1
|
Anjos-Santos A, Erikson CM, Flores-Ramirez FJ, Rodriguez L, Barchiesi R, Vozella V, Borgonetti V, Cruz B, Zalfa C, Hughes K, Gandhi P, Bajo M, Vlkolinsky R, Mayfield RD, Martin-Fardon R, Roberto M. Noradrenaline modulates central amygdala GABA transmission and alcohol drinking in female rats. Biol Psychiatry 2025:S0006-3223(25)01114-X. [PMID: 40194754 DOI: 10.1016/j.biopsych.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a chronic relapsing disorder and a leading preventable cause of death worldwide. The central nucleus of the amygdala (CeA) is a hub for stress and AUD. Noradrenaline (norepinephrine; NE) regulates the brain's response to stress and alcohol. We previously reported that α1 adrenergic receptors drive moderate alcohol intake, while β receptors contribute to excessive drinking associated with dependence in male rats. METHODS Here, we determined that alcohol dependence and withdrawal alter the CeA noradrenergic system in female rats using ex vivo electrophysiology, in situ hybridization, site-specific behavioral pharmacology, and RNA-sequencing data from postmortem CeA samples obtained from female donors with and without AUD. RESULTS NE bidirectionally (increase and decrease) modulated CeA GABAergic transmission via both α1 and β receptors. Prazosin, an α1 receptor antagonist, reduced moderate alcohol intake in non-dependent female rats and excessive drinking in dependent females, while propranolol, a β receptor antagonist, only reduced excessive drinking in dependent females. While withdrawal produced a partial functional recovery of the NE modulation of the CeA, some of the cellular patterns of adrenergic receptor mRNA expression persist. Although we did not observe any differences in adrenergic receptor gene expression in the CeA from our human AUD donors, we found a downregulation of ADRA1A in the basolateral amygdala and the dorsolateral prefrontal cortex, compared to controls. CONCLUSIONS Amygdalar α1 and β adrenergic receptors are key neural substrates of AUD. Our results support ongoing development of receptor-specific medication for AUD and highlight promising efficacy in females.
Collapse
Affiliation(s)
- Alexia Anjos-Santos
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA; Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04024-002, Brazil
| | | | - Francisco J Flores-Ramirez
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA; Department of Psychology, California State University, San Marcos, San Marcos, CA, USA
| | - Larry Rodriguez
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - Riccardo Barchiesi
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research (WCAAR), University of Texas at Austin, Austin, TX, USA
| | - Valentina Vozella
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - Vittoria Borgonetti
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA; Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Bryan Cruz
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - Cristina Zalfa
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - Kiley Hughes
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - Pauravi Gandhi
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - Michal Bajo
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - Roman Vlkolinsky
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - R Dayne Mayfield
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research (WCAAR), University of Texas at Austin, Austin, TX, USA
| | - Rémi Martin-Fardon
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA
| | - Marisa Roberto
- Department of Translational Medicine, Scripps Research; La Jolla, CA, USA.
| |
Collapse
|
2
|
Richardson RS, Kryszak LA, Vendruscolo JCM, Koob GF, Vendruscolo LF, Leggio L. GHSR blockade, but not reduction of peripherally circulating ghrelin via β 1-adrenergic receptor antagonism, decreases binge-like alcohol drinking in mice. Mol Psychiatry 2025; 30:1047-1056. [PMID: 39232198 PMCID: PMC11835741 DOI: 10.1038/s41380-024-02713-3] [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: 10/20/2023] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
Alcohol use disorder (AUD) and binge drinking are highly prevalent public health issues. The stomach-derived peptide ghrelin, and its receptor, the growth hormone secretagogue receptor (GHSR), both of which are expressed in the brain and periphery, are implicated in alcohol-related outcomes. We previously found that systemic and central administration of GHSR antagonists reduced binge-like alcohol drinking, whereas a ghrelin vaccine did not. Thus, we hypothesized that central GHSR drives binge-like alcohol drinking independently of peripheral ghrelin. To investigate this hypothesis, we antagonized β1-adrenergic receptors (β1ARs), which are required for peripheral ghrelin release, and combined them with GHSR blockers. We found that both systemic β1AR antagonism with atenolol (peripherally restricted) and metoprolol (brain permeable) robustly decreased plasma ghrelin levels. Also, ICV administration of atenolol had no effect on peripheral endogenous ghrelin levels. However, only metoprolol, but not atenolol, decreased binge-like alcohol drinking. The β1AR antagonism also did not prevent the effects of the GHSR blockers JMV2959 and PF-5190457 in decreasing binge-like alcohol drinking. These results suggest that the GHSR rather than peripheral endogenous ghrelin is involved in binge-like alcohol drinking. Thus, GHSRs and β1ARs represent possible targets for therapeutic intervention for AUD, including the potential combination of drugs that target these two systems.
Collapse
Affiliation(s)
- Rani S Richardson
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
- University of North Carolina School of Medicine MD/PhD Program, University of North Carolina, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - Lindsay A Kryszak
- National Institute on Drug Abuse Intramural Research Program Translational Analytical Core, National Institutes of Health, Baltimore, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, MD, USA.
- National Institute on Drug Abuse Intramural Research Program Translational Analytical Core, National Institutes of Health, Baltimore, MD, USA.
- Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA.
- Medication Development Program, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA.
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
3
|
De Oliveira Sergio T, Jane Smith R, Wean SE, Engleman EA, Hopf FW. Greater inhibition of female rat binge alcohol intake by adrenergic receptor blockers using a novel Two-Shot rat binge drinking model. Sci Rep 2024; 14:14029. [PMID: 38890353 PMCID: PMC11189554 DOI: 10.1038/s41598-024-64565-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Binge drinking (BD) contributes strongly to the harms of alcohol use disorder. Most rodent models do not result in binge-level blood alcohol concentrations (BACs), and to better understand individual and sex differences in neurobiological mechanisms related to BD, the use of outbred rat strains would be valuable. Here, we developed a novel BD model where after 3+ months of intermittent access to 20% alcohol Wistar rats drank, twice a week, with two 5-min intake (what we called Two-shot) separated by a 10-min break. Our findings showed during Two-Shot that most animals reached ≥ 80 mg% BAC levels (when briefly food-restricted). However, when increasing alcohol concentrations from 20 to 30%, 40%, or 50%, rats titrated to similar intake levels, suggesting rapid sensing of alcohol effects even when front-loading. Two-Shot drinking was reduced in both sexes by naltrexone (1 mg/kg), validating intake suppression by a clinical therapeutic agent for human problem drinking. Further, both propranolol (β-adrenergic receptor antagonist) and prazosin (α1-adrenergic receptor antagonist) reduced female but not male BD at the lower dose. Thus, our results provide a novel model for BD in outbred rats and suggest that female binging is more sensitive to adrenergic modulation than males, perhaps providing a novel sex-related therapy.
Collapse
Affiliation(s)
- Thatiane De Oliveira Sergio
- Department of Psychiatry, Indiana University School of Medicine, 320 W. 15th Street, NB 300E, Indianapolis, IN, 46202, USA
| | - Rebecca Jane Smith
- Department of Psychiatry, Indiana University School of Medicine, 320 W. 15th Street, NB 300E, Indianapolis, IN, 46202, USA
| | - Sarah E Wean
- Department of Psychiatry, Indiana University School of Medicine, 320 W. 15th Street, NB 300E, Indianapolis, IN, 46202, USA
| | - Eric A Engleman
- Department of Psychiatry, Indiana University School of Medicine, 320 W. 15th Street, NB 300E, Indianapolis, IN, 46202, USA
| | - Frederic W Hopf
- Department of Psychiatry, Indiana University School of Medicine, 320 W. 15th Street, NB 300E, Indianapolis, IN, 46202, USA.
| |
Collapse
|
4
|
Sergio TDO, Smith RJ, Wean SE, Engleman EA, Hopf FW. Greater inhibition of female rat binge alcohol intake by adrenergic receptor blockers using a novel Two-Shot rat binge drinking model. RESEARCH SQUARE 2024:rs.3.rs-4402198. [PMID: 38853968 PMCID: PMC11160926 DOI: 10.21203/rs.3.rs-4402198/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Binge drinking (BD) contributes strongly to the harms of alcohol use disorder. Most rodent models do not result in binge-level blood alcohol concentrations (BACs), and to better understand individual and sex differences in neurobiological mechanisms related to BD, the use of outbred rat strains would be valuable. Here, we developed a novel BD model where after 3+ months of intermittent access to 20% alcohol Wistar rats drank, twice a week, with two 5-minute intake (what we called Two-shot) separated by a 10-minute break. Our findings showed during Two-Shot that most animals reached ≥ 80mg% BAC levels (when briefly food-restricted). However, when increasing alcohol concentrations from 20% to 30%, 40%, or 50%, rats titrated to similar intake levels, suggesting rapid sensing of alcohol effects even when front-loading. Two-Shot drinking was reduced in both sexes by naltrexone (1mg/kg), validating intake suppression by a clinical therapeutic agent. Further, both propranolol (β adrenergic receptor antagonist) and prazosin (α1 adrenergic receptor antagonist) reduced female but not male BD at the lower dose. Thus, our results provide a novel model for BD in outbred rats and suggest that female binging is more sensitive to adrenergic modulation than males, perhaps providing a novel sex-related therapy.
Collapse
Affiliation(s)
| | - Rebecca Jane Smith
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis IN
| | - Sarah E Wean
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis IN
| | - Eric A Engleman
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis IN
| | - Frederic W Hopf
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis IN
| |
Collapse
|
5
|
Koob GF, Vendruscolo L. Theoretical Frameworks and Mechanistic Aspects of Alcohol Addiction: Alcohol Addiction as a Reward Deficit/Stress Surfeit Disorder. Curr Top Behav Neurosci 2023. [PMID: 37421551 DOI: 10.1007/7854_2023_424] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
Alcohol use disorder (AUD) can be defined by a compulsion to seek and take alcohol, the loss of control in limiting intake, and the emergence of a negative emotional state when access to alcohol is prevented. Alcohol use disorder impacts multiple motivational mechanisms and can be conceptualized as a disorder that includes a progression from impulsivity (positive reinforcement) to compulsivity (negative reinforcement). Compulsive drug seeking that is associated with AUD can be derived from multiple neuroadaptations, but the thesis argued herein is that a key component involves the construct of negative reinforcement. Negative reinforcement is defined as drug taking that alleviates a negative emotional state. The negative emotional state that drives such negative reinforcement is hypothesized to derive from the dysregulation of specific neurochemical elements that are involved in reward and stress within basal forebrain structures that involve the ventral striatum and extended amygdala, respectively. Specific neurochemical elements in these structures include decreases in reward neurotransmission (e.g., decreases in dopamine and opioid peptide function in the ventral striatum) and the recruitment of brain stress systems (e.g., corticotropin-releasing factor [CRF]) in the extended amygdala, which contributes to hyperkatifeia and greater alcohol intake that is associated with dependence. Glucocorticoids and mineralocorticoids may play a role in sensitizing the extended amygdala CRF system. Other components of brain stress systems in the extended amygdala that may contribute to the negative motivational state of withdrawal include norepinephrine in the bed nucleus of the stria terminalis, dynorphin in the nucleus accumbens, hypocretin and vasopressin in the central nucleus of the amygdala, and neuroimmune modulation. Decreases in the activity of neuropeptide Y, nociception, endocannabinoids, and oxytocin in the extended amygdala may also contribute to hyperkatifeia that is associated with alcohol withdrawal. Such dysregulation of emotional processing may also significantly contribute to pain that is associated with alcohol withdrawal and negative urgency (i.e., impulsivity that is associated with hyperkatifeia during hyperkatifeia). Thus, an overactive brain stress response system is hypothesized to be activated by acute excessive drug intake, to be sensitized during repeated withdrawal, to persist into protracted abstinence, and to contribute to the compulsivity of AUD. The combination of the loss of reward function and recruitment of brain stress systems provides a powerful neurochemical basis for a negative emotional state that is responsible for the negative reinforcement that at least partially drives the compulsivity of AUD.
Collapse
Affiliation(s)
- George F Koob
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| | - Leandro Vendruscolo
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
6
|
Athanason A, Nadav T, Cates-Gatto C, Roberts A, Roberto M, Varodayan F. Chronic ethanol alters adrenergic receptor gene expression and produces cognitive deficits in male mice. Neurobiol Stress 2023; 24:100542. [PMID: 37197395 PMCID: PMC10184141 DOI: 10.1016/j.ynstr.2023.100542] [Citation(s) in RCA: 2] [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: 02/06/2023] [Revised: 04/12/2023] [Accepted: 04/27/2023] [Indexed: 05/19/2023] Open
Abstract
Hyperkateifia and stress-induced alcohol cravings drive relapse in individuals with alcohol use disorder (AUD). The brain stress signal norepinephrine (also known as noradrenaline) tightly controls cognitive and affective behavior and was thought to be broadly dysregulated with AUD. The locus coeruleus (LC) is a major source of forebrain norepinephrine, and it was recently discovered that the LC sends distinct projections to addiction-associated regions suggesting that alcohol-induced noradrenergic changes may be more brain region-specific than originally thought. Here we investigated whether ethanol dependence alters adrenergic receptor gene expression in the medial prefrontal cortex (mPFC) and central amgydala (CeA), as these regions mediate the cognitive impairment and negative affective state of ethanol withdrawal. We exposed male C57BL/6J mice to the chronic intermittent ethanol vapor-2 bottle choice paradigm (CIE-2BC) to induce ethanol dependence, and assessed reference memory, anxiety-like behavior and adrenergic receptor transcript levels during 3-6 days of withdrawal. Dependence bidirectionally altered mouse brain α1 and β receptor mRNA levels, potentially leading to reduced mPFC adrenergic signaling and enhanced noradrenergic influence over the CeA. These brain region-specific gene expression changes were accompanied by long-term retention deficits and a shift in search strategy in a modified Barnes maze task, as well as greater spontaneous digging behavior and hyponeophagia. Current clinical studies are evaluating adrenergic compounds as a treatment for AUD-associated hyperkatefia, and our findings can contribute to the refinement of these therapies by increasing understanding of the specific neural systems and symptoms that may be targeted.
Collapse
Affiliation(s)
- A.C. Athanason
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - T. Nadav
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - C. Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - A.J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - M. Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - F.P. Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
7
|
Raskind MA, Williams T, Holmes H, Hart K, Crews L, Poupore EL, Thomas RG, Darnell J, Daniels C, Goke K, Hendrickson R, Terry G, Mayer C, Simpson T, Saxon A, Rasmussen D, Peskind ER. A randomized controlled clinical trial of prazosin for alcohol use disorder in active duty soldiers: Predictive effects of elevated cardiovascular parameters. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:348-360. [PMID: 36809662 DOI: 10.1111/acer.14989] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/23/2022] [Accepted: 11/23/2022] [Indexed: 02/24/2023]
Abstract
BACKGROUND Excessive noradrenergic signaling contributes to aversive symptoms of alcohol withdrawal that interfere with abstinence or reductions in harmful use. METHODS To address this aspect of alcohol use disorder, 102 active-duty soldiers participating in command-mandated Army outpatient alcohol treatment were randomized to also receive the brain-penetrant alpha-1 adrenergic receptor antagonist prazosin or placebo for 13 weeks. Primary outcomes were scores on the Penn Alcohol Craving Scale (PACS), standard drink units (SDUs) per day averaged over each week, % days of any drinking per week, and % days of heavy drinking per week. RESULTS PACS declines did not differ significantly between the prazosin and placebo groups in the overall sample. In the subgroup with comorbid PTSD (n = 48), PACS declines were significantly greater in the prazosin than in the placebo condition (p < 0.05). Baseline alcohol consumption was markedly reduced by the pre-randomization outpatient alcohol treatment program, but the addition of prazosin treatment produced a greater slope of decline in SDUs per day compared to placebo (p = 0.01). Preplanned subgroup analyses were performed in soldiers with elevated baseline cardiovascular measures consistent with increased noradrenergic signaling. In soldiers with elevated standing heart rate (n = 15), prazosin reduced SDUs per day (p = 0.01), % days drinking (p = 0.03), and % days heavy drinking (p = 0.001) relative to placebo. In soldiers with elevated standing systolic blood pressure (n = 27), prazosin reduced SDUs per day (p = 0.04) and tended to reduce % days drinking (p = 0.056). Prazosin also reduced depressive symptoms and the incidence of emergent depressed mood more than placebo (p = 0.05 and p = 0.01, respectively). During the final 4 weeks of prazosin vs. placebo treatment that followed completion of Army outpatient AUD treatment, alcohol consumption in soldiers with elevated baseline cardiovascular measures increased in those receiving placebo but remained suppressed in those receiving prazosin. CONCLUSIONS These results extend reports that higher pretreatment cardiovascular measures predict beneficial effects of prazosin, which may be useful for relapse prevention in patients with AUD.
Collapse
Affiliation(s)
- Murray A Raskind
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
| | - Tammy Williams
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Madigan Army Medical Center, Joint Base Lewis-McChord, Tacoma, Washington, USA
| | - Hollie Holmes
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Kim Hart
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Laura Crews
- Madigan Army Medical Center, Joint Base Lewis-McChord, Tacoma, Washington, USA
| | - Eileen L Poupore
- Madigan Army Medical Center, Joint Base Lewis-McChord, Tacoma, Washington, USA
| | | | - Jolee Darnell
- Madigan Army Medical Center, Joint Base Lewis-McChord, Tacoma, Washington, USA
| | - Colin Daniels
- Madigan Army Medical Center, Joint Base Lewis-McChord, Tacoma, Washington, USA
| | - Kevin Goke
- Madigan Army Medical Center, Joint Base Lewis-McChord, Tacoma, Washington, USA
| | - Rebecca Hendrickson
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
| | - Garth Terry
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
| | - Cynthia Mayer
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Tracy Simpson
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
- Center of Excellence in Substance Addiction Treatment and Education, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Andrew Saxon
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
- Center of Excellence in Substance Addiction Treatment and Education, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Dennis Rasmussen
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
| | - Elaine R Peskind
- VA Northwest Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
8
|
Zhan B, Zhu Y, Xia J, Li W, Tang Y, Beesetty A, Ye JH, Fu R. Comorbidity of Post-Traumatic Stress Disorder and Alcohol Use Disorder: Animal Models and Associated Neurocircuitry. Int J Mol Sci 2022; 24:ijms24010388. [PMID: 36613829 PMCID: PMC9820348 DOI: 10.3390/ijms24010388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) and alcohol use disorder (AUD) are prevalent neuropsychiatric disorders and frequently co-occur concomitantly. Individuals suffering from this dual diagnosis often exhibit increased symptom severity and poorer treatment outcomes than those with only one of these diseases. Lacking standard preclinical models limited the exploration of neurobiological mechanisms underlying PTSD and AUD comorbidity. In this review, we summarize well-accepted preclinical model paradigms and criteria for developing successful models of comorbidity. We also outline how PTSD and AUD affect each other bidirectionally in the nervous nuclei have been heatedly discussed recently. We hope to provide potential recommendations for future research.
Collapse
Affiliation(s)
- Bo Zhan
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Yingxin Zhu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Jianxun Xia
- Department of Basic Medical Sciences, Yunkang School of Medicine and Health, Nanfang College, Guangzhou 510970, China
| | - Wenfu Li
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Ying Tang
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Anju Beesetty
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ 07103, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ 07103, USA
- Correspondence: (J.-H.Y.); (R.F.)
| | - Rao Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
- Correspondence: (J.-H.Y.); (R.F.)
| |
Collapse
|
9
|
Curley DE, Vasaturo-Kolodner TR, Cannella N, Ciccocioppo R, Haass-Koffler CL. Yohimbine as a pharmacological probe for alcohol research: a systematic review of rodent and human studies. Neuropsychopharmacology 2022; 47:2111-2122. [PMID: 35760866 PMCID: PMC9556614 DOI: 10.1038/s41386-022-01363-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/12/2022] [Accepted: 06/02/2022] [Indexed: 11/08/2022]
Abstract
Alcohol use disorder (AUD) is a significant public health concern, contributing to a myriad of social, psychological, and physiological issues. Despite substantial efforts within the alcohol research field, promising preclinical findings have failed to translate to clinical use, highlighting the necessity to develop safe and effective pharmacological probes with the ability to be used in preclinical and clinical research. Yohimbine, an α2 adrenergic receptor antagonist, is a well-validated pharmacological tool that has been widely employed in alcohol studies to evaluate noradrenergic activation. This scoping systematic review examines published literature in rodent and human studies involving the use of yohimbine relevant to alcohol research. We conducted a systematic literature review of MEDLINE, Embase, Web of Science Core Collection, CINAHL, PsycInfo, and Cochrane Central Register of Controlled Trials to identify: (1) Experimental Characteristics and Methodology, (2) Sex Differences, (3) Neurochemical Systems and Brain Regions, and (4) Discussion of Applications for Medication Development. Sixty-seven (62 preclinical and 5 clinical) studies were identified meeting the stated criteria, comprising extensive evidence supporting the use of yohimbine as a safe, titratable pharmacological agent for translational alcohol research. Support for the use of yohimbine as a fully translational tool, however, is hindered by limited available findings from human laboratory studies, as well as a dearth of studies examining sex differences in yohimbine's mechanistic actions. Additional consideration should be given to further translational modeling, ideally allowing for parallel preclinical and clinical assessment of yohimbine, methodological assessment of neurochemical systems and brain regions.
Collapse
Affiliation(s)
- Dallece E Curley
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
- Neuroscience Graduate Program, Department of Neuroscience, Brown University, Providence, RI, USA
| | - Talia R Vasaturo-Kolodner
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Carolina L Haass-Koffler
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA.
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI, USA.
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
10
|
Alcohol’s Negative Emotional Side: The Role of Stress Neurobiology in Alcohol Use Disorder. Alcohol Res 2022; 42:12. [PMID: 36338609 PMCID: PMC9621746 DOI: 10.35946/arcr.v42.1.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
This article is part of a Festschrift commemorating the 50th anniversary of the National Institute on Alcohol Abuse and Alcoholism (NIAAA). Established in 1970, first as part of the National Institute of Mental Health and later as an independent institute of the National Institutes of Health, NIAAA today is the world’s largest funding agency for alcohol research. In addition to its own intramural research program, NIAAA supports the entire spectrum of innovative basic, translational, and clinical research to advance the diagnosis, prevention, and treatment of alcohol use disorder and alcohol-related problems. To celebrate the anniversary, NIAAA hosted a 2-day symposium, “Alcohol Across the Lifespan: 50 Years of Evidence-Based Diagnosis, Prevention, and Treatment Research,” devoted to key topics within the field of alcohol research. This article is based on Dr. Sinha’s presentation at the event. NIAAA Director George F. Koob, Ph.D., serves as editor of the Festschrift.
Collapse
|
11
|
Varodayan FP, Patel RR, Matzeu A, Wolfe SA, Curley DE, Khom S, Gandhi PJ, Rodriguez L, Bajo M, D'Ambrosio S, Sun H, Kerr TM, Gonzales RA, Leggio L, Natividad LA, Haass-Koffler CL, Martin-Fardon R, Roberto M. The Amygdala Noradrenergic System Is Compromised With Alcohol Use Disorder. Biol Psychiatry 2022; 91:1008-1018. [PMID: 35430085 PMCID: PMC9167785 DOI: 10.1016/j.biopsych.2022.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a leading preventable cause of death. The central amygdala (CeA) is a hub for stress and AUD, while dysfunction of the noradrenaline stress system is implicated in AUD relapse. METHODS Here, we investigated whether alcohol (ethanol) dependence and protracted withdrawal alter noradrenergic regulation of the amygdala in rodents and humans. Male adult rats were housed under control conditions, subjected to chronic intermittent ethanol vapor exposure to induce dependence, or withdrawn from chronic intermittent ethanol vapor exposure for 2 weeks, and ex vivo electrophysiology, biochemistry (catecholamine quantification by high-performance liquid chromatography), in situ hybridization, and behavioral brain-site specific pharmacology studies were performed. We also used real-time quantitative polymerase chain reaction to assess gene expression of α1B, β1, and β2 adrenergic receptors in human postmortem brain tissue from men diagnosed with AUD and matched control subjects. RESULTS We found that α1 receptors potentiate CeA GABAergic (gamma-aminobutyric acidergic) transmission and drive moderate alcohol intake in control rats. In dependent rats, β receptors disinhibit a subpopulation of CeA neurons, contributing to their excessive drinking. Withdrawal produces CeA functional recovery with no change in local noradrenaline tissue concentrations, although there are some long-lasting differences in the cellular patterns of adrenergic receptor messenger RNA expression. In addition, postmortem brain analyses reveal increased α1B receptor messenger RNA in the amygdala of humans with AUD. CONCLUSIONS CeA adrenergic receptors are key neural substrates of AUD. Identification of these novel mechanisms that drive alcohol drinking, particularly during the alcohol-dependent state, supports ongoing new medication development for AUD.
Collapse
Affiliation(s)
- Florence P Varodayan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California; Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, New York
| | - Reesha R Patel
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California; Systems Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California
| | - Alessandra Matzeu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Sarah A Wolfe
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Dallece E Curley
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island; Neuroscience Graduate Program, Department of Neuroscience, Brown University, Providence, Rhode Island
| | - Sophia Khom
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Pauravi J Gandhi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Larry Rodriguez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Shannon D'Ambrosio
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Hui Sun
- Clinical Core Laboratory, Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Tony M Kerr
- College of Pharmacy, Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, Texas
| | - Rueben A Gonzales
- College of Pharmacy, Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, Texas
| | - Lorenzo Leggio
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island; Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland; Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland; Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland; Department of Neuroscience, Georgetown University Medical Center, Washington, DC
| | - Luis A Natividad
- College of Pharmacy, Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, Texas
| | - Carolina L Haass-Koffler
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island; Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island; Carney Institute for Brain Science, Brown University, Providence, Rhode Island; Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland
| | - Rémi Martin-Fardon
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California.
| |
Collapse
|
12
|
Yang W, Singla R, Maheshwari O, Fontaine CJ, Gil-Mohapel J. Alcohol Use Disorder: Neurobiology and Therapeutics. Biomedicines 2022; 10:1192. [PMID: 35625928 PMCID: PMC9139063 DOI: 10.3390/biomedicines10051192] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
Alcohol use disorder (AUD) encompasses the dysregulation of multiple brain circuits involved in executive function leading to excessive consumption of alcohol, despite negative health and social consequences and feelings of withdrawal when access to alcohol is prevented. Ethanol exerts its toxicity through changes to multiple neurotransmitter systems, including serotonin, dopamine, gamma-aminobutyric acid, glutamate, acetylcholine, and opioid systems. These neurotransmitter imbalances result in dysregulation of brain circuits responsible for reward, motivation, decision making, affect, and the stress response. Despite serious health and psychosocial consequences, this disorder still remains one of the leading causes of death globally. Treatment options include both psychological and pharmacological interventions, which are aimed at reducing alcohol consumption and/or promoting abstinence while also addressing dysfunctional behaviours and impaired functioning. However, stigma and social barriers to accessing care continue to impact many individuals. AUD treatment should focus not only on restoring the physiological and neurological impairment directly caused by alcohol toxicity but also on addressing psychosocial factors associated with AUD that often prevent access to treatment. This review summarizes the impact of alcohol toxicity on brain neurocircuitry in the context of AUD and discusses pharmacological and non-pharmacological therapies currently available to treat this addiction disorder.
Collapse
Affiliation(s)
- Waisley Yang
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (W.Y.); (R.S.)
| | - Rohit Singla
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (W.Y.); (R.S.)
| | - Oshin Maheshwari
- Psychiatry Residency Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8W 3P5, Canada;
| | | | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (W.Y.); (R.S.)
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada;
| |
Collapse
|
13
|
Downs AM, McElligott ZA. Noradrenergic circuits and signaling in substance use disorders. Neuropharmacology 2022; 208:108997. [PMID: 35176286 PMCID: PMC9498225 DOI: 10.1016/j.neuropharm.2022.108997] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 12/20/2022]
Abstract
The central noradrenergic system innervates almost all regions of the brain and, as such, is well positioned to modulate many neural circuits implicated in behaviors and physiology underlying substance use disorders. Ample pharmacological evidence demonstrates that α1, α2, and β adrenergic receptors may serve as therapeutic targets to reduce drug -seeking behavior and drug withdrawal symptoms. Further, norepinephrine is a key modulator of the stress response, and stress has been heavily implicated in reinstatement of drug taking. In this review, we discuss recent advances in our understanding of noradrenergic circuitry and noradrenergic receptor signaling in the context of opioid, alcohol, and psychostimulant use disorders.
Collapse
Affiliation(s)
- Anthony M Downs
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Zoe A McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
14
|
Is There a Novel Biosynthetic Pathway in Mice That Converts Alcohol to Dopamine, Norepinephrine and Epinephrine? Molecules 2022; 27:molecules27092726. [PMID: 35566075 PMCID: PMC9105937 DOI: 10.3390/molecules27092726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/21/2022] [Indexed: 11/29/2022] Open
Abstract
Previous studies in animals and humans have shown multiple types of interaction between alcohol (ethanol) intake and the catecholamine signaling molecules: dopamine, norepinephrine and epinephrine. This literature suggests that the administration of alcohol to rodents affects the central and peripheral (blood plasma) levels of these catecholamines. Two prior publications (Fitzgerald 2012, 2020) put forth the hypothesis that there may be a currently unidentified biosynthetic pathway, in a range of organisms, that actually converts alcohol to dopamine, norepinephrine and epinephrine. This publication describes the details for how to test this hypothesis in mice. Mice can be systemically injected with an intoxicating dose of commercially available stable isotope-labeled ethanol (ethanol-1-13C), and blood plasma samples and brains can be collected approximately two to 24 h post-injection. Liquid chromatography-mass spectrometry analysis can then be used to test whether some of the labeled ethanol molecules have been incorporated into new dopamine, norepinephrine, and epinephrine molecules, in plasma and brain samples. If confirmed, this hypothesis may have broadly reaching implications both for basic neuroscience and our understanding of alcohol abuse and alcoholism.
Collapse
|
15
|
Sinha R, Fogelman N, Wemm S, Angarita G, Seo D, Hermes G. Alcohol withdrawal symptoms predict corticostriatal dysfunction that is reversed by prazosin treatment in alcohol use disorder. Addict Biol 2022; 27:e13116. [PMID: 34856641 PMCID: PMC9872962 DOI: 10.1111/adb.13116] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 01/27/2023]
Abstract
Chronic alcohol use increases risk of alcohol withdrawal symptoms (AW) and disrupts stress biology and resilient coping, thereby promoting excessive alcohol intake. Chronic alcohol intake and multiple alcohol detoxifications are known to impair brain medial prefrontal cortex (mPFC) and striatal functioning, regions involved in regulating stress, craving and alcohol intake. In two related studies, we examined whether AW predicts this functional brain pathology and whether Prazosin versus Placebo treatment may reverse these effects. In Study 1, patients with Alcohol Use Disorder (AUD) (N = 45) with varying AW levels at treatment entry were assessed to examine AW effects on corticostriatal responses to stress, alcohol cue and neutral visual images with functional magnetic resonance imaging (fMRI). In Study 2, 23 AUD patients entering a 12-week randomised controlled trial (RCT) of Prazosin, an alpha1 adrenergic antagonist that decreased withdrawal-related alcohol intake in laboratory animals, participated in two fMRI sessions at pretreatment and also at week 9-10 of chronic treatment (Placebo: N = 13; Prazosin: N = 10) to assess Prazosin treatment effects on alcohol-related cortico-striatal dysfunction. Study 1 results indicated that higher AW predicted greater disruption in brain mPFC and striatal response to stress and alcohol cues (p < 0.001, family-wise error [FWE] correction) and also subsequently greater heavy drinking days (HDD) in early treatment (p < 0.01). In Study 2, Prazosin versus Placebo treatment reversed mPFC-striatal dysfunction (p < 0.001, FWE), which in turn predicted fewer drinking days (p < 0.01) during the 12-week treatment period. These results indicate that AW is a significant predictor of alcohol-related prefrontal-striatal dysfunction, and Prazosin treatment reversed these effects that in turn contributed to improved alcohol treatment outcomes.
Collapse
Affiliation(s)
- Rajita Sinha
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nia Fogelman
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Stephanie Wemm
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gustavo Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dongju Seo
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gretchen Hermes
- Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
16
|
Abstract
Alcohol use disorder (AUD) is a highly prevalent but severely under-treated disorder, with only three widely-approved pharmacotherapies. Given that AUD is a very heterogeneous disorder, it is unlikely that one single medication will be effective for all individuals with an AUD. As such, there is a need to develop new, more effective, and diverse pharmacological treatment options for AUD with the hopes of increasing utilization and improving care. In this qualitative literature review, we discuss the efficacy, mechanism of action, and tolerability of approved, repurposed, and novel pharmacotherapies for the treatment of AUD with a clinical perspective. Pharmacotherapies discussed include: disulfiram, acamprosate, naltrexone, nalmefene, topiramate, gabapentin, varenicline, baclofen, sodium oxybate, aripiprazole, ondansetron, mifepristone, ibudilast, suvorexant, prazosin, doxazosin, N-acetylcysteine, GET73, ASP8062, ABT-436, PF-5190457, and cannabidiol. Overall, many repurposed and novel agents discussed in this review demonstrate clinical effectiveness and promise for the future of AUD treatment. Importantly, these medications also offer potential improvements towards the advancement of precision medicine and personalized treatment for the heterogeneous AUD population. However, there remains a great need to improve access to treatment, increase the menu of approved pharmacological treatments, and de-stigmatize and increase treatment-seeking for AUD.
Collapse
|
17
|
Grinevich VP, Krupitsky EM, Gainetdinov RR, Budygin EA. Linking Ethanol-Addictive Behaviors With Brain Catecholamines: Release Pattern Matters. Front Behav Neurosci 2022; 15:795030. [PMID: 34975429 PMCID: PMC8716449 DOI: 10.3389/fnbeh.2021.795030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/26/2021] [Indexed: 12/30/2022] Open
Abstract
Using a variety of animal models that simulate key features of the alcohol use disorder (AUD), remarkable progress has been made in identifying neurochemical targets that may contribute to the development of alcohol addiction. In this search, the dopamine (DA) and norepinephrine (NE) systems have been long thought to play a leading role in comparison with other brain systems. However, just recent development and application of optogenetic approaches into the alcohol research field provided opportunity to identify neuronal circuits and specific patterns of neurotransmission that govern the key components of ethanol-addictive behaviors. This critical review summarizes earlier findings, which initially disclosed catecholamine substrates of ethanol actions in the brain and shows how the latest methodologies help us to reveal the significance of DA and NE release changes. Specifically, we focused on recent optogenetic investigations aimed to reveal cause-effect relationships between ethanol-drinking (seeking and taking) behaviors and catecholamine dynamics in distinct brain pathways. These studies gain the knowledge that is needed for the better understanding addiction mechanisms and, therefore, for development of more effective AUD treatments. Based on the reviewed findings, new messages for researches were indicated, which may have broad applications beyond the field of alcohol addiction.
Collapse
Affiliation(s)
- Vladimir P Grinevich
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia
| | - Evgeny M Krupitsky
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, St. Petersburg, Russia.,Laboratory of Clinical Psychopharmacology of Addictions, St.-Petersburg First Pavlov State Medical University, St. Petersburg, Russia
| | - Raul R Gainetdinov
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia.,Institute of Translational Biomedicine and St. Petersburg State University Hospital, St. Petersburg State University, St. Petersburg, Russia
| | - Evgeny A Budygin
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
18
|
Minnig MA, Park T, Echeveste Sanchez M, Cottone P, Sabino V. Viral-Mediated Knockdown of Nucleus Accumbens Shell PAC1 Receptor Promotes Excessive Alcohol Drinking in Alcohol-Preferring Rats. Front Behav Neurosci 2021; 15:787362. [PMID: 34924973 PMCID: PMC8678417 DOI: 10.3389/fnbeh.2021.787362] [Citation(s) in RCA: 4] [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: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 01/04/2023] Open
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing disorder whose genetic and environmental susceptibility components are not fully understood. Neuropeptidergic signaling has been repeatedly implicated in modulating excessive alcohol drinking, especially within sub-regions of the striatum. Here, we investigated the potential involvement of the selective receptor for pituitary adenylate cyclase-activating polypeptide (PACAP), PAC1R, in the nucleus accumbens shell (NAcc Shell) in excessive alcohol drinking in alcohol-preferring rats, an established animal model of the genetic propensity for alcoholism. Scr:sP alcohol-preferring rats were trained to operantly self-administer alcohol and then either an AAV virus short-hairpin RNA (shRNA) targeted to knockdown PAC1R, or an AAV control virus were microinfused into the NAcc Shell. NAcc Shell PAC1R shRNA knockdown virus was confirmed to significantly decrease PAC1R levels in the NAcc Shell. The effects of NAcc Shell PAC1R shRNA knockdown on ethanol self-administration were investigated using a Fixed Ratio (FR) 1 and a Progressive Ratio (PR) schedule of reinforcement. The effect of PAC1R knockdown on self-administration of an alternative reinforcer, saccharin, was also assessed. The results showed that the reduction in PAC1R in the NAcc Shell led to excessive ethanol drinking, increased preference for ethanol, and higher motivation to drink. NAcc Shell PAC1R shRNA knockdown did not comparably increase saccharin self-administration, suggesting selectivity of action. These data suggest that NAcc Shell PAC1R may serves as a "brake" on alcohol drinking, and thereby the loss of function of PAC1R leads to excessive alcohol consumption. Therefore, the PACAP/PAC1R system may represent a novel target for the treatment of AUD.
Collapse
Affiliation(s)
| | | | | | | | - Valentina Sabino
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
19
|
De Oliveira Sergio T, Lei K, Kwok C, Ghotra S, Wegner SA, Walsh M, Waal J, Darevsky D, Hopf FW. The role of anterior insula-brainstem projections and alpha-1 noradrenergic receptors for compulsion-like and alcohol-only drinking. Neuropsychopharmacology 2021; 46:1918-1926. [PMID: 34168279 PMCID: PMC8429444 DOI: 10.1038/s41386-021-01071-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 02/06/2023]
Abstract
Compulsion-like alcohol drinking (CLAD), where consumption continues despite negative consequences, is a major obstacle to treating alcohol use disorder. The locus coeruleus area in the brainstem and norepinephrine receptor (NER) signaling in forebrain cortical regions have been implicated in adaptive responding under stress, which is conceptually similar to compulsion-like responding (adaptive responding despite the presence of stress or conflict). Thus, we examined whether anterior insula (aINS)-to-brainstem connections and alpha-1 NERs regulated compulsion-like intake and alcohol-only drinking (AOD). Halorhodopsin inhibition of aINS-brainstem significantly reduced CLAD, with no effect on alcohol-only or saccharin intake, suggesting a specific aINS-brainstem role in aversion-resistant drinking. In contrast, prazosin inhibition of alpha-1 NERs systemically reduced both CLAD and AOD. Similar to systemic inhibition, intra-aINS alpha-1-NER antagonism reduced both CLAD and AOD. Global aINS inhibition with GABAR agonists also strongly reduced both CLAD and AOD, without impacting saccharin intake or locomotion, while aINS inhibition of calcium-permeable AMPARs (with NASPM) reduced CLAD without impacting AOD. Finally, prazosin inhibition of CLAD and AOD was not correlated with each other, systemically or within aINS, suggesting the possibility that different aINS pathways regulate CLAD versus AOD, which will require further study to definitively address. Together, our results provide important new information showing that some aINS pathways (aINS-brainstem and NASPM-sensitive) specifically regulate compulsion-like alcohol consumption, while aINS more generally may contain parallel pathways promoting CLAD versus AOD. These findings also support the importance of the adaptive stress response system for multiple forms of alcohol drinking.
Collapse
Affiliation(s)
- Thatiane De Oliveira Sergio
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Kelly Lei
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Claudina Kwok
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Shahbaj Ghotra
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Scott A Wegner
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Margaret Walsh
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Jaclyn Waal
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - David Darevsky
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Frederic W Hopf
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
20
|
Cameron CM, Nieto S, Bosler L, Wong M, Bishop I, Mooney L, Cahill CM. Mechanisms Underlying the Anti-Suicidal Treatment Potential of Buprenorphine. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2021; 1. [PMID: 35265942 PMCID: PMC8903193 DOI: 10.3389/adar.2021.10009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Death by suicide is a global epidemic with over 800 K suicidal deaths worlwide in 2012. Suicide is the 10th leading cause of death among Americans and more than 44 K people died by suicide in 2019 in the United States. Patients with chronic pain, including, but not limited to, those with substance use disorders, are particularly vulnerable. Chronic pain patients have twice the risk of death by suicide compared to those without pain, and 50% of chronic pain patients report that they have considered suicide at some point due to their pain. The kappa opioid system is implicated in negative mood states including dysphoria, depression, and anxiety, and recent evidence shows that chronic pain increases the function of this system in limbic brain regions important for affect and motivation. Additionally, dynorphin, the endogenous ligand that activates the kappa opioid receptor is increased in the caudate putamen of human suicide victims. A potential treatment for reducing suicidal ideation and suicidal attempts is buprenorphine. Buprenorphine, a partial mu opioid agonist with kappa opioid antagonist properties, reduced suicidal ideation in chronic pain patients with and without an opioid use disorder. This review will highlight the clinical and preclinical evidence to support the use of buprenorphine in mitigating pain-induced negative affective states and suicidal thoughts, where these effects are at least partially mediated via its kappa antagonist properties.
Collapse
Affiliation(s)
- Courtney M. Cameron
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven Nieto
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lucienne Bosler
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Megan Wong
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Isabel Bishop
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Larissa Mooney
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Catherine M. Cahill
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
- Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Correspondence: Catherine M. Cahill,
| |
Collapse
|
21
|
Vanderkam P, Solinas M, Ingrand I, Doux N, Ebrahimighavam S, Jaafari N, Lafay-Chebassier C. Effectiveness of drugs acting on adrenergic receptors in the treatment for tobacco or alcohol use disorders: systematic review and meta-analysis. Addiction 2021; 116:1011-1020. [PMID: 32959918 DOI: 10.1111/add.15265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/08/2020] [Accepted: 09/11/2020] [Indexed: 11/27/2022]
Abstract
AIM To assess the efficacy of drugs directly acting on alpha- and beta-adrenergic receptors in the treatment of patients suffering from tobacco or alcohol use disorder. METHODS Using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, studies were identified through PUBMED, EMBASE, the Cochrane Central Register of Controlled Trials and clinicaltrial.gov. We selected only randomized controlled trials with adult patients with tobacco or alcohol use disorders according to DSM-5 criteria. Interventions included any molecule having a direct pharmacological action on alpha- or beta-adrenergic receptors (agonist or antagonist). Comparators were placebo or other validated pharmacotherapies. The duration of the intervention was a minimum of 1 month, with 3 months of follow-up. Measurements included smoking cessation for tobacco; for alcohol, we selected abstinence, alcohol consumption (drinks per day or week) and heavy drinking days (HDD). Ten studies with tobacco and six with alcohol use disorder were included in the qualitative synthesis and fifteen studies in the quantitative analysis. RESULTS We found that clonidine, an alpha-2 agonist, significantly increased smoking abstinence [relative risk = 1.39 with a 95% confidence interval (CI) = 1.04, 1.84]. Beta-blockers had no significant effect on smoking abstinence. The alpha-1 antagonists prazosin and doxazosin decreased alcohol consumption [SMD = -0.32 (-0.56, -0.07)] but had no effect on abstinence or HDD. CONCLUSIONS The noradrenaline system may represent a promising mechanism to target in tobacco and alcohol use disorders.
Collapse
Affiliation(s)
- Paul Vanderkam
- INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, Poitiers, France
- Unité de recherche clinique intersectorielle en psychiatrie, Centre Hospitalier Henri Laborit, Poitiers, France
| | - Marcello Solinas
- INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, Poitiers, France
- Unité de recherche clinique intersectorielle en psychiatrie, Centre Hospitalier Henri Laborit, Poitiers, France
| | - Isabelle Ingrand
- Service de Pharmacologie Clinique et Vigilances, CHU de Poitiers, Poitiers, France
- Unité d'Epidémiologie et Biostatistique, Registre Général des Cancers Poitou-Charentes, INSERM CIC 1402, Université, CHU de Poitiers, France
| | - Nicolas Doux
- Service Commun de Documentation, Bibliothèque universitaire de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France
| | - Soghra Ebrahimighavam
- Department of Educational Psychology, Faculty of Psychology and Educational Science, Allameh Tabatabai University, Iran
| | - Nematollah Jaafari
- Unité de recherche clinique intersectorielle en psychiatrie, Centre Hospitalier Henri Laborit, Poitiers, France
| | - Claire Lafay-Chebassier
- INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, Poitiers, France
- Service de Pharmacologie Clinique et Vigilances, CHU de Poitiers, Poitiers, France
| |
Collapse
|
22
|
Sinha R, Wemm S, Fogelman N, Milivojevic V, Morgan PM, Angarita GA, Hermes G, Fox HC. Moderation of Prazosin's Efficacy by Alcohol Withdrawal Symptoms. Am J Psychiatry 2021; 178:447-458. [PMID: 33207935 PMCID: PMC8119326 DOI: 10.1176/appi.ajp.2020.20050609] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Alcohol use disorder (AUD) is a leading cause of global disease burden. Chronic, heavy use increases the likelihood of alcohol withdrawal symptoms and associated secondary outcomes of alcohol craving and mood, anxiety, and sleep disturbances, which are predictive of poor treatment outcomes. The authors examined whether alcohol withdrawal symptoms moderate the efficacy of prazosin in reducing alcohol intake and associated secondary outcomes. METHODS A 12-week, double-blind, randomized, controlled proof-of-concept trial of prazosin (16 mg/day, with a 2-week titration) was conducted in community-recruited adults with current alcohol dependence (N=100) with varying levels of alcohol withdrawal symptoms assessed at treatment entry. Primary outcomes were daily self-reported drinking days and heavy drinking days, and secondary outcomes were average drinks/day and mood, anxiety, craving, and sleep quality ratings. RESULTS Modified intent-to-treat analyses indicated a significant interaction of alcohol withdrawal symptom score by treatment by full-dose treatment period (weeks 3-12) for drinking days, heavy drinking days, and average drinks/day. By week 12, participants with high alcohol withdrawal symptoms on prazosin reported 7.07% heavy drinking days and 27.46% drinking days, while those on placebo had 35.58% heavy drinking days and 58.47% drinking days (heavy drinking days: odds ratio=0.14, 95% CI=0.058, 0.333; drinking days: odds ratio=0.265, 95% CI=0.146, 0.481). No such benefit of prazosin was observed in those reporting low or no alcohol withdrawal symptoms. Individuals with high alcohol withdrawal symptoms on prazosin compared with placebo also showed significantly improved anxiety, depression, and alcohol craving over the course of the trial. CONCLUSIONS The findings indicate that alcohol withdrawal symptoms are a significant moderator of prazosin treatment response for alcohol use outcomes and for associated symptoms of alcohol craving, anxiety, and mood symptoms. These data support further evaluation of alcohol withdrawal symptoms as a prognostic indicator of prazosin's efficacy in the treatment of AUD.
Collapse
Affiliation(s)
- Rajita Sinha
- Yale Stress Center (Sinha, Wemm, Fogelman, Milivojevic, Hermes), Department of Psychiatry (Sinha, Wemm, Fogelman, Milivojevic, Morgan, Angarita, Hermes), and Department of Neuroscience (Sinha), Yale University School of Medicine, New Haven, Conn.; Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, N.Y. (Fox)
| | - Stephanie Wemm
- Yale Stress Center (Sinha, Wemm, Fogelman, Milivojevic, Hermes), Department of Psychiatry (Sinha, Wemm, Fogelman, Milivojevic, Morgan, Angarita, Hermes), and Department of Neuroscience (Sinha), Yale University School of Medicine, New Haven, Conn.; Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, N.Y. (Fox)
| | - Nia Fogelman
- Yale Stress Center (Sinha, Wemm, Fogelman, Milivojevic, Hermes), Department of Psychiatry (Sinha, Wemm, Fogelman, Milivojevic, Morgan, Angarita, Hermes), and Department of Neuroscience (Sinha), Yale University School of Medicine, New Haven, Conn.; Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, N.Y. (Fox)
| | - Verica Milivojevic
- Yale Stress Center (Sinha, Wemm, Fogelman, Milivojevic, Hermes), Department of Psychiatry (Sinha, Wemm, Fogelman, Milivojevic, Morgan, Angarita, Hermes), and Department of Neuroscience (Sinha), Yale University School of Medicine, New Haven, Conn.; Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, N.Y. (Fox)
| | - Peter M Morgan
- Yale Stress Center (Sinha, Wemm, Fogelman, Milivojevic, Hermes), Department of Psychiatry (Sinha, Wemm, Fogelman, Milivojevic, Morgan, Angarita, Hermes), and Department of Neuroscience (Sinha), Yale University School of Medicine, New Haven, Conn.; Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, N.Y. (Fox)
| | - Gustavo A Angarita
- Yale Stress Center (Sinha, Wemm, Fogelman, Milivojevic, Hermes), Department of Psychiatry (Sinha, Wemm, Fogelman, Milivojevic, Morgan, Angarita, Hermes), and Department of Neuroscience (Sinha), Yale University School of Medicine, New Haven, Conn.; Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, N.Y. (Fox)
| | - Gretchen Hermes
- Yale Stress Center (Sinha, Wemm, Fogelman, Milivojevic, Hermes), Department of Psychiatry (Sinha, Wemm, Fogelman, Milivojevic, Morgan, Angarita, Hermes), and Department of Neuroscience (Sinha), Yale University School of Medicine, New Haven, Conn.; Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, N.Y. (Fox)
| | - Helen C Fox
- Yale Stress Center (Sinha, Wemm, Fogelman, Milivojevic, Hermes), Department of Psychiatry (Sinha, Wemm, Fogelman, Milivojevic, Morgan, Angarita, Hermes), and Department of Neuroscience (Sinha), Yale University School of Medicine, New Haven, Conn.; Department of Psychiatry, Stony Brook University School of Medicine, Stony Brook, N.Y. (Fox)
| |
Collapse
|
23
|
Buhr TJ, Reed CH, Shoeman A, Bauer EE, Valentine RJ, Clark PJ. The Influence of Moderate Physical Activity on Brain Monoaminergic Responses to Binge-Patterned Alcohol Ingestion in Female Mice. Front Behav Neurosci 2021; 15:639790. [PMID: 33716684 PMCID: PMC7947191 DOI: 10.3389/fnbeh.2021.639790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/25/2021] [Indexed: 01/16/2023] Open
Abstract
Monoamine neurotransmitter activity in brain reward, limbic, and motor areas play key roles in the motivation to misuse alcohol and can become modified by exercise in a manner that may affect alcohol craving. This study investigated the influence of daily moderate physical activity on monoamine-related neurochemical concentrations across the mouse brain in response to high volume ethanol ingestion. Adult female C57BL/6J mice were housed with or without 2.5 h of daily access to running wheels for 30 days. On the last 5 days, mice participated in the voluntary binge-like ethanol drinking procedure, “Drinking in the dark” (DID). Mice were sampled immediately following the final episode of DID. Monoamine-related neurochemical concentrations were measured across brain regions comprising reward, limbic, and motor circuits using ultra High-Performance Liquid Chromatography (UHPLC). The results suggest that physical activity status did not influence ethanol ingestion during DID. Moreover, daily running wheel access only mildly influenced alcohol-related norepinephrine concentrations in the hypothalamus and prefrontal cortex, as well as serotonin turnover in the hippocampus. However, access to alcohol during DID eliminated wheel running-related decreases of norepinephrine, serotonin, and 5-HIAA content in the hypothalamus, but also to a lesser extent for norepinephrine in the hippocampus and caudal cortical areas. Finally, alcohol access increased serotonin and dopamine-related neurochemical turnover in the striatum and brainstem areas, regardless of physical activity status. Together, these data provide a relatively thorough assessment of monoamine-related neurochemical levels across the brain in response to voluntary binge-patterned ethanol drinking, but also adds to a growing body of research questioning the utility of moderate physical activity as an intervention to curb alcohol abuse.
Collapse
Affiliation(s)
- Trevor J Buhr
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States.,Neuroscience Program, Iowa State University, Ames, IA, United States
| | - Carter H Reed
- Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States.,Department of Kinesiology, Iowa State University, Ames, IA, United States
| | - Allyse Shoeman
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States.,Neuroscience Program, Iowa State University, Ames, IA, United States
| | - Ella E Bauer
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States.,Neuroscience Program, Iowa State University, Ames, IA, United States.,Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States
| | - Rudy J Valentine
- Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States.,Department of Kinesiology, Iowa State University, Ames, IA, United States
| | - Peter J Clark
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States.,Neuroscience Program, Iowa State University, Ames, IA, United States.,Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States
| |
Collapse
|
24
|
Drug addiction co-morbidity with alcohol: Neurobiological insights. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 157:409-472. [PMID: 33648675 DOI: 10.1016/bs.irn.2020.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Addiction is a chronic disorder that consists of a three-stage cycle of binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation. These stages involve, respectively, neuroadaptations in brain circuits involved in incentive salience and habit formation, stress surfeit and reward deficit, and executive function. Much research on addiction focuses on the neurobiology underlying single drug use. However, alcohol use disorder (AUD) can be co-morbid with substance use disorder (SUD), called dual dependence. The limited epidemiological data on dual dependence indicates that there is a large population of individuals suffering from addiction who are dependent on more than one drug and/or alcohol, yet dual dependence remains understudied in addiction research. Here, we review neurobiological data on neurotransmitter and neuropeptide systems that are known to contribute to addiction pathology and how the involvement of these systems is consistent or divergent across drug classes. In particular, we highlight the dopamine, opioid, corticotropin-releasing factor, norepinephrine, hypocretin/orexin, glucocorticoid, neuroimmune signaling, endocannabinoid, glutamate, and GABA systems. We also discuss the limited research on these systems in dual dependence. Collectively, these studies demonstrate that the use of multiple drugs can produce neuroadaptations that are distinct from single drug use. Further investigation into the neurobiology of dual dependence is necessary to develop effective treatments for addiction to multiple drugs.
Collapse
|
25
|
Koob GF. Drug Addiction: Hyperkatifeia/Negative Reinforcement as a Framework for Medications Development. Pharmacol Rev 2021; 73:163-201. [PMID: 33318153 PMCID: PMC7770492 DOI: 10.1124/pharmrev.120.000083] [Citation(s) in RCA: 193] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Compulsive drug seeking that is associated with addiction is hypothesized to follow a heuristic framework that involves three stages (binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation) and three domains of dysfunction (incentive salience/pathologic habits, negative emotional states, and executive function, respectively) via changes in the basal ganglia, extended amygdala/habenula, and frontal cortex, respectively. This review focuses on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the addiction cycle. Hyperkatifeia provides an additional source of motivation for compulsive drug seeking via negative reinforcement. Negative reinforcement reflects an increase in the probability of a response to remove an aversive stimulus or drug seeking to remove hyperkatifeia that is augmented by genetic/epigenetic vulnerability, environmental trauma, and psychiatric comorbidity. Neurobiological targets for hyperkatifeia in addiction involve neurocircuitry of the extended amygdala and its connections via within-system neuroadaptations in dopamine, enkephalin/endorphin opioid peptide, and γ-aminobutyric acid/glutamate systems and between-system neuroadaptations in prostress corticotropin-releasing factor, norepinephrine, glucocorticoid, dynorphin, hypocretin, and neuroimmune systems and antistress neuropeptide Y, nociceptin, endocannabinoid, and oxytocin systems. Such neurochemical/neurocircuitry dysregulations are hypothesized to mediate a negative hedonic set point that gradually gains allostatic load and shifts from a homeostatic hedonic state to an allostatic hedonic state. Based on preclinical studies and translational studies to date, medications and behavioral therapies that reset brain stress, antistress, and emotional pain systems and return them to homeostasis would be promising new targets for medication development. SIGNIFICANCE STATEMENT: The focus of this review is on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the drug addiction cycle and a driving force for negative reinforcement in addiction. Medications and behavioral therapies that reverse hyperkatifeia by resetting brain stress, antistress, and emotional pain systems and returning them to homeostasis would be promising new targets for medication development.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
26
|
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]
|
27
|
Lopez MF, Reasons SE, Carper BA, Nolen TL, Williams RL, Becker HC. Evaluation of the effect of doxasozin and zonisamide on voluntary ethanol intake in mice that experienced chronic intermittent ethanol exposure and stress. Alcohol 2020; 89:37-42. [PMID: 32712186 PMCID: PMC7719616 DOI: 10.1016/j.alcohol.2020.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/15/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022]
Abstract
The comorbidity between alcohol use disorder and post-traumatic stress disorder represents a serious health care burden with few effective treatment options. The current study was designed to evaluate the effect of an alpha 1 receptor antagonist (doxazosin) and a novel anticonvulsant (zonisamide) in a model of alcohol (ethanol) dependence and stress exposure. The main dependent variable was voluntary ethanol intake in mice that experienced chronic intermittent ethanol (CIE) exposure and forced swim stress (FSS) alone, and in combination. Adult male and female C57BL/6J mice had access to a single bottle of 15% (v/v) ethanol for 1-hr in the home cage, 3-hr into the dark phase of the light/dark cycle. Once stable ethanol intake was established (~4 weeks), mice were separated into four groups (CTL, CIE, FSS, CIE + FSS). Mice in the FSS condition received 10-min FSS exposure 4-hr prior to drinking sessions (remaining mice were not disturbed). During baseline and the first two test cycles, all mice received vehicle (saline) injections (IP) 30-min before ethanol access. As previously observed, FSS increased ethanol drinking in dependent (CIE-exposed) mice but not in nondependent control (CTL) mice. In the following test cycles mice were evaluated for ethanol intake after administration of doxazosin, zonisamide or their combination. Results indicated that the three doses of doxazosin evaluated significantly reduced voluntary ethanol intake in all mice. Zonisamide had a more modest effect and may require a more prolonged treatment regime. The combined administration of both compounds was not more effective than each drug alone. This study suggests that doxazosin is reliable at reducing voluntary ethanol intake in mice independently of their history of ethanol dependence and stress exposure.
Collapse
Affiliation(s)
- Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Sarah E Reasons
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Benjamin A Carper
- Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, NC, 27709, USA
| | - Tracy L Nolen
- Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, NC, 27709, USA
| | - Rick L Williams
- Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, NC, 27709, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA; RHJ Department of Veterans Affairs Medical Center, Charleston, SC, 29401, USA
| |
Collapse
|
28
|
Vena AA, Zandy SL, Cofresí RU, Gonzales RA. Behavioral, neurobiological, and neurochemical mechanisms of ethanol self-administration: A translational review. Pharmacol Ther 2020; 212:107573. [PMID: 32437827 PMCID: PMC7580704 DOI: 10.1016/j.pharmthera.2020.107573] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2020] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder has multiple characteristics including excessive ethanol consumption, impaired control over drinking behaviors, craving and withdrawal symptoms, compulsive seeking behaviors, and is considered a chronic condition. Relapse is common. Determining the neurobiological targets of ethanol and the adaptations induced by chronic ethanol exposure is critical to understanding the clinical manifestation of alcohol use disorders, the mechanisms underlying the various features of the disorder, and for informing medication development. In the present review, we discuss ethanol's interactions with a variety of neurotransmitter systems, summarizing findings from preclinical and translational studies to highlight recent progress in the field. We then describe animal models of ethanol self-administration, emphasizing the value, limitations, and validity of commonly used models. Lastly, we summarize the behavioral changes induced by chronic ethanol self-administration, with an emphasis on cue-elicited behavior, the role of ethanol-related memories, and the emergence of habitual ethanol seeking behavior.
Collapse
Affiliation(s)
- Ashley A Vena
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, United States of America
| | | | - Roberto U Cofresí
- Psychological Sciences, University of Missouri, United States of America
| | - Rueben A Gonzales
- Division of Pharmacology and Toxicology, College of Pharmacy and Institute for Neuroscience, The University of Texas at Austin, United States of America.
| |
Collapse
|
29
|
Jaime S, Vena AA, Gonzales RA. Intravenous Ethanol Administration and Operant Self-Administration Alter Extracellular Norepinephrine Concentration in the Mesocorticolimbic Systems of Male Long Evans Rats. Alcohol Clin Exp Res 2020; 44:1529-1539. [PMID: 32573991 DOI: 10.1111/acer.14397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/10/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Norepinephrine has been suggested to regulate ethanol (EtOH)-related behaviors, but little is known about the effects of EtOH on norepinephrine release in mesocortical and mesolimbic brain areas that are targets of EtOH actions. METHODS We used in vivo microdialysis to examine the effects of EtOH on extracellular norepinephrine concentrations in mesocorticolimbic brain regions of male Long Evans rats. We determined the effects of intravenous infusion of saline or EtOH in the medial prefrontal cortex (mPFC) and the basal forebrain. We also measured dialysate norepinephrine concentrations during operant self-administration of EtOH in the mPFC. RESULTS Intravenous infusion (1 or 0.25 ml/min) of 1.0 g/kg EtOH stimulated an increase in dialysate norepinephrine in mPFC and in basal forebrain. In the basal forebrain, an infusion of 0.5 g/kg EtOH did not stimulate dialysate norepinephrine concentrations. In both regions, saline infusions did not increase dialysate norepinephrine concentrations. In the behavioral experiment, 1 week of experience with operant self-administration of sweetened EtOH resulted in an apparent reduction in basal dialysate norepinephrine concentrations in the mPFC relative to the sucrose control. Dialysate norepinephrine increased during the transfer from home cage to the operant chamber in all groups. CONCLUSIONS We conclude that acute EtOH stimulates both the locus coeruleus (which projects to the mPFC) and the nucleus tractus solitarius (which projects to the basal forebrain) noradrenergic neurons. Additionally, limited EtOH self-administration experience alters dialysate norepinephrine in the mPFC in a manner consistent with a decrease in tonic norepinephrine release. Further studies are necessary to elucidate the mechanisms by which EtOH exerts these variable effects.
Collapse
Affiliation(s)
- Saul Jaime
- From the, Division of Pharmacology and Toxicology (SJ, RAG), College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Ashley A Vena
- From the, Division of Pharmacology and Toxicology (SJ, RAG), College of Pharmacy, The University of Texas at Austin, Austin, Texas.,Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois
| | - Rueben A Gonzales
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois
| |
Collapse
|
30
|
Hicks C, Sabino V, Cottone P. The Alpha-1 Adrenergic Receptor Antagonist Prazosin Reduces Binge-Like Eating in Rats. Nutrients 2020; 12:nu12061569. [PMID: 32481494 PMCID: PMC7352795 DOI: 10.3390/nu12061569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 01/19/2023] Open
Abstract
Background: Binge-eating disorder is a pervasive addiction-like disorder that is defined by excessive and uncontrollable consumption of food within brief periods of time. The aim of the current study was to examine the role of the brain noradrenergic system in binge-like eating through the use of the alpha-1 adrenergic receptor antagonist prazosin. Methods: For this purpose, we employed a limited access model whereby male Wistar rats were allowed to nosepoke for either chow (Chow rats) or a sugary, highly palatable food (Palatable rats) for 1 h/day. The effects of prazosin (0, 0.5, 1 and 2 mg/kg, i.p.) were tested in a fixed ratio 1 (FR1) and progressive ratio (PR) schedule of reinforcement. Results: The results show that prazosin preferentially reduced the responses for palatable food in a FR1 reinforcement schedule; when tested in a PR schedule of reinforcement, prazosin increased breakpoint in both Chow and Palatable rats, but more potently and more efficaciously in the latter. Our results suggest that prazosin treatment preferentially increased the motivational properties of the palatable diet. Conclusions: The current findings provide the characterization of the effects of prazosin on binge-like eating and offer support to the existing literature showing the important role of the noradrenergic system in addiction-like behavior.
Collapse
|
31
|
Milivojevic V, Angarita GA, Hermes G, Sinha R, Fox HC. Effects of Prazosin on Provoked Alcohol Craving and Autonomic and Neuroendocrine Response to Stress in Alcohol Use Disorder. Alcohol Clin Exp Res 2020; 44:1488-1496. [PMID: 32449942 DOI: 10.1111/acer.14378] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/13/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Chronic alcohol use results in changes to stress biology and autonomic arousal contributing to acute alcohol withdrawal symptoms, neuroendocrine tolerance of the hypothalamic-pituitary-adrenal axis responses, high stress-induced craving, and risk of alcohol relapse. Thus, stress coping and recovery from alcohol during early abstinence may be jeopardized by such stress system dysfunction. Significant preclinical evidence suggests that noradrenergic disruption may contribute to these alcohol-related stress arousal changes and that alpha-1 adrenergic antagonists, such as prazosin, may normalize these stress system adaptations and reduce alcohol intake. Thus, we hypothesized that prazosin would reduce stress-induced craving and improve neuroendocrine and autonomic response to stress and alcohol cue exposure during early abstinence. We secondarily also assessed the role of lifetime anxiety disorders on these prazosin effects. METHODS Forty inpatient treatment-seeking alcohol-dependent individuals were randomly assigned to receive placebo (n = 18) or 16 mg/d, T.I.D., prazosin (n = 22) in a double-blind manner, titrated over 2 weeks. In weeks 3 to 4 after achieving full dose, patients were exposed to 3 5-minute personalized guided imagery conditions (stress cue, alcohol cue, neutral/relaxing cue), on 3 consecutive days in a random, counterbalanced order. Alcohol craving, anxiety, heart rate, cortisol, and adrenocorticotropic hormone (ACTH) levels were assessed at baseline, following imagery and at repeated recovery timepoints. RESULTS Prazosin reduced stress cue-induced alcohol craving (p < 0.05) and stress- and alcohol cue-induced anxiety (p < 0.05) and increased heart rate responses in all imagery conditions (p < 0.05). Prazosin lowered basal cortisol and ACTH (p's < 0.05) and attenuated stress cue-induced rises in cortisol (p < 0.05) versus placebo. Finally, in those without lifetime anxiety disorder, the placebo group showed stress- and alcohol cue-induced increases in cortisol (p's < 0.05), while the prazosin group did not. CONCLUSIONS Prazosin may attenuate stress cue-induced alcohol craving and anxiety during early abstinence while improving adrenergic and stress system function, effects which are independent of a history of lifetime anxiety disorders.
Collapse
Affiliation(s)
- Verica Milivojevic
- From the, Department of Psychiatry, (VM, GH, RS), The Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut
| | - Gustavo A Angarita
- Clinical Neuroscience Research Unit, (GAA), Department of Psychiatry, Connecticut Mental Health Center, Yale University School of Medicine, New Haven, Connecticut
| | - Gretchen Hermes
- From the, Department of Psychiatry, (VM, GH, RS), The Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut
| | - Rajita Sinha
- From the, Department of Psychiatry, (VM, GH, RS), The Yale Stress Center, Yale University School of Medicine, New Haven, Connecticut
| | - Helen C Fox
- Department of Psychiatry, (HCF), Stony Brook University School of Medicine, Stony Brook, New York
| |
Collapse
|
32
|
María-Ríos CE, Morrow JD. Mechanisms of Shared Vulnerability to Post-traumatic Stress Disorder and Substance Use Disorders. Front Behav Neurosci 2020; 14:6. [PMID: 32082127 PMCID: PMC7006033 DOI: 10.3389/fnbeh.2020.00006] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Psychoactive substance use is a nearly universal human behavior, but a significant minority of people who use addictive substances will go on to develop an addictive disorder. Similarly, though ~90% of people experience traumatic events in their lifetime, only ~10% ever develop post-traumatic stress disorder (PTSD). Substance use disorders (SUD) and PTSD are highly comorbid, occurring in the same individual far more often than would be predicted by chance given the respective prevalence of each disorder. Some possible reasons that have been proposed for the relationship between PTSD and SUD are self-medication of anxiety with drugs or alcohol, increased exposure to traumatic events due to activities involved in acquiring illegal substances, or addictive substances altering the brain's stress response systems to make users more vulnerable to PTSD. Yet another possibility is that some people have an intrinsic vulnerability that predisposes them to both PTSD and SUD. In this review, we integrate clinical and animal data to explore these possible etiological links between SUD and PTSD, with an emphasis on interactions between dopaminergic, adrenocorticotropic, GABAergic, and glutamatergic neurobehavioral mechanisms that underlie different emotional learning styles.
Collapse
Affiliation(s)
| | - Jonathan D. Morrow
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
33
|
Patkar OL, Belmer A, Beecher K, Jacques A, Bartlett SE. Pindolol Rescues Anxiety-Like Behavior and Neurogenic Maladaptations of Long-Term Binge Alcohol Intake in Mice. Front Behav Neurosci 2019; 13:264. [PMID: 31849624 PMCID: PMC6895681 DOI: 10.3389/fnbeh.2019.00264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 11/20/2019] [Indexed: 12/30/2022] Open
Abstract
Long-term binge alcohol consumption alters the signaling of numerous neurotransmitters in the brain including noradrenaline (NE) and serotonin (5-HT). Alterations in the signaling of these neuronal pathways result in dysfunctional emotional states like anxiety and depression which are typically seen during alcohol withdrawal. Interestingly, studies have demonstrated that the development of alcohol-induced negative affective states is linked to disrupted neurogenesis in the dentate gyrus (DG) region of the hippocampus in alcohol-dependent animals. We have previously shown that modulation of NE and 5-HT activity by pharmacological targeting of β-adrenoreceptors (β-ARs) and 5-HT1A/1B receptors with pindolol reduces consumption in long-term alcohol-consuming mice. Since these receptors are also involved in emotional homeostasis and hippocampal neurogenesis, we investigated the effects of pindolol administration on emotional and neurogenic deficits in mice consuming long-term alcohol (18 weeks). We report that acute administration of pindolol (32 mg/kg) reduces anxiety-like behavior in mice at 24 h withdrawal in the marble-burying test (MBT) and the elevated plus-maze (EPM). We also show that chronic (2 weeks) pindolol treatment (32 mg/kg/day) attenuates alcohol-induced impairments in the density of immature neurons (DCX+) but not newborn cells (BrdU+) in the hippocampal DG. Pindolol treatment also restores the normal proportion of newborn proliferating cells (BrdU+/Ki67+/DCX−), newborn proliferating immature neurons (BrdU+/Ki67+/DCX+) and newborn non-proliferating immature neurons (BrdU+/Ki67−/DCX+) following long-term alcohol intake. These results suggest that pindolol, through its unique pharmacology may rescue some but not all deficits of long-term alcohol abuse on the brain, adding further value to its properties as a strong pharmaceutical option for alcohol use disorders (AUDs).
Collapse
Affiliation(s)
- Omkar L Patkar
- Addiction and Obesity Laboratory, Department of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, QLD, Australia
| | - Arnauld Belmer
- Addiction and Obesity Laboratory, Department of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, QLD, Australia
| | - Kate Beecher
- Addiction and Obesity Laboratory, Department of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, QLD, Australia
| | - Angela Jacques
- Addiction and Obesity Laboratory, Department of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, QLD, Australia
| | - Selena E Bartlett
- Addiction and Obesity Laboratory, Department of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
34
|
Funk D, Coen K, Tamadon S, Lê AD. Effects of the Alpha-1 Antagonist Prazosin on KOR Agonist-Induced Reinstatement of Alcohol Seeking. Int J Neuropsychopharmacol 2019; 22:724-734. [PMID: 31556948 PMCID: PMC6872965 DOI: 10.1093/ijnp/pyz049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/20/2019] [Accepted: 09/16/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Stress is associated with relapse to alcohol seeking during abstinence, but the processes underlying this relationship are poorly understood. Noradrenaline is a key transmitter in stress responses and in stress-induced drug seeking. The alpha-1 adrenoceptor antagonist prazosin has been investigated as a treatment for alcoholism and for chronic stress disorders that are frequently comorbid with alcoholism. In rats, we previously showed that prazosin blocks reinstatement of alcohol seeking induced by footshock and yohimbine stressors and reduces yohimbine-induced brain activation. The role of alpha-1 adrenoceptors in reinstatement induced by other stressors is not known. Our most recent work is on the role of kappa opioid receptors in stress-induced reinstatement of alcohol seeking and have reported that the selective kappa opioid receptor agonist U50,488 induces reinstatement and neuronal activation in stress- and relapse-related brain regions. Here we determine the involvement of alpha-1 receptors in reinstatement and brain activation induced by U50,488. METHODS We trained male Long-Evans rats to self-administer alcohol (12% w/v), extinguished alcohol-reinforced responding, and then determined the effects of prazosin (1 mg/kg) on U50,488 (2.5 mg/kg)-induced reinstatement and regional Fos expression. RESULTS Prazosin blocked U50,488-induced reinstatement and decreased U50,488-induced Fos expression in the orbitofrontal cortex, nucleus accumbens core, ventral bed nucleus of the stria terminalis, central and basolateral amygdalar nuclei and ventral tegmental area. CONCLUSIONS These findings suggest that prazosin may reduce U50,488-induced relapse by inhibiting activity in 1 or more of these brain areas.
Collapse
Affiliation(s)
- Douglas Funk
- Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada,Correspondence: Douglas Funk; Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Russell St., Toronto, Ontario, Canada M5S 2S1 ()
| | - Kathleen Coen
- Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Sahar Tamadon
- Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - A D Lê
- Neurobiology of Alcohol Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada,Department of Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|
35
|
Patkar OL, Belmer A, Holgate JY, Klenowski PM, Bartlett SE. Modulation of serotonin and noradrenaline in the BLA by pindolol reduces long-term ethanol intake. Addict Biol 2019; 24:652-663. [PMID: 30022582 DOI: 10.1111/adb.12630] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 01/05/2023]
Abstract
Repeated cycles of binge-like alcohol consumption and abstinence change the activity of several neurotransmitter systems. Some of these changes are consolidated following prolonged alcohol use and are thought to play an important role in the development of dependence. We have previously shown that systemic administration of the dual beta-adrenergic antagonist and 5-HT1A/1B partial agonist pindolol selectively reduces long-term but not short-term binge-like consumption of ethanol and alters excitatory postsynaptic currents in basolateral amygdala (BLA) principal neurons. The aim of this study was to investigate the effects of pindolol microinfusions in the BLA on long-term ethanol intake using the drinking-in-the-dark paradigm in mice. We also microinfused RU24969 (5-HT1A/1B receptor partial agonist) and CGP12177 (β1/2 adrenergic antagonist) following long-term ethanol intake and determined the densities of 5-HT1A/1B receptors and β1/2 adrenergic in the BLA following short-term (4 weeks) and long-term ethanol (12 weeks) consumption. We show that intra-BLA infusion of pindolol (1000 pmol/0.5 μl), RU24969 (0.3 and 3 pmol/0.5 μl) and CGP12177 (500 pmol/0.5 μl) produce robust decreases in long-term ethanol consumption. Additionally, we identified reduced β1/2 adrenergic receptor expression and no change in 5-HT1A/1B receptor density in the BLA of long-term ethanol-consuming mice. Collectively, our data highlight the effects of pindolol on voluntary, binge-like ethanol consumption behavior following long-term intake.
Collapse
Affiliation(s)
- Omkar L. Patkar
- Translational Research InstituteQueensland University of Technology Brisbane Australia
- Institute of Health and Biomedical Innovation (IHBI)Queensland University of Technology Brisbane Australia
| | - Arnauld Belmer
- Translational Research InstituteQueensland University of Technology Brisbane Australia
- Institute of Health and Biomedical Innovation (IHBI)Queensland University of Technology Brisbane Australia
| | - Joan Y. Holgate
- Translational Research InstituteQueensland University of Technology Brisbane Australia
- Institute of Health and Biomedical Innovation (IHBI)Queensland University of Technology Brisbane Australia
| | - Paul M. Klenowski
- Department of NeurobiologyUniversity of Massachusetts Medical School Worcester MA 01605 USA
| | - Selena E. Bartlett
- Translational Research InstituteQueensland University of Technology Brisbane Australia
- Institute of Health and Biomedical Innovation (IHBI)Queensland University of Technology Brisbane Australia
| |
Collapse
|
36
|
Murnane KS, Guner OF, Bowen JP, Rambacher KM, Moniri NH, Murphy TJ, Daphney CM, Oppong-Damoah A, Rice KC. The adrenergic receptor antagonist carvedilol interacts with serotonin 2A receptors both in vitro and in vivo. Pharmacol Biochem Behav 2019; 181:37-45. [PMID: 30998954 DOI: 10.1016/j.pbb.2019.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/08/2019] [Accepted: 04/12/2019] [Indexed: 11/19/2022]
Abstract
There is increasing support for the potential clinical use of compounds that interact with serotonin 2A (5-HT2A) receptors. It is therefore of interest to discover novel compounds that interact with 5-HT2A receptors. In the present study, we used computational chemistry to identify critical ligand structural features of 5-HT2A receptor binding and function. Query of compound databases using those ligand features revealed the adrenergic receptor antagonist carvedilol as a high priority match. As carvedilol is used clinically for cardiovascular diseases, we conducted experiments to assess whether it has any interactions with 5-HT2A receptors. In vitro experiments demonstrated that carvedilol has high nanomolar affinity for 5-HT2A receptors. In vivo experiments demonstrated that carvedilol increases the ethanol-induced loss of the righting reflex and suppresses operant responding in mice, and that these effects are attenuated by pretreatment with the selective 5-HT2A receptor antagonist M100907. Moreover, carvedilol did not induce the head-twitch response in mice, suggesting a lack of psychedelic effects. However, carvedilol did not activate canonical 5-HT2A receptor signaling pathways and antagonized serotonin-mediated signaling. It also reduced the head-twitch response induced by 2,5-Dimethoxy-4-iodoamphetamine, suggesting potential in vivo antagonism, allosteric modulation, or functional bias. These data suggest that carvedilol has functionally relevant interactions with 5-HT2A receptors, providing a novel mechanism of action for a clinically used compound. However, our findings do not clearly delineate the precise mechanism of action of carvedilol at 5-HT2A receptors, and additional experiments are needed to elucidate the role of 5-HT2A receptors in the behavioral and clinical effects of carvedilol.
Collapse
Affiliation(s)
- Kevin Sean Murnane
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA.
| | - Osman F Guner
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - J Phillip Bowen
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Kalyn M Rambacher
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Tyler J Murphy
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA; Department of Biology, Oglethorpe University, Atlanta, GA, USA
| | - Cedrick Maceo Daphney
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Aboagyewaah Oppong-Damoah
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Kenner C Rice
- Section on Drug Design and Synthesis, National Institute on Drug Abuse and Chemical Biology Research Branch, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Abstract
In this review, the effects of stress on alcohol drinking are discussed. The interactions between biological stress systems and alcohol drinking are examined, with a focus on the hypothalamic pituitary adrenal axis, corticotropin releasing factor, dynorphin, neuropeptide Y, and norepinephrine systems. Findings from animal models suggest that these biological stress systems may be useful targets for medications development for alcohol use disorder and co-occurring stress-related disorders in humans.
Collapse
Affiliation(s)
- Marcus M Weera
- Marcus M. Weera, Ph.D., is a postdoctoral fellow in the Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana. Nicholas W. Gilpin, Ph.D., is a professor in the Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Nicholas W Gilpin
- Marcus M. Weera, Ph.D., is a postdoctoral fellow in the Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana. Nicholas W. Gilpin, Ph.D., is a professor in the Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
38
|
Simpson TL, Saxon AJ, Stappenbeck C, Malte CA, Lyons R, Tell D, Millard SP, Raskind M. Double-Blind Randomized Clinical Trial of Prazosin for Alcohol Use Disorder. Am J Psychiatry 2018; 175:1216-1224. [PMID: 30153753 PMCID: PMC6395537 DOI: 10.1176/appi.ajp.2018.17080913] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Current medications for alcohol use disorder do not target brain noradrenergic pathways. Theoretical and preclinical evidence suggests that noradrenergic circuits may be involved in alcohol reinforcement and relapse. After a positive pilot study, the authors tested the α-1 adrenergic receptor antagonist prazosin to treat alcohol use disorder in a larger sample. METHOD Ninety-two participants with alcohol use disorder but without posttraumatic stress disorder were randomly assigned to receive prazosin or placebo in a 12-week double-blind study. Medication was titrated to a target dosing schedule of 4 mg in the morning, 4 mg in the afternoon, and 8 mg at bedtime by the end of week 2. The behavioral platform was medical management. Participants provided daily data on alcohol consumption. Generalized linear mixed-effects models were used to examine the impact of prazosin compared with placebo on number of drinks per week, number of drinking days per week, and number of heavy drinking days per week. RESULTS Eighty participants completed the titration period and were included in the primary analyses. There was a significant interaction between condition and week for both number of drinks and number of heavy drinking days, such that the rate of drinking and the probability of heavy drinking showed a greater decrease over time for participants in the prazosin condition compared with those in the placebo condition. Participants in the prazosin condition were more likely to report drowsiness and edema than participants in the placebo condition. CONCLUSIONS Prazosin holds promise as a harm-reduction pharmacologic treatment for alcohol use disorder and deserves further evaluation by independent research groups.
Collapse
Affiliation(s)
- Tracy L. Simpson
- Center of Excellence in Substance Abuse Treatment and Education, VA Puget Sound Health Care System, Seattle WA, Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| | - Andrew J. Saxon
- Center of Excellence in Substance Abuse Treatment and Education, VA Puget Sound Health Care System, Seattle WA, Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| | - Cynthia Stappenbeck
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| | - Carol A. Malte
- Center of Excellence in Substance Abuse Treatment and Education, VA Puget Sound Health Care System, Seattle WA
| | - Robert Lyons
- Seattle Institute for Biomedical and Clinical Research, Seattle, WA
| | - Dana Tell
- Seattle Institute for Biomedical and Clinical Research, Seattle, WA
| | - Steven P. Millard
- Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, Seattle Institute for Biomedical and Clinical Research, Seattle, WA
| | - Murray Raskind
- Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| |
Collapse
|
39
|
Clark SL, Costin BN, Chan RF, Johnson AW, Xie L, Jurmain JL, Kumar G, Shabalin AA, Pandey AK, Aberg KA, Miles MF, van den Oord E. A Whole Methylome Study of Ethanol Exposure in Brain and Blood: An Exploration of the Utility of Peripheral Blood as Proxy Tissue for Brain in Alcohol Methylation Studies. Alcohol Clin Exp Res 2018; 42:2360-2368. [PMID: 30320886 DOI: 10.1111/acer.13905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/06/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Recent reviews have highlighted the potential use of blood-based methylation biomarkers as diagnostic and prognostic tools of current and future alcohol use and addiction. Due to the substantial overlap that often exists between methylation patterns across different tissues, including blood and brain, blood-based methylation may track methylation changes in brain; however, little work has explored the overlap in alcohol-related methylation in these tissues. METHODS To study the effects of alcohol on the brain methylome and identify possible biomarkers of these changes in blood, we performed a methylome-wide association study in brain and blood from 40 male DBA/2J mice that received either an acute ethanol (EtOH) or saline intraperitoneal injection. To investigate all 22 million CpGs in the mouse genome, we enriched for the methylated genomic fraction using methyl-CpG binding domain (MBD) protein capture followed by next-generation sequencing (MBD-seq). We performed association tests in blood and brain separately followed by enrichment testing to determine whether there was overlapping alcohol-related methylation in the 2 tissues. RESULTS The top result for brain was a CpG located in an intron of Ttc39b (p = 5.65 × 10-08 ), and for blood, the top result was located in Espnl (p = 5.11 × 10-08 ). Analyses implicated pathways involved in inflammation and neuronal differentiation, such as CXCR4, IL-7, and Wnt signaling. Enrichment tests indicated significant overlap among the top results in brain and blood. Pathway analyses of the overlapping genes converge on MAPKinase signaling (p = 5.6 × 10-05 ) which plays a central role in acute and chronic responses to alcohol and glutamate receptor pathways, which can regulate neuroplastic changes underlying addictive behavior. CONCLUSIONS Overall, we have shown some methylation changes in brain and blood after acute EtOH administration and that the changes in blood partly mirror the changes in brain suggesting the potential for DNA methylation in blood to be biomarkers of alcohol use.
Collapse
Affiliation(s)
- Shaunna L Clark
- Department of Psychology , Michigan State University, East Lansing, Michigan.,Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Blair N Costin
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia
| | - Robin F Chan
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Alexander W Johnson
- Department of Psychology , Michigan State University, East Lansing, Michigan
| | - Linying Xie
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Jessica L Jurmain
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia
| | - Gaurav Kumar
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Andrey A Shabalin
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Ashutosh K Pandey
- Department of Anatomy and Neurobiology , Center for Integrative and Translational Genomics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Karolina A Aberg
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| | - Michael F Miles
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia
| | - Edwin van den Oord
- Center for Biomarker Research and Precision Medicine , Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
40
|
CRF modulation of central monoaminergic function: Implications for sex differences in alcohol drinking and anxiety. Alcohol 2018; 72:33-47. [PMID: 30217435 DOI: 10.1016/j.alcohol.2018.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/03/2018] [Accepted: 01/19/2018] [Indexed: 01/06/2023]
Abstract
Decades of research have described the importance of corticotropin-releasing factor (CRF) signaling in alcohol addiction, as well as in commonly co-expressed neuropsychiatric diseases, including anxiety and mood disorders. However, CRF signaling can also acutely regulate binge alcohol consumption, anxiety, and affect in non-dependent animals, possibly via modulation of central monoaminergic signaling. We hypothesize that basal CRF tone is particularly high in animals and humans with an inherent propensity for high anxiety and alcohol consumption, and thus these individuals are at increased risk for the development of alcohol use disorder and comorbid neuropsychiatric diseases. The current review focuses on extrahypothalamic CRF circuits, particularly those stemming from the bed nucleus of the stria terminalis (BNST), found to play a role in basal phenotypes, and examines whether the intrinsic hyperactivity of these circuits is sufficient to escalate the expression of these behaviors and steepen the trajectory of development of disease states. We focus our efforts on describing CRF modulation of biogenic amine neuron populations that have widespread projections to the forebrain to modulate behaviors, including alcohol and drug intake, stress reactivity, and anxiety. Further, we review the known sex differences and estradiol modulation of these neuron populations and CRF signaling at their synapses to address the question of whether females are more susceptible to the development of comorbid addiction and stress-related neuropsychiatric diseases because of hyperactive extrahypothalamic CRF circuits compared to males.
Collapse
|
41
|
Haass-Koffler CL, Swift RM, Leggio L. Noradrenergic targets for the treatment of alcohol use disorder. Psychopharmacology (Berl) 2018; 235:1625-1634. [PMID: 29460163 PMCID: PMC5995154 DOI: 10.1007/s00213-018-4843-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/28/2018] [Indexed: 12/17/2022]
Abstract
The role of norepinephrine (NE) in the development of alcohol use disorder (AUD) has been studied over the past several decades. However, the NE system has been largely ignored for many years as a potential target for medication development for AUD. More recently, preclinical and clinical studies have demonstrated the potential value of targeting NE signaling for developing new pharmacological treatments for AUD. This review contributes to a special issue of Psychopharmacology focused on promising targets for alcohol addiction. Specifically, this review coalesces preclinical and clinical neuroscience that re-evaluate the noradrenergic system, and in particular the alpha-1 receptor, as a potential target for AUD.
Collapse
Affiliation(s)
- Carolina L Haass-Koffler
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, 02912, USA.
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA.
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
| | - Robert M Swift
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, 02912, USA
- Providence VA Medical Center, Providence, RI, 02912, USA
| | - Lorenzo Leggio
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
42
|
Abstract
Patients who suffer from alcohol use disorders (AUDs) usually go through various socio-behavioral and pathophysiological changes that take place in the brain and other organs. Recently, consumption of unhealthy food and excess alcohol along with a sedentary lifestyle has become a norm in both developed and developing countries. Despite the beneficial effects of moderate alcohol consumption, chronic and/or excessive alcohol intake is reported to negatively affect the brain, liver and other organs, resulting in cell death, organ damage/failure and death. The most effective therapy for alcoholism and alcohol related comorbidities is alcohol abstinence, however, chronic alcoholic patients cannot stop drinking alcohol. Therefore, targeted therapies are urgently needed to treat such populations. Patients who suffer from alcoholism and/or alcohol abuse experience harmful effects and changes that occur in the brain and other organs. Upon stopping alcohol consumption, alcoholic patients experience acute withdrawal symptoms followed by a protracted abstinence syndrome resulting in the risk of relapse to heavy drinking. For the past few decades, several drugs have been available for the treatment of AUDs. These drugs include medications to reduce or stop severe alcohol withdrawal symptoms during alcohol detoxification as well as recovery medications to reduce alcohol craving and support abstinence. However, there is no drug that completely antagonizes the adverse effects of excessive amounts of alcohol. This review summarizes the drugs which are available and approved by the FDA and their mechanisms of action as well as the medications that are under various phases of preclinical and clinical trials. In addition, the repurposing of the FDA approved drugs, such as anticonvulsants, antipsychotics, antidepressants and other medications, to prevent alcoholism and treat AUDs and their potential target mechanisms are summarized.
Collapse
Affiliation(s)
- Mohammed Akbar
- Division of Neuroscience and Behavior, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA.
| | - Mark Egli
- Division of Neuroscience and Behavior, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - Young-Eun Cho
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - Antonio Noronha
- Division of Neuroscience and Behavior, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
43
|
Di Ciano P, Le Foll B. The Rat Gambling Task as a model for the preclinical development of treatments for gambling disorder. INTERNATIONAL GAMBLING STUDIES 2018. [DOI: 10.1080/14459795.2018.1448428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Patricia Di Ciano
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health (CAMH) , Toronto, Canada
| | - Bernard Le Foll
- Addiction Division, Centre for Addiction and Mental Health (CAMH) , Toronto, Canada
- Departments of Pharmacology and Toxicology, Psychiatry, Family and Community Medicine, Institute of Medical Sciences, University of Toronto , Toronto, Canada
| |
Collapse
|
44
|
Vazey EM, den Hartog CR, Moorman DE. Central Noradrenergic Interactions with Alcohol and Regulation of Alcohol-Related Behaviors. Handb Exp Pharmacol 2018; 248:239-260. [PMID: 29687164 DOI: 10.1007/164_2018_108] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder (AUD) results from disruption of a number of neural systems underlying motivation, emotion, and cognition. Patients with AUD exhibit not only elevated motivation for alcohol but heightened stress and anxiety, and disruptions in cognitive domains such as decision-making. One system at the intersection of these functions is the central norepinephrine (NE) system. This catecholaminergic neuromodulator, produced by several brainstem nuclei, plays profound roles in a wide range of behaviors and functions, including arousal, attention, and other aspects of cognition, motivation, emotional regulation, and control over basic physiological processes. It has been known for some time that NE has an impact on alcohol seeking and use, but the mechanisms of its influence are still being revealed. This chapter will discuss the influence of NE neuron activation and NE release at alcohol-relevant targets on behaviors and disruptions underlying alcohol motivation and AUD. Potential NE-based pharmacotherapies for AUD treatment will also be discussed. Given the basic properties of NE function, the strong relationship between NE and alcohol use, and the effectiveness of current NE-related treatments, the studies presented here indicate an encouraging direction for the development of precise and efficacious future therapies for AUD.
Collapse
Affiliation(s)
- Elena M Vazey
- Department of Biology & Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA.
| | - Carolina R den Hartog
- Department of Biology & Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - David E Moorman
- Department of Psychological and Brain Sciences & Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| |
Collapse
|
45
|
Effects of disulfiram on choice behavior in a rodent gambling task: association with catecholamine levels. Psychopharmacology (Berl) 2018; 235:23-35. [PMID: 29085979 PMCID: PMC5750121 DOI: 10.1007/s00213-017-4744-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/08/2017] [Indexed: 10/18/2022]
Abstract
RATIONALE Gambling disorder is a growing societal concern, as recognized by its recent classification as an addictive disorder in the DSM-5. Case reports have shown that disulfiram reduces gambling-related behavior in humans. OBJECTIVES The purpose of the present study was to determine whether disulfiram affects performance on a rat gambling task, a rodent version of the Iowa gambling task in humans, and whether any changes were associated with alterations in dopamine and/or norepinephrine levels. METHODS Rats were administered disulfiram prior to testing on the rat gambling task or prior to analysis of dopamine or norepinephrine levels in brain homogenates. Rats in the behavioral task were divided into two subgroups (optimal vs suboptimal) based on their baseline levels of performance in the rat gambling task. Rats in the optimal group chose the advantageous strategy more, and rats in the suboptimal group (a parallel to problem gambling) chose the disadvantageous strategy more. Rats were not divided into optimal or suboptimal groups prior to neurochemical analysis. RESULTS Disulfiram administered 2 h, but not 30 min, before the task dose-dependently improved choice behavior in the rats with an initial disadvantageous "gambling-like" strategy, while having no effect on the rats employing an advantageous strategy. The behavioral effects of disulfiram were associated with increased striatal dopamine and decreased striatal norepinephrine. CONCLUSIONS These findings suggest that combined actions on dopamine and norepinephrine may be a useful treatment for gambling disorders.
Collapse
|
46
|
Barajaz AM, Kliethermes CL. An assessment of the utilization of the preclinical rodent model literature in clinical trials of putative therapeutics for the treatment of alcohol use disorders. Drug Alcohol Depend 2017; 181:77-84. [PMID: 29035708 DOI: 10.1016/j.drugalcdep.2017.09.022] [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: 05/26/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Rodent models of Alcohol Use Disorders (AUD) are used extensively by preclinical researchers to develop new therapeutics for the treatment of AUD. Although these models play an important role in the development of novel, targeted therapeutics, their role in bringing therapeutics to clinical trials is unclear, as off-label use of existing medications not approved for the treatment of AUD is commonly seen in the clinic and clinical trials. METHOD In the current study, we used the Clinicaltrials.gov database to obtain a list of drugs that have been tested for efficacy in a clinical trial between 1997 and 2017. We then conducted a set of literature searches to determine which of the 98 unique drugs we identified had shown efficacy in a rodent model of an AUD prior to being tested in a clinical trial. RESULTS We found that slightly less than half of the drugs tested in clinical trials (48%) had shown prior efficacy in any rodent model of an AUD, while the remaining 52% of drugs were used off-label, or in some cases, following non-published studies. CONCLUSION This study raises the question of how clinical researchers incorporate results from preclinical studies in the decision to bring a drug to a clinical trial. Our results underscore the need for ongoing communication among preclinical and clinical researchers.
Collapse
Affiliation(s)
- Ashley M Barajaz
- Drake University, Department of Psychology and Neuroscience, 1344 27th Street, Des Moines, IA 50311, United States
| | - Christopher L Kliethermes
- Drake University, Department of Psychology and Neuroscience, 1344 27th Street, Des Moines, IA 50311, United States.
| |
Collapse
|
47
|
Kimbrough A, de Guglielmo G, Kononoff J, Kallupi M, Zorrilla EP, George O. CRF 1 Receptor-Dependent Increases in Irritability-Like Behavior During Abstinence from Chronic Intermittent Ethanol Vapor Exposure. Alcohol Clin Exp Res 2017; 41:1886-1895. [PMID: 28833238 DOI: 10.1111/acer.13484] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/17/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND In humans, emotional and physical signs of withdrawal from ethanol are commonly seen. Many of these symptoms, including anxiety-like and depression-like behavior, have been characterized in animal models of ethanol dependence. One issue with several current behavioral tests that measure withdrawal in animal models is that they are often not repeatable within subjects over time. Additionally, irritability, one of the most common symptoms of ethanol withdrawal in humans, has not been well characterized in animal models. The corticotropin-releasing factor (CRF)-CRF1 receptor system has been suggested to be critical for the emergence of anxiety-like behavior in ethanol dependence, but the role of this system in irritability-like behavior has not been characterized. METHODS The present study compared the effects of chronic intermittent ethanol (CIE) vapor exposure-induced ethanol dependence on irritability-like behavior in rats using the bottle-brush test during acute withdrawal and protracted abstinence. Rats were trained to self-administer ethanol in operant chambers and then either left in a nondependent state or made dependent via CIE. Naïve, nondependent, and dependent rats were tested for irritability-like behavior in the bottle-brush test 8 hours and 2 weeks into abstinence from ethanol. Separate cohorts of dependent and nondependent rats were used to examine the effect of the specific CRF1 receptor antagonist R121919 on irritability-like behavior. RESULTS Dependent rats exhibited escalated ethanol intake compared with their own pre-CIE baseline and nondependent rats. At both time points of abstinence, ethanol-dependent rats exhibited increased aggressive-like responses compared with naïve and nondependent rats. R121919 reduced irritability-like behavior in both dependent and nondependent rats, but dependent rats were more sensitive to R121919. CONCLUSIONS Irritability-like behavior is a clinically relevant and reliable measure of negative emotional states that is partially mediated by activation of the CRF-CRF1 system and remains elevated during protracted abstinence in ethanol-dependent rats.
Collapse
Affiliation(s)
- Adam Kimbrough
- Department of Neuroscience (AK, GdG, JK, MK, EPZ, OG), The Scripps Research Institute, La Jolla, California
| | - Giordano de Guglielmo
- Department of Neuroscience (AK, GdG, JK, MK, EPZ, OG), The Scripps Research Institute, La Jolla, California
| | - Jenni Kononoff
- Department of Neuroscience (AK, GdG, JK, MK, EPZ, OG), The Scripps Research Institute, La Jolla, California
| | - Marsida Kallupi
- Department of Neuroscience (AK, GdG, JK, MK, EPZ, OG), The Scripps Research Institute, La Jolla, California
| | - Eric P Zorrilla
- Department of Neuroscience (AK, GdG, JK, MK, EPZ, OG), The Scripps Research Institute, La Jolla, California
| | - Olivier George
- Department of Neuroscience (AK, GdG, JK, MK, EPZ, OG), The Scripps Research Institute, La Jolla, California
| |
Collapse
|
48
|
Haass-Koffler CL, Goodyear K, Zywiak WH, Magill M, Eltinge SE, Wallace PM, Long VM, Jayaram-Lindström N, Swift RM, Kenna GA, Leggio L. Higher pretreatment blood pressure is associated with greater alcohol drinking reduction in alcohol-dependent individuals treated with doxazosin. Drug Alcohol Depend 2017; 177:23-28. [PMID: 28551590 PMCID: PMC5534374 DOI: 10.1016/j.drugalcdep.2017.03.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND Preclinical and clinical research suggest that the α1 receptor antagonist prazosin reduces alcohol consumption. Furthermore, clinical studies indicate a role for prazosin in treating Post-Traumatic Stress Disorder (PTSD) symptoms and a recent trial suggested that pre-treatment blood pressure (BP) predicts therapeutic response for prazosin in PTSD patients. Whether pre-treatment BP may predict response to α1 blockers in alcohol-dependent (AD) patients is unknown. We previously reported a randomized controlled trial (RCT) where doxazosin, an α1 receptor antagonist with a more favorable pharmacokinetic profile than prazosin, reduced drinks per week (DPW) and heavy drinking days (HDD) in AD patients with a high family history density of alcoholism. In this study, we tested pre-treatment BP as another potentially valuable clinical moderator of doxazosin's response on alcohol consumption. METHODS This was a double-blind placebo-controlled RCT testing doxazosin up to 16mg/day in AD treatment-seeking patients (N=41). The hypothesized moderator effect of baseline standing systolic and diastolic BP on DPW and HDD was tested. RESULTS With pre-treatment standing diastolic BP as a moderator, there were significant BP x medication interactions for both DPW [**p=0.009, d=0.80] and HDD [*p=0.018, d=1.11]. Post-hoc analyses indicated significant doxazosin effects in patients with higher standing BP in reducing both DPW and HDD. CONCLUSION These findings suggest that higher standing diastolic BP at baseline (pre-treatment) may represent a predictor of doxazosin's response on alcohol consumption in AD patients. These results further elucidate the possible efficacy and mechanisms of action of α1 receptor antagonism in AD individuals.
Collapse
Affiliation(s)
- Carolina L. Haass-Koffler
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD,Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI,Correspondence: Carolina L. Haass-Koffler, Pharm.D., Center for Alcohol and Addiction Studies, Department Psychiatry and Human Behavior, Brown University, 121 South Main Street, Providence, RI 02912, Phone: 401-863-6630, , Lorenzo Leggio, M.D., Ph.D., M.Sc., Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, NIAAA DICBR and NIDA IRP, National Institutes of Health, 10 Center Drive (10CRC/15330) Room 1-5429, Bethesda, MD 20892-1108, Phone: 301-435-9398,
| | - Kimberly Goodyear
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI
| | - William H. Zywiak
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI
| | - Molly Magill
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI
| | - Sarah E. Eltinge
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI
| | | | - Victoria M. Long
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI
| | - Nitya Jayaram-Lindström
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Robert M. Swift
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI,Veterans Affairs Medical Center, Providence, RI
| | - George A. Kenna
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, United States; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, United States.
| |
Collapse
|
49
|
Becker HC. Influence of stress associated with chronic alcohol exposure on drinking. Neuropharmacology 2017; 122:115-126. [PMID: 28431971 PMCID: PMC5497303 DOI: 10.1016/j.neuropharm.2017.04.028] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/12/2017] [Accepted: 04/17/2017] [Indexed: 12/24/2022]
Abstract
Stress is commonly regarded as an important trigger for relapse and a significant factor that promotes increased motivation to drink in some individuals. However, the relationship between stress and alcohol is complex, likely changing in form during the transition from early moderated alcohol use to more heavy uncontrolled alcohol intake. A growing body of evidence indicates that prolonged excessive alcohol consumption serves as a potent stressor, producing persistent dysregulation of brain reward and stress systems beyond normal homeostatic limits. This progressive dysfunctional (allostatic) state is characterized by changes in neuroendocrine and brain stress pathways that underlie expression of withdrawal symptoms that reflect a negative affective state (dysphoria, anxiety), as well as increased motivation to self-administer alcohol. This review highlights literature supportive of this theoretical framework for alcohol addiction. In particular, evidence for stress-related neural, physiological, and behavioral changes associated with chronic alcohol exposure and withdrawal experience is presented. Additionally, this review focuses on the effects of chronic alcohol-induced changes in several pro-stress neuropeptides (corticotropin-releasing factor, dynorphin) and anti-stress neuropeptide systems (nocicepton, neuropeptide Y, oxytocin) in contributing to the stress, negative emotional, and motivational consequences of chronic alcohol exposure. Studies involving use of animal models have significantly increased our understanding of the dynamic stress-related physiological mechanisms and psychological underpinnings of alcohol addiction. This, in turn, is crucial for developing new and more effective therapeutics for treating excessive, harmful drinking, particularly stress-enhanced alcohol consumption. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Department of Neuroscience, Medical University of South Carolina, RHJ Department of Veterans Affairs, Charleston, SC 29464, USA.
| |
Collapse
|
50
|
Mason BJ. Emerging pharmacotherapies for alcohol use disorder. Neuropharmacology 2017; 122:244-253. [PMID: 28454983 PMCID: PMC5643030 DOI: 10.1016/j.neuropharm.2017.04.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 01/29/2023]
Abstract
The identification of different stages within the alcohol use disorder (AUD) cycle that are linked to neurocircuitry changes in pathophysiology associated with the negative emotional states of abstinence has provided a view of medication development for AUD that emphasizes changes in the brain reward and stress systems. Alcohol use disorder can be defined as a chronic relapsing disorder that involves compulsive alcohol seeking and taking, loss of control over alcohol intake, and emergence of a negative emotional state during abstinence. The focus of early medications development was to block the motivation to seek alcohol in the binge/intoxication stage. More recent work has focused on reversing the motivational dysregulations associated with the withdrawal/negative affect and preoccupation/anticipation stages during protracted abstinence. Advances in our understanding of the neurocircuitry and neuropharmacological mechanisms that are involved in the development and maintenance of the withdrawal/negative affect stage using validated animal models have provided viable targets for future medications. Another major advance has been proof-of-concept testing of potential therapeutics and clinical validation of relevant pharmacological targets using human laboratory models of protracted abstinence. This review focuses on future targets for medication development associated with reversal of the loss of reward function and gain in brain stress function that drive negative reinforcement in the withdrawal/negative affect stage of addiction. Basic research has identified novel neurobiological targets associated with the withdrawal/negative affect stage and preoccupation/anticipation stage, with a focus on neuroadaptive changes within the extended amygdala that account for the transition to dependence and vulnerability to relapse. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Barbara J Mason
- The Pearson Center on Alcoholism and Addiction Research, Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, TPC-5 La Jolla, CA 92037 USA.
| |
Collapse
|