhibition of cation channels. Whereas application of Ba2+ at 300 μM which is effective for the inward rectifier K+ (Kir) channels slightly inhibited NOP-induced hyperpolarization, the current-voltage (I-V) curve of the net currents induced by NOP was linear without showing inward rectification. However, a role of NOP-induced inhibition of cation channels was revealed after inhibition of Kir channels by Ba2+. Furthermore, NOP-mediated augmentation of membrane currents was differently affected by application of the blockers selective for distinct subfamilies of Kir channels. Whereas SCH23390 or ML133 blocked NOP-induced augmentation of membrane currents at negative potentials, application of tertiapin-Q exerted no actions on NOP-induced alteration of membrane currents. Our results demonstrated a novel cellular and molecular mechanism whereby activation of ORL-1 receptors depresses epilepsy.
Collapse
Affiliation(s)
- Huiming Li
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Binqi Hu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Hao-Peng Zhang
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Cody A Boyle
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Saobo Lei
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA.
| |
Collapse
|
22
|
Abstract
Nociceptin/orphanin FQ (N/OFQ) is an endogenous neuropeptide of 17 amino acids, related to opioid peptides but with its own receptor, distinct from conventional opioid receptors, the ORL1 or NOP receptor. The NOP receptor is a G protein-coupled receptor which activates Gi/o proteins and thus induces an inhibition of neuronal activity. The peptide and its receptor are widely expressed in the central nervous system with a high density of receptors in regions involved in learning and memory. This review describes the consequences of the pharmacological manipulation of the N/OFQ system by NOP receptor ligands on learning processes and on the consolidation of various types of long-term memory. We also discuss the role of endogenous N/OFQ release in the modulation of learning and memory. Finally we propose several putative neuronal mechanisms taking place at the level of the hippocampus and amygdala and possibly underlying the behavioral amnestic or promnesic effects of NOP ligands.
Collapse
Affiliation(s)
- Lionel Moulédous
- Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France.
| |
Collapse
|
23
|
Ciccocioppo R, Borruto AM, Domi A, Teshima K, Cannella N, Weiss F. NOP-Related Mechanisms in Substance Use Disorders. Handb Exp Pharmacol 2019; 254:187-212. [PMID: 30968214 PMCID: PMC6641545 DOI: 10.1007/164_2019_209] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Nociceptin/orphanin FQ (N/OFQ) is a 17 amino acid peptide that was deorphanized in 1995 and has been widely studied since. The role of the N/OFQ system in drug abuse has attracted researchers' attention since its initial discovery. The first two scientific papers describing the effect of intracranial injection of N/OFQ appeared 20 years ago and reported efficacy of the peptide in attenuating alcohol intake, whereas heroin self-administration was insensitive. Since then more than 100 scientific articles investigating the role of the N/OFQ and N/OFQ receptor (NOP) system in drug abuse have been published. The present article provides an historical overview of the advances in the field with focus on three major elements. First, the most robust data supportive of the efficacy of NOP agonists in treating drug abuse come from studies in the field of alcohol research, followed by psychostimulant and opioid research. In contrast, activation of NOP appears to facilitate nicotine consumption. Second, emerging data challenge the assumption that activation of NOP is the most appropriate strategy to attenuate consumption of substances of abuse. This assumption is based first on the observation that animals carrying an overexpression of NOP system components are more prone to consume substances of abuse, whereas NOP knockout rats are less motivated to self-administer heroin, alcohol, and cocaine. Third, administration of NOP antagonists also reduces alcohol consumption. In addition, NOP blockade reduces nicotine self-administration. Hypothetical mechanisms explaining this apparent paradox are discussed. Finally, we focus on the possibility that co-activation of NOP and mu opioid (MOP) receptors is an alternative strategy, readily testable in the clinic, to reduce the consumption of psychostimulants, opiates, and, possibly, alcohol.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy.
| | - Anna Maria Borruto
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Ana Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Koji Teshima
- Research Unit/Neuroscience, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Friedbert Weiss
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
24
|
Abstract
Whilst the nociceptin/orphanin FQ (N/OFQ) receptor (NOP) has similar intracellular coupling mechanisms to opioid receptors, it has distinct modulatory effects on physiological functions such as pain. These actions range from agonistic to antagonistic interactions with classical opioids within the spinal cord and brain, respectively. Understanding the electrophysiological actions of N/OFQ has been crucial in ascertaining the mechanisms by which these agonistic and antagonistic interactions occur. These similarities and differences between N/OFQ and opioids are due to the relative location of NOP versus opioid receptors on specific neuronal elements within these CNS regions. These mechanisms result in varied cellular actions including postsynaptic modulation of ion channels and presynaptic regulation of neurotransmitter release.
Collapse
|
25
|
Abstract
Animal models provide rapid, inexpensive assessments of an investigational drug's therapeutic potential. Ideally, they support the plausibility of therapeutic efficacy and provide a rationale for further investigation. Here, I discuss how the absence of clear effective-ineffective categories for alcohol use disorder (AUD) medications and biases in the clinical and preclinical literature affect the development of predictive preclinical alcohol dependence (AD) models. Invoking the analogical argument concept from the philosophy of science field, I discuss how models of excessive alcohol drinking support the plausibility of clinical pharmacotherapy effects. Even though these models are not likely be completely discriminative, they are sensitive to clinically effective medications and have revealed dozens of novel medication targets. In that context, I discuss recent preclinical work on GLP-1 receptor agonists, phosphodiesterase inhibitors, glucocorticoid receptor antagonists, nociception agonists and antagonists, and CRF1 antagonists. Clinically approved medications are available for each of these drug classes. I conclude by advocating a translational approach in which drugs are evaluated highly congruent preclinical models and human laboratory studies. Once translation is established, I suggest the burden is to develop hypothesis-based therapeutic interventions maximizing the impact of the confirmed pharmacotherapeutic effects in the context of additional variables falling outside the model.
Collapse
Affiliation(s)
- Mark Egli
- Division of Neuroscience and Behavior, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
26
|
Varodayan FP, Correia D, Kirson D, Khom S, Oleata CS, Luu G, Schweitzer P, Roberto M. CRF modulates glutamate transmission in the central amygdala of naïve and ethanol-dependent rats. Neuropharmacology 2017; 125:418-428. [PMID: 28807676 DOI: 10.1016/j.neuropharm.2017.08.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/21/2017] [Accepted: 08/09/2017] [Indexed: 11/15/2022]
Abstract
Corticotropin-releasing factor (CRF) signaling in the central nucleus of the amygdala (CeA) is hypothesized to drive the development of alcohol dependence, as it regulates ethanol intake and several anxiogenic behaviors linked to withdrawal. Excitatory glutamatergic neurotransmission contributes to alcohol reinforcement, tolerance and dependence. Therefore, in this study we used in vitro slice electrophysiology to investigate the effects of CRF and its receptor subtype (CRF1 and CRF2) antagonists on both evoked and spontaneous action potential-independent glutamatergic transmission in the CeA of naive and ethanol-dependent Sprague-Dawley rats. We found that CRF (25-200 nM) concentration-dependently diminished evoked compound excitatory postsynaptic potentials (EPSPs), but increased miniature excitatory postsynaptic current (mEPSC) frequencies similarly in CeA neurons of both naïve and ethanol-dependent rats, indicating reduced evoked glutamatergic responses and enhanced vesicular glutamate release, respectively. This CRF-induced vesicular glutamate release was prevented by the CRF1/2 antagonist (Astressin B) and the CRF1 antagonist (R121919), but not by the CRF2 antagonist (Astressin 2B). Similarly, CRF's effects on evoked glutamatergic responses were completely blocked by CRF1 antagonism, but only slightly decreased in the presence of the CRF2 antagonist. Moreover, CRF1 antagonism reveals a tonic facilitation of vesicular glutamate, whereas the CRF2 antagonism revealed a tonic inhibition of vesicular glutamate release. Collectively our data show that CRF primarily acts at presynaptic CRF1 to produce opposite effects on CeA evoked and spontaneous glutamate release and that the CRF system modulates CeA glutamatergic synapses throughout the development of alcohol dependence.
Collapse
Affiliation(s)
| | - Diego Correia
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA; Department of Pharmacology, Universidade Federal do Paraná, Jardim das Américas, Curitiba, Paraná, CEP 81531-990, Brazil
| | - Dean Kirson
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Sophia Khom
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | | | - George Luu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Paul Schweitzer
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
27
|
Epigenetic mechanisms of alcoholism and stress-related disorders. Alcohol 2017; 60:7-18. [PMID: 28477725 DOI: 10.1016/j.alcohol.2017.01.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/30/2016] [Accepted: 01/03/2017] [Indexed: 12/20/2022]
Abstract
Stress-related disorders, such as anxiety, early life stress, and posttraumatic stress disorder appear to be important factors in promoting alcoholism, as alcohol consumption can temporarily attenuate the negative affective symptoms of these disorders. Several molecules involved in signaling pathways may contribute to the neuroadaptation induced during alcohol dependence and stress disorders, and among these, brain-derived neurotrophic factor (BDNF), corticotropin releasing factor (CRF), neuropeptide Y (NPY) and opioid peptides (i.e., nociceptin and dynorphin) are involved in the interaction of stress and alcohol. In fact, alterations in the expression and function of these molecules have been associated with the pathophysiology of stress-related disorders and alcoholism. In recent years, various studies have focused on the epigenetic mechanisms that regulate chromatin architecture, thereby modifying gene expression. Interestingly, epigenetic modifications in specific brain regions have been shown to be associated with the neurobiology of psychiatric disorders, including alcoholism and stress. In particular, the enzymes responsible for chromatin remodeling (i.e., histone deacetylases and methyltransferases, DNA methyltransferases) have been identified as common molecular mechanisms for the interaction of stress and alcohol and have become promising therapeutic targets to treat or prevent alcoholism and associated emotional disorders.
Collapse
|
28
|
Korpi ER, den Hollander B, Farooq U, Vashchinkina E, Rajkumar R, Nutt DJ, Hyytiä P, Dawe GS. Mechanisms of Action and Persistent Neuroplasticity by Drugs of Abuse. Pharmacol Rev 2015; 67:872-1004. [PMID: 26403687 DOI: 10.1124/pr.115.010967] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Adaptation of the nervous system to different chemical and physiologic conditions is important for the homeostasis of brain processes and for learning and remembering appropriate responses to challenges. Although processes such as tolerance and dependence to various drugs of abuse have been known for a long time, it was recently discovered that even a single pharmacologically relevant dose of various drugs of abuse induces neuroplasticity in selected neuronal populations, such as the dopamine neurons of the ventral tegmental area, which persist long after the drug has been excreted. Prolonged (self-) administration of drugs induces gene expression, neurochemical, neurophysiological, and structural changes in many brain cell populations. These region-specific changes correlate with addiction, drug intake, and conditioned drugs effects, such as cue- or stress-induced reinstatement of drug seeking. In rodents, adolescent drug exposure often causes significantly more behavioral changes later in adulthood than a corresponding exposure in adults. Clinically the most impairing and devastating effects on the brain are produced by alcohol during fetal development. In adult recreational drug users or in medicated patients, it has been difficult to find persistent functional or behavioral changes, suggesting that heavy exposure to drugs of abuse is needed for neurotoxicity and for persistent emotional and cognitive alterations. This review describes recent advances in this important area of research, which harbors the aim of translating this knowledge to better treatments for addictions and related neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.R.K., B.d.H., E.V., P.H.); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, and SINAPSE, Singapore Institute for Neurotechnology, Singapore (E.R.K., R.R., G.S.D.); Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut (U.F.); and Centre for Neuropsychopharmacology, Division of Brain Sciences, Burlington Danes Building, Imperial College London, London. United Kingdom (D.J.N.)
| | - Bjørnar den Hollander
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.R.K., B.d.H., E.V., P.H.); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, and SINAPSE, Singapore Institute for Neurotechnology, Singapore (E.R.K., R.R., G.S.D.); Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut (U.F.); and Centre for Neuropsychopharmacology, Division of Brain Sciences, Burlington Danes Building, Imperial College London, London. United Kingdom (D.J.N.)
| | - Usman Farooq
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.R.K., B.d.H., E.V., P.H.); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, and SINAPSE, Singapore Institute for Neurotechnology, Singapore (E.R.K., R.R., G.S.D.); Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut (U.F.); and Centre for Neuropsychopharmacology, Division of Brain Sciences, Burlington Danes Building, Imperial College London, London. United Kingdom (D.J.N.)
| | - Elena Vashchinkina
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.R.K., B.d.H., E.V., P.H.); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, and SINAPSE, Singapore Institute for Neurotechnology, Singapore (E.R.K., R.R., G.S.D.); Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut (U.F.); and Centre for Neuropsychopharmacology, Division of Brain Sciences, Burlington Danes Building, Imperial College London, London. United Kingdom (D.J.N.)
| | - Ramamoorthy Rajkumar
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.R.K., B.d.H., E.V., P.H.); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, and SINAPSE, Singapore Institute for Neurotechnology, Singapore (E.R.K., R.R., G.S.D.); Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut (U.F.); and Centre for Neuropsychopharmacology, Division of Brain Sciences, Burlington Danes Building, Imperial College London, London. United Kingdom (D.J.N.)
| | - David J Nutt
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.R.K., B.d.H., E.V., P.H.); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, and SINAPSE, Singapore Institute for Neurotechnology, Singapore (E.R.K., R.R., G.S.D.); Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut (U.F.); and Centre for Neuropsychopharmacology, Division of Brain Sciences, Burlington Danes Building, Imperial College London, London. United Kingdom (D.J.N.)
| | - Petri Hyytiä
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.R.K., B.d.H., E.V., P.H.); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, and SINAPSE, Singapore Institute for Neurotechnology, Singapore (E.R.K., R.R., G.S.D.); Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut (U.F.); and Centre for Neuropsychopharmacology, Division of Brain Sciences, Burlington Danes Building, Imperial College London, London. United Kingdom (D.J.N.)
| | - Gavin S Dawe
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland (E.R.K., B.d.H., E.V., P.H.); Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, and SINAPSE, Singapore Institute for Neurotechnology, Singapore (E.R.K., R.R., G.S.D.); Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut (U.F.); and Centre for Neuropsychopharmacology, Division of Brain Sciences, Burlington Danes Building, Imperial College London, London. United Kingdom (D.J.N.)
| |
Collapse
|
29
|
Ubaldi M, Del Bello F, Domi E, Pigini M, Nasuti C. Investigation of allyphenyline efficacy in the treatment of alcohol withdrawal symptoms. Eur J Pharmacol 2015; 760:122-8. [PMID: 25895641 DOI: 10.1016/j.ejphar.2015.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/31/2015] [Accepted: 04/02/2015] [Indexed: 10/23/2022]
Abstract
We have recently demonstrated that allyphenyline, behaving as α2C-adrenoceptor/serotonin 5-HT1A receptor agonist and α2A-adrenoceptor antagonist, in mice enhanced morphine analgesia, attenuated morphine withdrawal symptoms, showed significant antidepressant-like activity and was devoid of sedative side effects. Opioid and alcohol withdrawal shares several common neurobiological and molecular mechanisms. Therefore, in this study we expanded our analysis of the pharmacological properties of allyphenyline by investigating its ability to prevent the expression of somatic withdrawal signs, anxiety-like behavior and hyperlocomotion associated with chronic ethanol intoxication. Rats were subjected to induction of ethanol dependence via repeated daily intragastric ethanol (20%) administration for 4 consecutive days. Twelve hours after the last alcohol administration, somatic alcohol withdrawal signs were scored. Results revealed a significant expression of physical withdrawal signs that were not affected by intraperitoneal (i.p.) administration of allyphenyline at the doses of 0.05, 0.275 and 0.5 mg/kg. In contrast, allyphenyline (0.05 and 0.275 mg/kg i.p.) significantly reduced hyperanxiety-like behavior observed 6 days after alcohol intoxication as measured using the defensive burying test. Allyphenyline also reduced open field hyperlocomotor activity associated with alcohol withdrawal. Notably, the anxiolytic effect of the compound, as well as the already reported antidepressant action, was observed at very low doses, suggesting the involvement of its α2C-adrenoceptor/serotonin 5-HT1A receptor agonism. Therefore, the present investigation suggests that allyphenyline might represent an interesting pharmacological tool to investigate the potential of compounds exhibiting α2C-adrenoceptor/serotonin 5-HT1A receptor agonism and α2A-adrenoceptor antagonism in the treatment of hyperanxiety and hyperlocomotion occurring during alcohol withdrawal in dependent subjects.
Collapse
Affiliation(s)
- Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 62032 Camerino, Italy.
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy.
| | - Esi Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 62032 Camerino, Italy
| | - Maria Pigini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | - Cinzia Nasuti
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 62032 Camerino, Italy
| |
Collapse
|
30
|
de Guglielmo G, Martin-Fardon R, Teshima K, Ciccocioppo R, Weiss F. MT-7716, a potent NOP receptor agonist, preferentially reduces ethanol seeking and reinforcement in post-dependent rats. Addict Biol 2015; 20:643-51. [PMID: 24930632 PMCID: PMC4268094 DOI: 10.1111/adb.12157] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Dysregulation of the nociceptin (N/OFQ) system has been implicated in alcohol abuse and alcoholism, and growing evidence suggests that targeting this system may be beneficial for treating alcoholism. To further explore the treatment target potential of the N/OFQ system, the novel non-peptide, small-molecule N/OFQ (NOP) agonist MT-7716, (R)-2-{3-[1-(Acenaphthen-1-yl)piperidin-4-yl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl}-N-methylacetamide hydrochloride hydrate, was examined for its effects on ethanol self-administration and stress-induced reinstatement of alcohol seeking in non-dependent and post-dependent rats. Male Wistar rats were trained to self-administer ethanol and then made ethanol dependent via repeated intragastric ethanol intubation. The effects of MT-7716 (0.3 and 1 mg/kg; PO) on alcohol self-administration were determined 2 weeks following dependence induction, when baseline self-administration was restored. Effects of MT-7716 on stress-induced reinstatement were tested in separate cohorts of rats, 1 and 3 weeks post-withdrawal. MT-7716 reduced alcohol self-administration and stress-induced reinstatement of alcohol seeking in post-dependent rats, but was ineffective in non-dependent animals. Moreover, the prevention of stress-induced reinstatement by MT-7716 was more pronounced at 3 weeks post-dependence. The results further confirm treatment target potential for the NOP receptor and identify non-peptide NOP agonists as promising potential treatment drugs for alcohol abuse and relapse prevention. The findings also support dysregulation of the N/OFQ system as a factor in alcohol seeking and reinforcement.
Collapse
Affiliation(s)
- Giordano de Guglielmo
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 62032 Camerino, Italy
| | - Rémi Martin-Fardon
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Koji Teshima
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 62032 Camerino, Italy
| | - Friedbert Weiss
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
31
|
Abstract
In the past decades, a large number of neuropeptides with unknown functions have been identified in the brain. Among the newly discovered peptides, nociceptin or orphanin-FQ (N/OFQ) peptide has attracted considerable attention because of its sequence homology with the opioid peptide family. N/OFQ and its cognate receptor (NOP receptor) are distributed widely in the mammalian central nervous system, though particularly intense expression is found in corticolimbic structures. Such distinctive pattern of expression suggests a key role of N/OFQ system in higher brain functions, such as cognition and emotion. In this chapter, we will outline the findings supporting the role played by N/OFQ and NOP receptors in learning and memory and discuss the underlying mechanisms.
Collapse
|
32
|
Schröder W, Lambert DG, Ko MC, Koch T. Functional plasticity of the N/OFQ-NOP receptor system determines analgesic properties of NOP receptor agonists. Br J Pharmacol 2014; 171:3777-800. [PMID: 24762001 PMCID: PMC4128043 DOI: 10.1111/bph.12744] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/07/2014] [Accepted: 04/15/2014] [Indexed: 12/12/2022] Open
Abstract
Despite high sequence similarity between NOP (nociceptin/orphanin FQ opioid peptide) and opioid receptors, marked differences in endogenous ligand selectivity, signal transduction, phosphorylation, desensitization, internalization and trafficking have been identified; underscoring the evolutionary difference between NOP and opioid receptors. Activation of NOP receptors affects nociceptive transmission in a site-specific manner, with antinociceptive effects prevailing after peripheral and spinal activation, and pronociceptive effects after supraspinal activation in rodents. The net effect of systemically administered NOP receptor agonists on nociception is proposed to depend on the relative contribution of peripheral, spinal and supraspinal activation, and this may depend on experimental conditions. Functional expression and regulation of NOP receptors at peripheral and central sites of the nociceptive pathway exhibits a high degree of plasticity under conditions of neuropathic and inflammatory pain. In rodents, systemically administered NOP receptor agonists exerted antihypersensitive effects in models of neuropathic and inflammatory pain. However, they were largely ineffective in acute pain while concomitantly evoking severe motor side effects. In contrast, systemic administration of NOP receptor agonists to non-human primates (NHPs) exerted potent and efficacious antinociception in the absence of motor and sedative side effects. The reason for this species difference with respect to antinociceptive efficacy and tolerability is not clear. Moreover, co-activation of NOP and μ-opioid peptide (MOP) receptors synergistically produced antinociception in NHPs. Hence, both selective NOP receptor as well as NOP/MOP receptor agonists may hold potential for clinical use as analgesics effective in conditions of acute and chronic pain.
Collapse
Affiliation(s)
- W Schröder
- Department of Translational Science, Global Innovation, Grünenthal GmbH, Aachen, Germany
| | | | | | | |
Collapse
|
33
|
Filaferro M, Ruggieri V, Novi C, Calò G, Cifani C, Micioni Di Bonaventura MV, Sandrini M, Vitale G. Functional antagonism between nociceptin/orphanin FQ and corticotropin-releasing factor in rat anxiety-related behaviors: involvement of the serotonergic system. Neuropeptides 2014; 48:189-97. [PMID: 24894718 DOI: 10.1016/j.npep.2014.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 05/05/2014] [Accepted: 05/07/2014] [Indexed: 01/30/2023]
Abstract
Nociceptin/orphanin FQ (N/OFQ) acts as an anxiolytic-like agent in the rat and behaves as a functional antagonist of corticotropin-releasing factor (CRF) due to its ability to oppose CRF biological actions. In response to stress, CRF triggers changes in neurotransmitter systems including serotonin (5-HT). The role of 5-HT1A receptor in anxiety has been supported by preclinical and clinical studies. The present study investigated the possible functional antagonism between N/OFQ (1nmol/rat) and CRF (0.2nmol/rat) in anxiety-related conditions in rats, using elevated plus maze and defensive burying tests, in order to confirm previous literature results. Moreover, possible changes in the serotonergic system were studied in areas rich of serotonergic neurons: frontal cortex and pons. In both tests N/OFQ showed anxiolytic-like effects while CRF displayed anxiogenic-like effects. N/OFQ before CRF treatment counteracted the anxiogenic-like effects evoked by CRF. In frontal cortex, N/OFQ significantly decreased 5-HT levels but did not modify the hydroxyindoleacetic acid (5-HIAA) ones; CRF modified neither 5-HT nor 5-HIAA content but counteracted changes induced by N/OFQ alone. In pons, N/OFQ induced no change in serotonergic activity while CRF significantly decreased 5-HT levels and increased 5-HIAA content. The two peptides' combination reinstated serotonergic parameters to controls. In frontal cortex, N/OFQ increased the 5HT1A receptor density but reduced its affinity, while CRF alone did not induce any change. In pons, CRF decreased 5HT1ABmax and KD whereas N/OFQ was ineffective. All biochemical modifications were reverted by N/OFQ plus CRF treatment. The present study confirms that N/OFQ counteracts CRF anxiogenic-like effects in the behavioral tests evaluated. These effects may involve central serotonergic mechanisms since N/OFQ plus CRF induces a reversion of serotonergic changes provoked by single peptide. Our data support the hypothesis that N/OFQ may behave as functional CRF antagonist, this action being of interest for the treatment of anxiety disorders.
Collapse
Affiliation(s)
- M Filaferro
- Department of Biomedical, Metabolic Sciences and Neurosciences, Section of Pharmacology, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - V Ruggieri
- Department of Life Sciences, Section of Pharmacology, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - C Novi
- Department of Life Sciences, Section of Pharmacology, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - G Calò
- Department of Medical Sciences, Section of Pharmacology, and National Institute of Neuroscience, University of Ferrara, Via Fossato di Mortara 19, 44121 Ferrara, Italy
| | - C Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Macerata, Italy
| | - M V Micioni Di Bonaventura
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Macerata, Italy
| | - M Sandrini
- Department of Biomedical, Metabolic Sciences and Neurosciences, Section of Pharmacology, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - G Vitale
- Department of Life Sciences, Section of Pharmacology, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy.
| |
Collapse
|
34
|
Kallupi M, Varodayan FP, Oleata CS, Correia D, Luu G, Roberto M. Nociceptin/orphanin FQ decreases glutamate transmission and blocks ethanol-induced effects in the central amygdala of naive and ethanol-dependent rats. Neuropsychopharmacology 2014; 39:1081-92. [PMID: 24169802 PMCID: PMC3957102 DOI: 10.1038/npp.2013.308] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/08/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022]
Abstract
The central nucleus of the amygdala (CeA) mediates several addiction-related processes and nociceptin/orphanin FQ (nociceptin) regulates ethanol intake and anxiety-like behaviors. Glutamatergic synapses, in the CeA and throughout the brain, are very sensitive to ethanol and contribute to alcohol reinforcement, tolerance, and dependence. Previously, we reported that in the rat CeA, acute and chronic ethanol exposures significantly decrease glutamate transmission by both pre- and postsynaptic actions. In this study, using electrophysiological techniques in an in vitro CeA slice preparation, we investigated the effects of nociceptin on glutamatergic transmission and its interaction with acute ethanol in naive and ethanol-dependent rats. We found that nociceptin (100-1000 nM) diminished basal-evoked compound glutamatergic receptor-mediated excitatory postsynaptic potentials (EPSPs) and spontaneous and miniature EPSCs (s/mEPSCs) by mainly decreasing glutamate release in the CeA of naive rats. Notably, nociceptin blocked the inhibition induced by acute ethanol (44 mM) and ethanol blocked the nociceptin-induced inhibition of evoked EPSPs in CeA neurons of naive rats. In neurons from chronic ethanol-treated (ethanol-dependent) rats, the nociceptin-induced inhibition of evoked EPSP amplitude was not significantly different from that in naive rats. Application of [Nphe1]Nociceptin(1-13)NH2, a nociceptin receptor (NOP) antagonist, revealed tonic inhibitory activity of NOP on evoked CeA glutamatergic transmission only in ethanol-dependent rats. The antagonist also blocked nociceptin-induced decreases in glutamatergic responses, but did not affect ethanol-induced decreases in evoked EPSP amplitude. Taken together, these studies implicate a potential role for the nociceptin system in regulating glutamatergic transmission and a complex interaction with ethanol at CeA glutamatergic synapses.
Collapse
Affiliation(s)
- Marsida Kallupi
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Florence P Varodayan
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher S Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Diego Correia
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Pharmacology, Universidade Federal do Paraná, Jardim das Américas, Curitiba, Paraná, Brazil
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
35
|
Kallupi M, Oleata CS, Luu G, Teshima K, Ciccocioppo R, Roberto M. MT-7716, a novel selective nonpeptidergic NOP receptor agonist, effectively blocks ethanol-induced increase in GABAergic transmission in the rat central amygdala. Front Integr Neurosci 2014; 8:18. [PMID: 24600360 PMCID: PMC3927450 DOI: 10.3389/fnint.2014.00018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/01/2014] [Indexed: 11/13/2022] Open
Abstract
The GABAergic system in the central amygdala (CeA) plays a major role in ethanol dependence and the anxiogenic-like response to ethanol withdrawal. A large body of evidence shows that Nociceptin/Orphanin FQ (N/OFQ) regulates ethanol intake and anxiety-like behavior. In the rat, ethanol significantly augments CeA GABA release, whereas N/OFQ diminishes it. Using electrophysiological techniques in an in vitro slice preparation, in this study we investigated the effects of a nonpeptidergic NOP receptor agonist, MT-7716 [(R)-2-3-[1-(Acenaphthen-1-yl)piperidin-4-yl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl-N-methylacetamide hydrochloride hydrate], and its interaction with ethanol on GABAergic transmission in CeA slices of naïve rats. We found that MT-7716 dose-dependently (100-1000 nM) diminished evoked GABAA receptor-mediated inhibitory postsynaptic potentials (IPSPs) and increased paired-pulse facilitation (PPF) ratio of these evoked IPSPs, suggesting a presynaptic site of action of the MT-7716 by decreasing GABA release at CeA synapses. The presynaptic action of MT-7716 was also supported by the significant decrease in the frequency of miniature inhibitory postsynaptic currents (mIPSCs) induced by the nociceptin receptor (NOP) agonist. Interestingly, MT-7716 prevented the ethanol-induced augmentation of evoked IPSPs. A putative selective NOP antagonist, [Nphe1]Nociceptin(1-13)NH2, totally prevented the MT-7716-induced inhibition of IPSP amplitudes indicating that MT-7716 exerts its effect through NOPs. These data provide support for an interaction between the nociceptin and GABAergic systems in the CeA and for the anti-alcohol properties of the NOP activation. The development of a synthetic nonpeptidergic NOP receptor agonist such as MT-7716 may represent a useful therapeutic target for alcoholism.
Collapse
Affiliation(s)
- Marsida Kallupi
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
- Pharmacology Unit, School of Pharmacy, University of CamerinoCamerino, Italy
| | - Christopher S. Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| | - Koji Teshima
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma CorporationYokohama, Japan
| | - Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of CamerinoCamerino, Italy
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| |
Collapse
|
36
|
Ciccocioppo R, de Guglielmo G, Hansson AC, Ubaldi M, Kallupi M, Cruz MT, Oleata CS, Heilig M, Roberto M. Restraint stress alters nociceptin/orphanin FQ and CRF systems in the rat central amygdala: significance for anxiety-like behaviors. J Neurosci 2014; 34:363-72. [PMID: 24403138 PMCID: PMC3870926 DOI: 10.1523/jneurosci.2400-13.2014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 10/02/2013] [Accepted: 11/02/2013] [Indexed: 02/05/2023] Open
Abstract
Corticotropin releasing factor (CRF) is the primary mediator of stress responses, and nociceptin/orphanin FQ (N/OFQ) plays an important role in the modulation of these stress responses. Thus, in this multidisciplinary study, we explored the relationship between the N/OFQ and the CRF systems in response to stress. Using in situ hybridization (ISH), we assessed the effect of body restraint stress on the gene expression of CRF and N/OFQ-related genes in various subdivisions of the amygdala, a critical brain structure involved in the modulation of stress response and anxiety-like behaviors. We found a selective upregulation of the NOP and downregulation of the CRF1 receptor transcripts in the CeA and in the BLA after body restraint. Thus, we performed intracellular electrophysiological recordings of GABAA-mediated IPSPs in the central nucleus of the amygdala (CeA) to explore functional interactions between CRF and N/OFQ systems in this brain region. Acute application of CRF significantly increased IPSPs in the CeA, and this enhancement was blocked by N/OFQ. Importantly, in stress-restraint rats, baseline CeA GABAergic responses were elevated and N/OFQ exerted a larger inhibition of IPSPs compared with unrestraint rats. The NOP antagonist [Nphe1]-nociceptin(1-13)NH2 increased the IPSP amplitudes in restraint rats but not in unrestraint rats, suggesting a functional recruitment of the N/OFQ system after acute stress. Finally, we evaluated the anxiety-like response in rats subjected to restraint stress and nonrestraint rats after N/OFQ microinjection into the CeA. Intra-CeA injections of N/OFQ significantly and selectively reduced anxiety-like behavior in restraint rats in the elevated plus maze. These combined results demonstrate that acute stress increases N/OFQ systems in the CeA and that N/OFQ has antistress properties.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
| | - Giordano de Guglielmo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
| | - Anita C. Hansson
- Institute of Psychopharmacology at the Central Institute of Mental Health, Medical Faculty Mannheim/University of Heidelberg, Heidelberg 68159, Germany
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
| | - Marsida Kallupi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, and
| | - Maureen T. Cruz
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, and
| | - Christopher S. Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, and
| | - Markus Heilig
- National Institutes of Health/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland 20892-1108
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, and
| |
Collapse
|
37
|
Mallimo EM, Kusnecov AW. The role of orphanin FQ/nociceptin in neuroplasticity: relationship to stress, anxiety and neuroinflammation. Front Cell Neurosci 2013; 7:173. [PMID: 24155687 PMCID: PMC3792366 DOI: 10.3389/fncel.2013.00173] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 09/14/2013] [Indexed: 01/23/2023] Open
Abstract
The neuropeptide, orphanin FQ/nociceptin (OFQ/N or simply, nociceptin), is expressed in both neuronal and non-neuronal tissue, including the immune system. In the brain, OFQ/N has been investigated in relation to stress, anxiety, learning and memory, and addiction. More recently, it has also been found that OFQ/N influences glial cell functions, including oligodendrocytes, astrocytes, and microglial cells. However, this latter research is relatively small, but potentially important, when observations regarding the relationship of OFQ/N to stress and emotional functions is taken into consideration and integrated with the growing evidence for its involvement in cells that mediate inflammatory events. This review will first provide an overview and understanding of how OFQ/N has been implicated in the HPA axis response to stress, followed by an understanding of its influence on natural and learned anxiety-like behavior. What emerges from an examination of the literature is a neuropeptide that appears to counteract anxiogenic influences, but paradoxically, without attenuating HPA axis responses generated in response to stress. Studies utilized both central administration of OFQ/N, which was shown to activate the HPA axis, as well as antagonism of NOP-R, the OFQ/N receptor. In contrast, antagonist or transgenic OFQ/N or NOP-R knockout studies, showed augmentation of HPA axis responses to stress, suggesting that OFQ/N may be needed to control the magnitude of the HPA axis response to stress. Investigations of behavior in standard exploratory tests of anxiogenic behavior (eg., elevated plus maze) or learned fear responses have suggested that OFQ/N is needed to attenuate fear or anxiety-like behavior. However, some discrepant observations, in particular, those that involve appetitive behaviors, suggest a failure of NOP-R deletion to increase anxiety. However, it is also suggested that OFQ/N may operate in an anxiolytic manner when initial anxiogenic triggers (eg., the neuropeptide CRH) are initiated. Finally, the regulatory functions of OFQ/N in relation to emotion-related behaviors may serve to counteract potential neuroinflammatory events in the brain. This appears to be evident within the glial cell environment of the brain, since OFQ/N has been shown to reduce the production of proinflammatory cellular and cytokine events. Given that both OFQ/N and glial cells are activated in response to stress, it is possible that there is a possible convergence of these two systems that has important repercussions for behavior and neuroplasticity.
Collapse
Affiliation(s)
- Elyse M Mallimo
- Behavioral and Systems Neuroscience Program, Department of Psychology, Rutgers University New Brunswick, NJ, USA
| | | |
Collapse
|
38
|
Stock AK, Blaszkewicz M, Beste C. Effects of binge drinking on action cascading processes: an EEG study. Arch Toxicol 2013; 88:475-88. [DOI: 10.1007/s00204-013-1109-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 07/23/2013] [Indexed: 11/28/2022]
|
39
|
Kang-Park M, Kieffer BL, Roberts AJ, Siggins GR, Moore SD. κ-Opioid receptors in the central amygdala regulate ethanol actions at presynaptic GABAergic sites. J Pharmacol Exp Ther 2013; 346:130-7. [PMID: 23587526 PMCID: PMC3684840 DOI: 10.1124/jpet.112.202903] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 04/12/2013] [Indexed: 12/18/2022] Open
Abstract
Human and animal studies indicate that κ-opioid receptors (KORs) are involved in ethanol drinking and dependence (Xuei et al., 2006; Walker and Koob, 2008; Walker et al., 2011). Using in vitro single-cell recording techniques in mouse brain slices, we examined the physiologic effects of KOR activation in the central amygdala (CeA) on GABAergic neurotransmission and its interaction with acute ethanol. A selective KOR agonist (U69593, 1 μM) diminished evoked GABAergic inhibitory postsynaptic currents (IPSCs) by 18% (n = 10), whereas blockade of KORs with a selective antagonist (nor-binaltorphimine, 1 μM) augmented the baseline evoked GABAergic IPSCs by 14% (P < 0.01; n = 34), suggesting that the KOR system contributes to tonic inhibition of GABAergic neurotransmission in the CeA. In addition, the enhancement by acute ethanol of GABAergic IPSC amplitudes was further augmented by pharmacologic blockade of KORs, from 14% (n = 36) to 27% (n = 26; P < 0.01), or by genetic deletion of KORs, from 14% in wild-type mice (n = 19) to 34% in KOR knockout mice (n = 13; P < 0.01). Subsequent experiments using tetrodotoxin to block activity-dependent neurotransmission suggest that KORs regulate GABA release at presynaptic sites. Our data support the idea that KORs modulate GABAergic synaptic responses and ethanol effects as one of multiple opioid system-dependent actions of ethanol in the CeA, possibly in a circuit-specific manner.
Collapse
Affiliation(s)
- Maenghee Kang-Park
- Department of Psychiatry, Duke University Medical Center, and Research Service, Veterans Administration Medical Center, Durham, North Carolina 27705, USA
| | | | | | | | | |
Collapse
|
40
|
Lesscher HMB, Vanderschuren LJMJ. Compulsive drug use and its neural substrates. Rev Neurosci 2013; 23:731-45. [PMID: 23079511 DOI: 10.1515/revneuro-2012-0066] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 08/15/2012] [Indexed: 12/22/2022]
Abstract
Drug addiction is a chronic relapsing brain disease, characterized by compulsive drug use. Despite the fact that drug addiction affects millions of people worldwide, treatments for this disorder are limited in number and efficacy. In our opinion, understanding the neural underpinnings of drug addiction would open new avenues for the development of innovative treatments for this disorder. Based on an awareness that drug use and drug reward do not equal drug addiction, there has been increasing interest in developing animal models of addiction that mimick the loss of control over drug use more closely than existing models aimed at studying drug reward. The present review provides an overview of animal studies of compulsive drug use and the neural mechanisms involved. First, the employed models are summarized, with a particular emphasis on models of escalation of drug use and resistance to punishment. Next, we discuss mechanisms within the (ventral and dorsal) striatum and (central) amygdala that have recently been implicated in the compulsive seeking and taking of alcohol and cocaine. The studies discussed here provide a promising line of research that will advance our knowledge of the neural circuits involved in the self-destructive behavior that characterizes drug addiction.
Collapse
Affiliation(s)
- Heidi M B Lesscher
- Department of Animals in Science and Society, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
41
|
Aujla H, Cannarsa R, Romualdi P, Ciccocioppo R, Martin-Fardon R, Weiss F. Modification of anxiety-like behaviors by nociceptin/orphanin FQ (N/OFQ) and time-dependent changes in N/OFQ-NOP gene expression following ethanol withdrawal. Addict Biol 2013; 18:467-79. [PMID: 22804785 PMCID: PMC3477306 DOI: 10.1111/j.1369-1600.2012.00466.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Anxiety is a key consequence of ethanol withdrawal and important risk factor for relapse. The neuropeptide nociceptin/orphanin FQ (N/OFQ) or agonists at this peptide's receptor (NOP) exert anxiolytic-like and antistress actions. N/OFQ dysfunction has been linked to both a high-anxiety behavioral phenotype and excessive ethanol intake. Recent studies suggest a possible link between genetic polymorphisms of the NOP transcript and alcoholism. Thus, in the present study, the effects of intracerebroventricularly administered N/OFQ were tested for modification of anxiety-like behaviors, using the shock-probe defensive burying and elevated plus-maze tests, in ethanol-dependent versus non-dependent rats, 1 and 3 weeks following termination of ethanol exposure. Additionally, prepro-N/OFQ (ppN/OFQ) and NOP receptor gene expression was measured in the central nucleus of the amygdala, in the bed nucleus of the stria terminalis and in the lateral hypothalamus at the same timepoints in separate subjects. One week post-ethanol, N/OFQ dose-dependently attenuated elevated anxiety-like behavior in ethanol-dependent rats and produced anxiolytic-like effects in non-dependent controls in both behavioral tests. However, 3 weeks post-ethanol, N/OFQ altered behavior consistent with anxiogenic-like actions in ethanol-dependent rats but continued to exert anxiolytic-like actions in non-dependent controls. These findings were paralleled by ethanol history-dependent changes of ppN/OFQ and NOP gene expression that showed a distinctive time course in the examined brain structures. The results demonstrate that ethanol dependence and withdrawal are associated with neuroadaptive changes in the N/OFQ-NOP system, suggesting a role of this neuropeptidergic pathway as a therapeutic target for the treatment of alcohol abuse.
Collapse
Affiliation(s)
- H Aujla
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California
| | - R Cannarsa
- Department of Pharmacology, University of Bologna, Bologna, Italy
| | - P Romualdi
- Department of Pharmacology, University of Bologna, Bologna, Italy
| | - R Ciccocioppo
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - R Martin-Fardon
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California
| | - F Weiss
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
42
|
D'Addario C, Caputi FF, Rimondini R, Gandolfi O, Del Borrello E, Candeletti S, Romualdi P. Different alcohol exposures induce selective alterations on the expression of dynorphin and nociceptin systems related genes in rat brain. Addict Biol 2013; 18:425-33. [PMID: 21507157 DOI: 10.1111/j.1369-1600.2011.00326.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Molecular mechanisms of adaptive transformations caused by alcohol exposure on opioid dynorphin and nociceptin systems have been investigated in the rat brain. Alcohol was intragastrically administered to rats to resemble human drinking with several hours of exposure: water or alcohol (20% in water) at a dose of 1.5 g/kg three times daily for 1 or 5 days. The development of tolerance and dependence were recorded daily. Brains were dissected 30 minutes (1- and 5-day groups) or 1, 3 or 7 days after the last administration for the three other 5-day groups (groups under withdrawal). Specific alterations in opioid genes expression were ascertained. In the amygdala, an up-regulation of prodynorphin and pronociceptin was observed in the 1-day group; moreover, pronociceptin and the kappa opioid receptor mRNAs in the 5-day group and both peptide precursors in the 1-day withdrawal group were also up-regulated. In the prefrontal cortex, an increase in prodynorhin expression in the 1-day group was detected. These data indicate a relevant role of the dynorphinergic system in the negative hedonic states associated with multiple alcohol exposure. The pattern of alterations observed for the nociceptin system appears to be consistent with its role of functional antagonism towards the actions of ethanol associated with other opioid peptides. Our findings could help to the understanding of how alcohol differentially affects the opioid systems in the brain and also suggest the dynorphin and nociceptin systems as possible targets for the treatment and/or prevention of alcohol dependence.
Collapse
Affiliation(s)
- Claudio D'Addario
- Department of Pharmacology, University of Bologna, Via Irnerio 48, Bologna, Italy.
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Neuropeptides are found in many mammalian CNS neurons where they play key roles in modulating neuronal activity. In contrast to amino acid transmitter release at the synapse, neuropeptide release is not restricted to the synaptic specialization, and after release, a neuropeptide may diffuse some distance to exert its action through a G protein-coupled receptor. Some neuropeptides such as hypocretin/orexin are synthesized only in single regions of the brain, and the neurons releasing these peptides probably have similar functional roles. Other peptides such as neuropeptide Y (NPY) are synthesized throughout the brain, and neurons that synthesize the peptide in one region have no anatomical or functional connection with NPY neurons in other brain regions. Here, I review converging data revealing a complex interaction between slow-acting neuromodulator peptides and fast-acting amino acid transmitters in the control of energy homeostasis, drug addiction, mood and motivation, sleep-wake states, and neuroendocrine regulation.
Collapse
|
44
|
Roberto M, Gilpin NW, Siggins GR. The central amygdala and alcohol: role of γ-aminobutyric acid, glutamate, and neuropeptides. Cold Spring Harb Perspect Med 2012; 2:a012195. [PMID: 23085848 PMCID: PMC3543070 DOI: 10.1101/cshperspect.a012195] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug seeking and drug taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptative mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid-ergic (GABAergic) transmission in CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and AMPA receptors in CeA, whereas chronic alcohol up-regulates N-methyl-d-aspartate receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and anti-stress (e.g., NPY, nociceptin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.
Collapse
Affiliation(s)
- Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
45
|
Abstract
Ethanol (EtOH) has effects on numerous cellular molecular targets, and alterations in synaptic function are prominent among these effects. Acute exposure to EtOH activates or inhibits the function of proteins involved in synaptic transmission, while chronic exposure often produces opposing and/or compensatory/homeostatic effects on the expression, localization, and function of these proteins. Interactions between different neurotransmitters (e.g., neuropeptide effects on release of small molecule transmitters) can also influence both acute and chronic EtOH actions. Studies in intact animals indicate that the proteins affected by EtOH also play roles in the neural actions of the drug, including acute intoxication, tolerance, dependence, and the seeking and drinking of EtOH. This chapter reviews the literature describing these acute and chronic synaptic effects of EtOH and their relevance for synaptic transmission, plasticity, and behavior.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, NIAAA, 5625 Fishers Lane, Room TS-13A, Rockville, MD 20852, USA.
| | | |
Collapse
|
46
|
Bajo M, Madamba SG, Lu X, Sharkey LM, Bartfai T, Siggins GR. Receptor subtype-dependent galanin actions on gamma-aminobutyric acidergic neurotransmission and ethanol responses in the central amygdala. Addict Biol 2012; 17:694-705. [PMID: 21955024 DOI: 10.1111/j.1369-1600.2011.00360.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The neuropeptide galanin and its three receptor subtypes (GalR1-3) are expressed in the central amygdala (CeA), a brain region involved in stress- and anxiety-related behaviors, as well as alcohol dependence. Galanin also has been suggested to play a role in alcohol intake and alcohol dependence. We examined the effects of galanin in CeA slices from wild-type and knockout (KO) mice deficient of GalR2 and both GalR1 and GalR2 receptors. Galanin had dual effects on gamma-aminobutyric acid (GABA)-ergic transmission, decreasing the amplitudes of pharmacologically isolated GABAergic inhibitory postsynaptic potentials (IPSPs) in over half of CeA neurons but augmenting IPSPs in the others. The increase in IPSP size was absent after superfusion of the GalR3 antagonist SNAP 37889, whereas the IPSP depression was absent in CeA neurons of GalR1 × GalR2 double KO and GalR2 KO mice. Paired-pulse facilitation studies showed weak or infrequent effects of galanin on GABA release. Thus, galanin may act postsynaptically through GalR3 to augment GABAergic transmission in some CeA neurons, whereas GalR2 receptors likely are involved in the depression of IPSPs. Co-superfusion of ethanol, which augments IPSPs presynaptically, together with galanin caused summated effects of ethanol and galanin in those CeA neurons showing galanin-augmented IPSPs, suggesting the two agents act via different mechanisms in this population. However, in neurons showing IPSP-diminishing galanin effects, galanin blunted the ethanol effects, suggesting a preemptive effect of galanin. These findings may increase understanding of the complex cellular mechanisms that underlie the anxiety-related behavioral effects of galanin and ethanol in CeA.
Collapse
Affiliation(s)
- Michal Bajo
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
47
|
Gilpin NW. Corticotropin-releasing factor (CRF) and neuropeptide Y (NPY): effects on inhibitory transmission in central amygdala, and anxiety- & alcohol-related behaviors. Alcohol 2012; 46:329-37. [PMID: 22560367 PMCID: PMC3613993 DOI: 10.1016/j.alcohol.2011.11.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 11/09/2011] [Accepted: 11/28/2011] [Indexed: 12/25/2022]
Abstract
The central amygdala (CeA) is uniquely situated to function as an interface between stress- and addiction-related processes. This brain region has long been attributed an important role in aversive (e.g., fear) conditioning, as well as the negative emotional states that define alcohol dependence and withdrawal. The CeA is the major output region of the amygdala and receives complex inputs from other amygdaloid nuclei as well as regions that integrate sensory information from the external environment (e.g., thalamus, cortex). The CeA is functionally and anatomically divided into lateral and medial subdivisions that themselves are interconnected and populated by inhibitory interneurons and projections neurons. Neuropeptides are highly expressed in the CeA, particularly in the lateral subdivision, and the role of many of these peptides in regulating anxiety- and alcohol-related behaviors has been localized to the CeA. This review focuses on two of these peptides, corticotropin-releasing factor (CRF) and neuropeptide Y (NPY), that exhibit a high degree of neuroanatomical overlap (e.g., in CeA) and largely opposite behavioral profiles (e.g., in regulating anxiety- and alcohol-related behavior). CRF and NPY systems in the CeA appear to be recruited and/or up-regulated during the transition to alcohol dependence. These and other neuropeptides may converge on GABA synapses in CeA to control projection neurons and downstream effector regions, thereby translating negative affective states into anxiety-like behavior and excessive alcohol consumption.
Collapse
Affiliation(s)
- Nicholas W Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
48
|
Cruz MT, Herman MA, Kallupi M, Roberto M. Nociceptin/orphanin FQ blockade of corticotropin-releasing factor-induced gamma-aminobutyric acid release in central amygdala is enhanced after chronic ethanol exposure. Biol Psychiatry 2012; 71:666-76. [PMID: 22153590 PMCID: PMC3838304 DOI: 10.1016/j.biopsych.2011.10.032] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 10/26/2011] [Accepted: 10/29/2011] [Indexed: 11/24/2022]
Abstract
BACKGROUND The central nucleus of the amygdala (CeA) mediates stress- and addiction-related processes. Corticotropin-releasing factor (CRF) and nociceptin/orphanin FQ (nociceptin) regulate ethanol intake and anxiety-like behavior. In the rat, CRF and ethanol significantly augment CeA gamma-aminobutyric acid (GABA) release, whereas nociceptin diminishes it. METHODS Using electrophysiologic techniques in an in vitro slice preparation, we investigated the interaction of nociceptin and CRF on evoked and spontaneous GABAergic transmission in CeA slices of naive and ethanol-dependent rats and the mechanistic role of protein kinase A. RESULTS In neurons from naive animals, nociceptin dose-dependently diminished basal-evoked GABA(A) receptor-mediated inhibitory postsynaptic potentials (IPSPs) by decreasing GABA release and prevented, as well as reversed, CRF-induced augmentation of IPSPs, actions that required PKA signaling. In neurons from ethanol-dependent animals, nociceptin decreased basal GABAergic transmission and blocked the CRF-induced increase in GABA release to a greater extent than in naive controls. CONCLUSIONS These data provide new evidence for an interaction between the nociceptin and CRF systems in the CeA. Nociceptin opposes CRF effects on CeA GABAergic transmission with sensitization of this effect in dependent animals. These properties of nociceptin may underlie its anti-alcohol and anxiolytic properties and identify the nociceptin receptor as a useful therapeutic target for alcoholism.
Collapse
Affiliation(s)
- Maureen T Cruz
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
49
|
Gilpin NW, Roberto M. Neuropeptide modulation of central amygdala neuroplasticity is a key mediator of alcohol dependence. Neurosci Biobehav Rev 2012; 36:873-88. [PMID: 22101113 PMCID: PMC3325612 DOI: 10.1016/j.neubiorev.2011.11.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 11/01/2011] [Accepted: 11/05/2011] [Indexed: 01/08/2023]
Abstract
Alcohol use disorders are characterized by compulsive drug-seeking and drug-taking, loss of control in limiting intake, and withdrawal syndrome in the absence of drug. The central amygdala (CeA) and neighboring regions (extended amygdala) mediate alcohol-related behaviors and chronic alcohol-induced plasticity. Acute alcohol suppresses excitatory (glutamatergic) transmission whereas chronic alcohol enhances glutamatergic transmission in CeA. Acute alcohol facilitates inhibitory (GABAergic) transmission in CeA, and chronic alcohol increases GABAergic transmission. Electrophysiology techniques are used to explore the effects of neuropeptides/neuromodulators (CRF, NPY, nociceptin, dynorphin, endocannabinoids, galanin) on inhibitory transmission in CeA. In general, pro-anxiety peptides increase, and anti-anxiety peptides decrease CeA GABAergic transmission. These neuropeptides facilitate or block the action of acute alcohol in CeA, and chronic alcohol produces plasticity in neuropeptide systems, possibly reflecting recruitment of negative reinforcement mechanisms during the transition to alcohol dependence. A disinhibition model of CeA output is discussed in the context of alcohol dependence- and anxiety-related behaviors.
Collapse
Affiliation(s)
- Nicholas W Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, 1901 Perdido Street, New Orleans, LA 70112, USA.
| | | |
Collapse
|
50
|
McCool BA. Ethanol modulation of synaptic plasticity. Neuropharmacology 2011; 61:1097-108. [PMID: 21195719 PMCID: PMC3149748 DOI: 10.1016/j.neuropharm.2010.12.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 12/10/2010] [Accepted: 12/22/2010] [Indexed: 12/19/2022]
Abstract
Synaptic plasticity in the most general terms represents the flexibility of neurotransmission in response to neuronal activity. Synaptic plasticity is essential both for the moment-by-moment modulation of neural activity in response to dynamic environmental cues and for long-term learning and memory formation. These temporal characteristics are served by an array of pre- and post-synaptic mechanisms that are frequently modulated by ethanol exposure. This modulation likely makes significant contributions to both alcohol abuse and dependence. In this review, I discuss the modulation of both short-term and long-term synaptic plasticity in the context of specific ethanol-sensitive cellular substrates. A general discussion of the available preclinical, animal-model based neurophysiology literature provides a comparison between results from in vitro and in vivo studies. Finally, in the context of alcohol abuse and dependence, the review proposes potential behavioral contributions by ethanol modulation of plasticity.
Collapse
Affiliation(s)
- Brian A McCool
- Department of Physiology & Pharmacology and the Translational Center for the Neurobehavioral Study of Alcohol, Wake Forest University School of Medicine, Winston-Salem NC 27157, USA.
| |
Collapse
|