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
|
Varodayan FP, Erikson CM, Scroger MV, Roberto M. Noradrenergic Mechanisms and Circuitry of Hyperkatifeia in Alcohol Use Disorder. Biol Psychiatry 2025; 97:580-589. [PMID: 39304172 PMCID: PMC11839382 DOI: 10.1016/j.biopsych.2024.09.009] [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: 03/15/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Hyperkatifeia, the manifestation of emotional distress or pain, is a conceptual framework gaining traction throughout the alcohol and other substance use fields as an important driver of addiction. It is well known that previous or current negative life experiences can serve as powerful motivators for excessive alcohol consumption and precipitate the development of an alcohol use disorder (AUD). A major hallmark of later stages of AUD is the emergence of hyperkatifeia during withdrawal, which can persist well into protracted abstinence to drive relapse. Given these complex interactions, understanding the specific neuroadaptations that lie at the intersection of hyperkatifeia and AUD can inform ongoing therapeutic development. The monoamine norepinephrine is of particular interest. Noradrenergic dysfunction is implicated in AUD, anxiety, chronic stress, depression, and emotional and physical pain. Importantly, there are key sexual dimorphisms within the noradrenergic system that are thought to differentially impact the development and trajectory of AUD in women and men. In the current review, we discuss past and recent work on noradrenergic influences at each stage of the AUD cycle (binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation) through the lens of hyperkatifeia. Evidence from these studies support the prioritization of norepinephrine-specific drug development to treat AUD and the identification of AUD subpopulations that may benefit the most from these therapies (e.g., women or people with comorbid chronic pain or anxiety/stress disorders).
Collapse
Affiliation(s)
- Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University, SUNY, Binghamton, New York.
| | - Chloe M Erikson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Marcis V Scroger
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University, SUNY, Binghamton, New York
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California.
| |
Collapse
|
3
|
Ortelli OA, Weiner JL. Evaluating the impact of concurrent sucrose availability on operant ethanol self-administration in male and female Long Evans rats. ADDICTION NEUROSCIENCE 2025; 14:100196. [PMID: 40161352 PMCID: PMC11951412 DOI: 10.1016/j.addicn.2025.100196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Investigating how environmental factors, such as the availability of non-ethanol alternative reinforcers, influences ethanol self-administration is critical for understanding the pathology of alcohol use disorder (AUD). Here we established the first operant choice paradigm that leverages the strengths of the sipper tube self-administration model to investigate how concurrent access to sucrose altered ethanol self-administration in male and female Long Evans rats. Choice behavior was examined using two distinct paradigms, including a novel adaptation of the response requirement paradigm. Under both a fixed-ratio or response requirement paradigm, we observed that concurrent availability of an alternative reinforcer significantly reduced appetitive and consummatory ethanol drinking-related behaviors. Furthermore, we assessed the sensitivity of the response requirement choice paradigm by administering the pharmacological stressor yohimbine and by altering the taste of the ethanol solution. Yohimbine administration non-selectively increased ethanol and sucrose intake, but not seeking, while taste adulteration decreased ethanol seeking and intake. These experiments demonstrate the utility of two concurrent choice paradigms that can more accurately capture AUD-like phenotypes, such as ethanol-directed choice in the face of alternative reinforcers. Future studies should investigate how models of vulnerability and dependence alter ethanol choice behavior under these paradigms.
Collapse
Affiliation(s)
- Olivia A. Ortelli
- Wake Forest University School of Medicine, Department of Translational Neuroscience, United States
| | - Jeffrey L. Weiner
- Wake Forest University School of Medicine, Department of Translational Neuroscience, United States
| |
Collapse
|
4
|
Petrakis IL, Nolen T, Vandergrift N, Hirsch S, Krystal JH, De Vivo M, Sabados J, Pisani E, Newcomb J, Kosten TR. Dexmedetomidine HCL (BXCL501) as a potential treatment for alcohol use disorder and comorbid PTSD: A phase 1b, placebo-controlled crossover laboratory study. Am J Addict 2025; 34:7-14. [PMID: 39152094 DOI: 10.1111/ajad.13637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Noradrenergic dysregulation is important in the pathophysiology of posttraumatic stress disorder (PTSD) and alcohol use disorder (AUD); pharmacotherapies targeting adrenergic function have potential as treatment for comorbidity. Dexmedetomidine (sublingual film formulation-BXCL501; IGALMI) is a highly potent, selective ⍺2-adrenergic receptor agonist and may be superior to other pharmacotherapeutic approaches. A within subjects, phase 1b safety laboratory study was conducted to evaluate adverse effects of BXCL501 when combined with alcohol; BXCL501's potential efficacy was also explored. METHODS Heavy drinker participants with a diagnosis of or who were at risk for PTSD participated in three separate test days which included pretreatment with BXCL501 (40 µg, 80 µg or placebo) administered in a randomized, double-blind fashion, followed by three testing conditions: alcohol cue reactivity, trauma-induced reactivity, and IV ethanol administration. Safety outcomes included blood pressure (BP) and sedation. Exploratory outcomes included alcohol craving, trauma-induced anxiety and craving and subjective effects of alcohol. RESULTS Ten of twelve randomized participants competed the entire study. BXCL501 (80 µg) was associated with expected mild changes in BP and sedation; administration with alcohol did not affect those parameters. There were no clinically significant adverse effects. BXCL501 attenuated trauma-induced anxiety and attenuated subjective effects of alcohol. DISCUSSIONS AND CONCLUSIONS BXCL501 is safe for use in humans who may drink alcohol while undergoing treatment. BXCL501 may be explored as a potential treatment for PTSD and AUD. SCIENTIFIC SIGNIFICANCE This is the first study to provide scientific support for BXCL501's potential to treat PTSD and comorbid AUD.
Collapse
Affiliation(s)
- Ismene L Petrakis
- Department of Veterans Affairs, VA Connecticut Healthcare System, West Haven, Connecticut, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Tracy Nolen
- Social, Statistical, & Environmental Sciences, RTI International, Research Triangle Park, North Carolina, USA
| | - Nathan Vandergrift
- Social, Statistical, & Environmental Sciences, RTI International, Research Triangle Park, North Carolina, USA
| | - Shawn Hirsch
- Social, Statistical, & Environmental Sciences, RTI International, Research Triangle Park, North Carolina, USA
| | - John H Krystal
- Department of Veterans Affairs, VA Connecticut Healthcare System, West Haven, Connecticut, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Jeff Sabados
- BioXcel Therapeutics Inc., New Haven, Connecticut, USA
| | - Emily Pisani
- Department of Veterans Affairs, VA Connecticut Healthcare System, West Haven, Connecticut, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jenelle Newcomb
- Department of Veterans Affairs, VA Connecticut Healthcare System, West Haven, Connecticut, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
5
|
Gully BJ, Brown ZE, Hornbacher R, Brown JC, Back SE, McCance-Katz EF, Swift RM, Haass-Koffler CL. Oxytocin Reduces Noradrenergic-Induced Opioid-Like Withdrawal Symptoms in Individuals on Opioid Agonist Therapy. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100395. [PMID: 39534517 PMCID: PMC11555595 DOI: 10.1016/j.bpsgos.2024.100395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background Intranasal administration of the neuropeptide oxytocin has been explored as a potential therapeutic agent for substance use disorder including opioid use disorder (OUD). Methods This phase 1, crossover, randomized, double-blind, placebo-controlled trial tested the safety, tolerability, and efficacy of intranasal oxytocin (80 IU) twice a day for 7 days in participants (N = 20) with OUD who were taking an opioid agonist therapy. In the laboratory, participants underwent opioid cue exposure paired with noradrenergic activation produced by yohimbine (32.4 mg) or placebo. Assessments included, 1) subjective response: craving, withdrawal, anxiety, and stress; 2) biomedical markers: hypothalamic-pituitary-adrenal axis response (cortisol) and noradrenergic activation (α-amylase); and 3) safety measures: hemodynamics and adverse event evaluation. Generalized linear model with model-based estimator in the covariance matrix was used, with medication (oxytocin/placebo) and noradrenergic activation (yohimbine/placebo) as within-subject factors. Results Oxytocin significantly reduced opioid-like withdrawal, anxiety symptoms, and cortisol levels elicited by cue exposure under noradrenergic activation produced by yohimbine. This effect was specific because oxytocin did not reduce craving, hemodynamics, or α-amylase levels increased by yohimbine administration. A single dose of yohimbine elicited the noradrenergic stimulation, and 7-day oxytocin administration was safe and well tolerated among individuals diagnosed with OUD and taking opioid agonist therapy. Conclusions The findings of this study suggest that oxytocin alleviates opioid-like withdrawal symptoms and anxiety by modulating the hypothalamic-pituitary-adrenal axis.
Collapse
Affiliation(s)
- Brian J. Gully
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island
| | - Zoe E. Brown
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island
| | - Rivkah Hornbacher
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island
| | - Joshua C. Brown
- Transcranial Magnetic Stimulation (TMS) Center, McLean Hospital, Belmont, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Sudie E. Back
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Healthcare System, Charleston, South Carolina
| | - Elinore F. McCance-Katz
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Robert M. Swift
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island
- Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island
| | - Carolina L. Haass-Koffler
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island
- Carney Institute for Brain Science, Brown University, Providence Rhode Island
| |
Collapse
|
6
|
Dali G, Logge W, Kranzler HR, Hurzeler T, Gallagher H, Haber PS, Morley KC. Comparative effects of topiramate and naltrexone on neural activity during anticipatory anxiety in individuals with alcohol use disorder. Alcohol Alcohol 2024; 60:agae078. [PMID: 39558663 PMCID: PMC11573881 DOI: 10.1093/alcalc/agae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/01/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024] Open
Abstract
Topiramate has been found to be effective in reducing alcohol use and may also attenuate anxiety severity in patients with alcohol use disorder (AUD). This study compared the neural response of treatment-seeking patients with AUD on either topiramate or naltrexone during an anticipatory anxiety task. Participants were 42 patients with AUD who were randomized to receive either topiramate (n = 23; titrated dose up to 200 mg/day) or naltrexone (n = 19; 50 mg/day) for 12-weeks as part of a larger randomized controlled trial. Following 6 weeks of treatment, participants completed an anticipatory anxiety task during a functional magnetic resonance imaging (fMRI) session. The task presented a series of high-threat and low-threat stimuli followed by an unpleasant or pleasant image, respectively. Primary whole-brain analyses revealed no significant differences in neural activation between the topiramate and naltrexone groups. Deactivation for safe cues relative to threat cues was observed within the precuneus, inferior parietal lobule and the cingulate gyrus. In the precentral and middle frontal gyri, threat cues elicited greater activation. Exploratory analyses revealed an effect of change in anxiety from baseline to week 6, with a greater reduction associated with a reduced response to threat cues relative to safe cues in the cuneus and lingual gyrus. The current study is the first to examine and compare neural activation during anticipatory anxiety in treatment-seeking individuals on topiramate and naltrexone. This preliminary research contributes to our understanding of the therapeutic mechanisms of these alcohol pharmacotherapies.
Collapse
Affiliation(s)
- Gezelle Dali
- Specialty of Addiction Medicine, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
- Edith Collins Centre for Translational Research (Alcohol, Drugs & Toxicology), Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW 2050, Australia
| | - Warren Logge
- Specialty of Addiction Medicine, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
- Edith Collins Centre for Translational Research (Alcohol, Drugs & Toxicology), Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW 2050, Australia
| | - Henry R Kranzler
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine and Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, United States
| | - Tristan Hurzeler
- Specialty of Addiction Medicine, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
| | - Hugh Gallagher
- Specialty of Addiction Medicine, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
| | - Paul S Haber
- Specialty of Addiction Medicine, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
- Edith Collins Centre for Translational Research (Alcohol, Drugs & Toxicology), Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW 2050, Australia
| | - Kirsten C Morley
- Specialty of Addiction Medicine, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
- Edith Collins Centre for Translational Research (Alcohol, Drugs & Toxicology), Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW 2050, Australia
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Tonetto S, Weikop P, Thomsen M. Nutritional ketosis as treatment for alcohol withdrawal symptoms in female C57BL/6J mice. Sci Rep 2024; 14:5092. [PMID: 38429369 PMCID: PMC10907582 DOI: 10.1038/s41598-024-55310-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/22/2024] [Indexed: 03/03/2024] Open
Abstract
Upon both acute and prolonged alcohol intake, the brain undergoes a metabolic shift associated with increased acetate metabolism and reduced glucose metabolism, which persists during abstinence, putatively leading to energy depletion in the brain. This study evaluates the efficacy of ketogenic treatments to rescue psychiatric and neurochemical alterations during long-term alcohol withdrawal. Female mice were intermittently exposed to alcohol vapor or air for three weeks, during which mice were introduced to either a ketogenic diet (KD), control diet supplemented with ketone ester (KE) or remained on control diet (CD). Withdrawal symptoms were assessed over a period of four weeks followed by re-exposure using several behavioral and biochemical tests. Alcohol-exposed mice fed CD displayed long-lasting depressive-like symptoms measured by saccharin preference and tail suspension, as well as decreased norepinephrine levels and serotonin turnover in the hippocampus. Both KD and KE rescued anhedonia for up to three weeks of abstinence. KD mice showed higher latency to first immobility in the tail suspension test, as well as lower plasma cholesterol levels. Our findings show promising effects of nutritional ketosis in ameliorating alcohol withdrawal symptoms in mice. KD seemed to better rescue these symptoms compared to KE.
Collapse
Affiliation(s)
- Simone Tonetto
- Laboratory of Neuropsychiatry, Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Copenhagen, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Morgan Thomsen
- Laboratory of Neuropsychiatry, Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark.
- Copenhagen Center for Translational Research, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Copenhagen, Denmark.
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Laboratory of Neuropsychiatry, Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Hovedvejen 17, 1., 2000, Frederiksberg, Denmark.
| |
Collapse
|
10
|
Schacht JP. Stress, Cues, and Craving: Does the Insula Hold the Key to Understanding Stress-Induced Drinking in Alcohol Use Disorder? Biol Psychiatry 2024; 95:200-201. [PMID: 38143097 DOI: 10.1016/j.biopsych.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/26/2023]
Affiliation(s)
- Joseph P Schacht
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
11
|
Intra J, Ippolito S, Lorenzini F, Mauro A, Mazzitello MC, Melzi S, Cappellani A, Cappellini F, Casati M. Elevated urine norepinephrine levels and alcohol use: A relationship that should be not neglected. Clin Biochem 2024; 123:110704. [PMID: 38158170 DOI: 10.1016/j.clinbiochem.2023.110704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Affiliation(s)
- Jari Intra
- Clinical Chemistry Laboratory, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
| | - Silvia Ippolito
- Clinical Chemistry Laboratory, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Franco Lorenzini
- Clinical Chemistry Laboratory, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Antonio Mauro
- Clinical Chemistry Laboratory, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | | | - Stefano Melzi
- Clinical Chemistry Laboratory, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Adele Cappellani
- Clinical Chemistry Laboratory, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Fabrizio Cappellini
- Clinical Chemistry Laboratory, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Marco Casati
- Clinical Chemistry Laboratory, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
12
|
Schacht JP, Kubicki M, Anton RF. A randomized trial of the effects of COMT inhibition on subjective response to alcohol: Moderation by baseline COMT activity and mediation of alcohol self-administration. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:178-187. [PMID: 38206282 DOI: 10.1111/acer.15227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 11/07/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Poor inhibitory control and enhanced subjective response to alcohol are interrelated risk factors for alcohol use disorder (AUD) that share underlying neural substrates, including dopamine signaling in the right prefrontal cortex, a potential target for pharmacological intervention. Cortical dopamine inactivation is primarily regulated by catechol-O-methyltransferase (COMT), an enzyme with large variation in activity as a function of the COMT rs4680 (val158met) single nucleotide polymorphism. In a previous randomized, placebo-controlled trial of the COMT inhibitor tolcapone (200 mg TID) in non-treatment-seeking participants with AUD, we found that tolcapone, relative to placebo, reduced alcohol self-administration only among rs4680 val-allele homozygotes, whose COMT activity is higher than in met-allele carriers. METHODS We conducted secondary analyses of the effects of tolcapone and baseline COMT activity, as indexed by both rs4680 genotype and an enzymatic activity assay, on the subjective response to alcohol in a bar-laboratory paradigm among 60 participants in the previous trial. RESULTS Tolcapone did not affect alcohol-induced stimulation or sedation more than placebo. However, baseline COMT activity moderated the effects of the drug on both outcomes, such that tolcapone-treated participants with higher baseline COMT activity had less stimulation (p = 0.008) and sedation (p = 0.053) than participants with lower baseline COMT activity and those treated with placebo. Additionally, alcohol-induced stimulation significantly mediated the interacting effects of baseline COMT activity and tolcapone on bar-laboratory self-administration. CONCLUSIONS Tolcapone may reduce subjective response to alcohol more effectively among individuals with preexisting high COMT activity an effect that could account for the drug's reduction of alcohol consumption among these individuals.
Collapse
Affiliation(s)
- Joseph P Schacht
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Matthew Kubicki
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Raymond F Anton
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
13
|
Gully B, Eaton E, Capone C, Haass-Koffler C. Treating posttraumatic stress disorder and alcohol use disorder comorbidity: Current pharmacological therapies and the future of MDMA-integrated psychotherapy. J Psychopharmacol 2023; 37:1182-1189. [PMID: 38009477 PMCID: PMC11549959 DOI: 10.1177/02698811231200880] [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] [Indexed: 11/29/2023]
Abstract
Posttraumatic stress disorder (PTSD) and alcohol use disorder (AUD) frequently co-occur in patients who have experienced trauma. This comorbidity leads to a vicious cycle where PTSD symptoms beget heavy drinking and vice versa. There are no FDA-approved medications to treat PTSD-AUD; therefore, individuals suffering from this comorbidity are treated with medication approved to treat the disorders separately or with off-label pharmacological interventions. However, these medications are limited in their efficacy for treating PTSD-AUD comorbidity. Emerging research on the nonclassical psychedelic drug 3,4-methylenedioxymethamphetamine (MDMA) suggests that it may be an effective drug used in conjunction with psychotherapy. The following reviews the current research for clinical pharmacotherapies, as well as MDMA-integrative psychotherapy as they pertain to PTSD and AUD in isolation and co-occurrence. Future directions for the role of psychedelic-integrative therapy for the treatment of this comorbidity are discussed.
Collapse
Affiliation(s)
- Brian Gully
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, USA
| | - Erica Eaton
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI, USA
- Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Christy Capone
- 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
- Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Carolina Haass-Koffler
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI, USA
- Carney Institute for Brain Science, Providence RI, Brown University
| |
Collapse
|
14
|
Diaz LA, Winder GS, Leggio L, Bajaj JS, Bataller R, Arab JP. New insights into the molecular basis of alcohol abstinence and relapse in alcohol-associated liver disease. Hepatology 2023:01515467-990000000-00605. [PMID: 37862466 DOI: 10.1097/hep.0000000000000645] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/11/2023] [Indexed: 10/22/2023]
Abstract
Alcohol use disorder remains a significant public health concern, affecting around 5% of adults worldwide. Novel pathways of damage have been described during the last years, providing insight into the mechanism of injury due to alcohol misuse beyond the direct effect of ethanol byproducts on the liver parenchyma and neurobehavioral mechanisms. Thus, the gut-liver-brain axis and immune system involvement could be therapeutic targets for alcohol use disorder. In particular, changes in gut microbiota composition and function, and bile acid homeostasis, have been shown with alcohol consumption and cessation. Alcohol can also directly disrupt intestinal and blood-brain barriers. Activation of the immune system can be triggered by intestinal barrier dysfunction and translocation of bacteria, pathogen-associated molecular patterns (such as lipopolysaccharide), cytokines, and damage-associated molecular patterns. These factors, in turn, promote liver and brain inflammation and the progression of liver fibrosis. Other involved mechanisms include oxidative stress, apoptosis, autophagy, and the release of extracellular vesicles and miRNA from hepatocytes. Potential therapeutic targets include gut microbiota (probiotics and fecal microbiota transplantation), neuroinflammatory pathways, as well as neuroendocrine pathways, for example, the ghrelin system (ghrelin receptor blockade), incretin mimetics (glucagon-like peptide-1 analogs), and the mineralocorticoid receptor system (spironolactone). In addition, support with psychological and behavioral treatments is essential to address the multiple dimensions of alcohol use disorder. In the future, a personalized approach considering these novel targets can contribute to significantly decreasing the alcohol-associated burden of disease.
Collapse
Affiliation(s)
- Luis Antonio Diaz
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institutes of Health, NIDA and NIAAA, Baltimore, Maryland, USA
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University and Central Virginia Veterans Health Care System, Richmond, Virginia, USA
| | - Ramon Bataller
- Liver Unit, Hospital Clinic, Institut d'Investigacions August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Medicine, Division of Gastroenterology, Schulich School of Medicine, Western University, London, Ontario, Canada
| |
Collapse
|
15
|
Teng PN, Barakat W, Tran SM, Tran ZM, Bateman NW, Conrads KA, Wilson KN, Oliver J, Gist G, Hood BL, Zhou M, Maxwell GL, Leggio L, Conrads TP, Lee MR. Brain proteomic atlas of alcohol use disorder in adult males. Transl Psychiatry 2023; 13:318. [PMID: 37833300 PMCID: PMC10575941 DOI: 10.1038/s41398-023-02605-0] [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: 03/14/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Alcohol use disorder (AUD) affects transcriptomic, epigenetic and proteomic expression in several organs, including the brain. There has not been a comprehensive analysis of altered protein abundance focusing on the multiple brain regions that undergo neuroadaptations occurring in AUD. We performed a quantitative proteomic analysis using a liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis of human postmortem tissue from brain regions that play key roles in the development and maintenance of AUD, the amygdala (AMG), hippocampus (HIPP), hypothalamus (HYP), nucleus accumbens (NAc), prefrontal cortex (PFC) and ventral tegmental area (VTA). Brain tissues were from adult males with AUD (n = 11) and matched controls (n = 16). Across the two groups, there were >6000 proteins quantified with differential protein abundance in AUD compared to controls in each of the six brain regions. The region with the greatest number of differentially expressed proteins was the AMG, followed by the HYP. Pathways associated with differentially expressed proteins between groups (fold change > 1.5 and LIMMA p < 0.01) were analyzed by Ingenuity Pathway Analysis (IPA). In the AMG, adrenergic, opioid, oxytocin, GABA receptor and cytokine pathways were among the most enriched. In the HYP, dopaminergic signaling pathways were the most enriched. Proteins with differential abundance in AUD highlight potential therapeutic targets such as oxytocin, CSNK1D (PF-670462), GABAB receptor and opioid receptors and may lead to the identification of other potential targets. These results improve our understanding of the molecular alterations of AUD across brain regions that are associated with the development and maintenance of AUD. Proteomic data from this study is publicly available at www.lmdomics.org/AUDBrainProteomeAtlas/ .
Collapse
Affiliation(s)
- Pang-Ning Teng
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Waleed Barakat
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Sophie M Tran
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Zoe M Tran
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Nicholas W Bateman
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Kelly A Conrads
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Katlin N Wilson
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Julie Oliver
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Glenn Gist
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Brian L Hood
- Women's Health Integrated Research Center, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Ming Zhou
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - G Larry Maxwell
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, Bethesda, Maryland, USA
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island, USA
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Thomas P Conrads
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Falls Church, VA, USA.
| | - Mary R Lee
- Veterans Affairs Medical Center, Washington, DC, USA.
| |
Collapse
|
16
|
De Oliveira Sergio T, Wean S, Katner SN, Hopf FW. The role of beta- and alpha-adrenergic receptors on alcohol drinking. Neuropharmacology 2023; 234:109545. [PMID: 37100382 PMCID: PMC11071639 DOI: 10.1016/j.neuropharm.2023.109545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 04/28/2023]
Abstract
Alcohol Use Disorders (AUD) is characterized by compulsion-like alcohol drinking (CLAD), where intake despite negative consequences can be a major clinical obstacle. With few treatment options available for AUD, there is a significant need for novel therapies. The noradrenergic system is an important hub for regulating stress responses and maladaptive drives for alcohol. Studies have shown that drugs targeting α1 adrenenergic receptors (ARs) may represent a pharmacological treatment for pathological drinking. However, the involvement of β ARs for treating human drinking has received scant investigation, and thus we sought to provide pre-clinical validation for possible AR utility for CLAD by analyzing whether β AR antagonists propranolol (β1/2), betaxolol (β1), and ICI, 118,551 (β2) impacted CLAD and alcohol-only drinking (AOD) in male Wistar rats. We found that the highest dose of propranolol tested systemically (10 mg/kg) reduced alcohol drinking, while 5 mg/kg propranolol reduced drinking with a trend to impact CLAD more than AOD, and with no effects of 2.5 mg/kg. Betaxolol (2.5 mg/kg) also decreased drinking, while ICI 118.551 had no effects. Also, while AR compounds might have utility for AUD, they can also lead to undesirable side effects. Here, a combination of ineffective doses of propranolol and prazosin reduced both CLAD and AOD. Finally, we investigated the effect of propranolol and betaxolol in two brain areas related to pathological drinking, the anterior insula (aINS) and medial prefrontal cortex (mPFC). Surprisingly, propranolol (1-10 μg) in aINS or mPFC did not affect CLAD or AOD. Together, our findings provide new pharmacological insights into noradrenergic regulation of alcohol consumption, which may inform AUD therapy.
Collapse
Affiliation(s)
| | - Sarah Wean
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA
| | - Simon N Katner
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA
| | - Frederic W Hopf
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA.
| |
Collapse
|
17
|
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
|
18
|
Munier J, Shen S, Rahal D, Hanna A, Marty V, O'Neill P, Fanselow M, Spigelman I. Chronic intermittent ethanol exposure disrupts stress-related tripartite communication to impact affect-related behavioral selection in male rats. Neurobiol Stress 2023; 24:100539. [PMID: 37131490 PMCID: PMC10149313 DOI: 10.1016/j.ynstr.2023.100539] [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: 11/22/2022] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 05/04/2023] Open
Abstract
Alcohol use disorder (AUD) is characterized by loss of intake control, increased anxiety, and susceptibility to relapse inducing stressors. Both astrocytes and neurons contribute to behavioral and hormonal consequences of chronic intermittent ethanol (CIE) exposure in animal models. Details on how CIE disrupts hypothalamic neuro-glial communication, which mediates stress responses are lacking. We conducted a behavioral battery (grooming, open field, reactivity to a single, uncued foot-shock, intermittent-access two-bottle choice ethanol drinking) followed by Ca2+ imaging in ex-vivo slices of paraventricular nucleus of the hypothalamus (PVN) from male rats exposed to CIE vapor or air-exposed controls. Ca2+ signals were evaluated in response to norepinephrine (NE) with or without selective α-adrenergic receptor (αAR) or GluN2B-containing N-methyl-D-aspartate receptor (NMDAR) antagonists, followed by dexamethasone (DEX) to mock a pharmacological stress response. Expectedly, CIE rats had altered anxiety-like, rearing, grooming, and drinking behaviors. Importantly, NE-mediated reductions in Ca2+ event frequency were blunted in both CIE neurons and astrocytes. Administration of the selective α1AR antagonist, prazosin, reversed this CIE-induced dysfunction in both cell types. Additionally, the pharmacological stress protocol reversed the altered basal Ca2+ signaling profile of CIE astrocytes. Signaling changes in astrocytes in response to NE were correlated with anxiety-like behaviors, such as the grooming:rearing ratio, suggesting tripartite synaptic function plays a role in switching between exploratory and stress-coping behavior. These data show how CIE exposure causes persistent changes to PVN neuro-glial function and provides the groundwork for how these physiological changes manifest in behavioral selection.
Collapse
Affiliation(s)
- J.J. Munier
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
- Corresponding author.
| | - S. Shen
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - D. Rahal
- Edna Bennett Pierce Prevention Research Center, The Pennsylvania State University, United States
| | - A. Hanna
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - V.N. Marty
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - P.R. O'Neill
- Hatos Center for Neuropharmacology, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, UCLA, United States
| | - M.S. Fanselow
- Department of Psychology, College of Life Sciences, Department of Psychiatry & Biobehavioral Science, David Geffen School of Medicine, UCLA, United States
| | - I. Spigelman
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
- Corresponding author. Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, 10833 Le Conte Avenue, 63-078 CHS, Los Angeles, CA, 90095-1668, United States.
| |
Collapse
|
19
|
Haass-Koffler CL. Utilizing precision medicine to treat alcohol use disorder: A commentary on the α1 receptor antagonist. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:432-434. [PMID: 36852535 PMCID: PMC10050091 DOI: 10.1111/acer.15014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/07/2023] [Indexed: 03/01/2023]
Affiliation(s)
- Carolina L Haass-Koffler
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island, USA
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island, USA
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
20
|
Tonetto S, Weikop P, Brudek T, Thomsen M. Behavioral and biochemical effects of alcohol withdrawal in female C3H/HeNRj and C57BL/6JRj mice. Front Behav Neurosci 2023; 17:1143720. [PMID: 36910126 PMCID: PMC9995974 DOI: 10.3389/fnbeh.2023.1143720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/08/2023] [Indexed: 02/25/2023] Open
Abstract
Background Alcohol use disorder (AUD) is a major problem of our society and is often characterized and worsened by relapse. Prolonged alcohol exposure leads to numerous biochemical alterations that, upon cessation of alcohol intake, cause an array of immediate and lasting withdrawal symptoms. Acute withdrawal and neuroinflammation can be harmful in themselves, and lasting withdrawal symptoms contribute to relapse. Here, we conducted an initial feasibility study assessing several behavioral and neurochemical factors in female C3H/HeNRj (C3H) and C57BL/6JRj (B6) mice to determine which strain showed the clearest alcohol withdrawal symptoms during long-term abstinence and neurochemical alterations following re-exposure. Methods Female C3H and B6 mice (n = 12 per group/strain) were intermittently exposed to alcohol-containing or control liquid diets for 3 weeks. Acute and prolonged withdrawal symptoms were assessed over a period of 3 weeks using a battery of behavioral test, comprised of alcohol self-administration, anhedonia, hyperalgesia, anxiety-like and depressive-like disturbances. Brain inflammation was measured by multiplex cytokine assay. Monoamine levels in the hippocampus and striatum, as well as exploratory analyses of cations levels in the cerebellum, were assessed by High-Performance Liquid Chromatography (HPLC). Results Both C3H and B6 alcohol-exposed mice displayed decreased saccharin intake or preference and higher stress levels assessed by ultrasonic vocalizations (USVs) recordings. B6 but not C3H alcohol-exposed mice also exhibited a slower decline of alcohol oral self-administration (OSA), hyperalgesia, elevated brain TNF-α and elevated serotonin turnover. Conclusion Our findings highlight the suitability of the B6 strain to study the behavioral and neurochemical alterations caused by alcohol withdrawal and the potential efficacy of experimental treatments, not only in early detoxification, but also in prolonged abstinence. The feasibility of these assays is important because long-lasting withdrawal symptoms are often the main cause of relapse in alcohol-dependent patients.
Collapse
Affiliation(s)
- Simone Tonetto
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, University Hospital of Copenhagen, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Tomasz Brudek
- Copenhagen Center for Translational Research, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Morgane Thomsen
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, University Hospital of Copenhagen, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
21
|
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
|
22
|
Haass-Koffler CL, Magill M, Cannella N, Brown JC, Aoun EG, Cioe PA, Sinha R, Swift RM, Ciccocioppo R, Leggio L. Mifepristone as a pharmacological intervention for stress-Induced alcohol craving: a translational crossover randomized trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.02.23284122. [PMID: 36711869 PMCID: PMC9882427 DOI: 10.1101/2023.01.02.23284122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Preclinical and clinical work suggests that mifepristone (glucocorticoid receptor antagonist), may be a viable treatment for alcohol use disorder (AUD). The aim of this work was to translate our preclinical mifepristone study using yohimbine (α2 receptor antagonist) stress-induced reinstatement of alcohol-seeking to a clinical setting. This was a Phase 1/2, outpatient, cross-over, randomized, double-blind, placebo-controlled trial with non-treatment-seeking individuals with AUD ( N =32). We investigated the safety, alcohol craving and consumption after oral administration of mifepristone (600mg daily for a week) in a human laboratory study comprised of administration of yohimbine in a cue-reactivity procedure and alcohol self-administration. Outcomes were assessed using Generalized Estimating Equations and mediation and moderation analyses assessed mechanisms of action and precision medicine targets. We did not observe serious adverse events related to the study drugs or study procedure and mild to moderate non-serious adverse events were reported by both study conditions. Also, there was no statistically-significant difference between the mifepristone and placebo in the hemodynamic response, alcohol subjective effects and pharmacokinetics parameters. Mifepristone significantly reduced alcohol craving and increased cortisol levels. Mifepristone-induced cortisol increase was not a mediator of alcohol craving. Moderation analysis with family history density of AUD (FHDA) and mifepristone, suggested that reduced craving was present in individuals with low , but not high FHDA. Mifepristone, compared to placebo, did not reduce alcohol consumption in the laboratory or in a naturalistic setting. This study successfully translated a preclinical paradigm to a human laboratory study confirming safety, tolerability and efficacy of mifepristone in an alcohol paradigm. Mediation analysis showed that the effect of mifepristone on craving was not related to mifepristone-induced increases in cortisol and moderation of FHDA suggested the importance of evaluating AUD endophenotypes for pharmacotherapies. Clinical trial registration Clinicaltrials.gov ; NCT02243709. IND/FDA 121984, mifepristone and yohimbine (Holder: Haass-Koffler).
Collapse
|
23
|
Fox HC, Milivojevic V, Sinha R. Therapeutics for Substance-Using Women: The Need to Elucidate Sex-Specific Targets for Better-Tailored Treatments. Handb Exp Pharmacol 2023; 282:127-161. [PMID: 37592081 DOI: 10.1007/164_2023_687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
In the last decade, alcohol consumption in the US has risen by 84% in women compared with 35% in men. Furthermore, research has shown that sex- and gender-related differences may disadvantage women in terms of developing a range of psychological, cognitive, and medical problems considerably earlier in their drinking history than men, and despite consuming a similar quantity of substances. While this "telescoping" process has been acknowledged in the literature, a concomitant understanding of the underlying biobehavioral mechanisms, and an increase in the development of specific treatments tailored to women, has not occurred. In the current chapter we focus on understanding why the need for personalized, sex-specific medications is imperative, and highlight some of the potential sex-specific gonadal and stress-related adaptations underpinning the accelerated progress from controlled to compulsive drug and alcohol seeking in women. We additionally discuss the efficacy of these mechanisms as novel targets for medications development, using exogenous progesterone and guanfacine as examples. Finally, we assess some of the challenges faced and progress made in terms of developing innovative medications in women. We suggest that agents such as exogenous progesterone and adrenergic medications, such as guanfacine, may provide some efficacy in terms of attenuating stress-induced craving for several substances, as well as improving the ability to emotionally regulate in the face of stress, preferentially in women. However, to fully leverage the potential of these therapeutics in substance-using women, greater focus needs to the placed on reducing barriers to treatment and research by encouraging women into clinical trials.
Collapse
Affiliation(s)
- Helen C Fox
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Verica Milivojevic
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Rajita Sinha
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
24
|
Ray LA, Nieto SJ, Grodin EN. Translational models of addiction phenotypes to advance addiction pharmacotherapy. Ann N Y Acad Sci 2023; 1519:118-128. [PMID: 36385614 PMCID: PMC10823887 DOI: 10.1111/nyas.14929] [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] [Indexed: 11/19/2022]
Abstract
Alcohol and substance use disorders are heterogeneous conditions with limited effective treatment options. While there have been prior attempts to classify addiction subtypes, they have not been translated into clinical practice. In an effort to better understand heterogeneity in psychiatric disorders, the National Institute for Mental Health Research Domain Criteria (RDoC) has challenged scientists to think beyond diagnostic symptoms and to consider the underlying features of psychopathology from a neuroscience-based framework. The field of addiction has grappled with this approach by considering several key constructs with the potential to capture RDoC domains. This critical review will focus on the efforts to apply translational models of addiction phenomenology in human clinical samples, including their relative strengths and weaknesses. Opportunities for forward and reverse translation are also discussed. Deep behavioral phenotyping using neuroscience-informed batteries shows promise for a better understanding of the clinical neuroscience of addiction and advancing precision medicine for alcohol and substance use disorders.
Collapse
Affiliation(s)
- Lara A. Ray
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
- Shirley & Stefan Hatos Center for Neuropharmacology, University of California at Los Angeles, Los Angeles, CA, USA
- Jane & Terry Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA, USA
| | - Steven J. Nieto
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Erica N. Grodin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
25
|
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
|
26
|
Price JL, Bates ME, Pawlak AP, Uhouse SG, Todaro SM, Morgano J, Buckman JF. Use and perceived usefulness of a just-in-time resonance breathing intervention adjunct for substance use disorder: Contextual and physiological predictors. Front Psychiatry 2022; 13:945751. [PMID: 36159943 PMCID: PMC9490325 DOI: 10.3389/fpsyt.2022.945751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/29/2022] [Indexed: 11/20/2022] Open
Abstract
Craving for alcohol and other drugs is often described as a momentary hyperarousal state that interferes with one's ability to use top-down strategies. As such, it may be best interrupted 'in the moment' through bottom-up modulation. We recently reported that episodic resonance paced breathing (eRPB) delivered via mobile phone app as an add-on to outpatient treatment for substance use disorder (SUD) was effective at dampening craving over the course of an 8-week intervention (NCT#02579317). However, not all participants engaged with the eRPB app and there was high intra- and inter-individual variability in weekly ratings of usefulness. Here we examined baseline demographic, physiological, and psychiatric measures as well as time-varying exposure to positive, negative, and temptation craving triggers as predictors of frequency of eRPB app use and ratings of usefulness. Seventy-seven outpatient women were randomized to an eRPB (0.1 Hz) or a faster paced breathing sham (0.23 Hz) condition. Baseline measures were assessed within the first 3 weeks of treatment entry prior to randomization. App use frequency, ratings of usefulness, and trigger exposure were measured weekly throughout the intervention. Variables were entered into marginal means models with forward stepwise model selection and examined as predictors of use and usefulness. Frequent app use was associated with a lifetime alcohol use disorder (AUD) diagnosis (p = 0.026), higher ratings of usefulness (p < 0.001), and fewer exposures to positive triggers (e.g., celebration, socialization; p < 0.001). There was a trend-level association between frequency of app use and greater cardiovascular capacity at baseline (p = 0.088). Higher ratings of usefulness were associated with greater exposure to negative triggers (e.g,. loneliness, frustration; p < 0.001) and parasympathetic dysregulation at baseline (p = 0.05). A positive relationship between app use frequency and ratings of usefulness was present only in the eRPB group (p = 0.045). Matching ideal candidates and moments to an arousal modulation anti-craving intervention can help streamline screening and implementation of eRPB in the treatment of SUD. Clinical Trial Registration https://clinicaltrials.gov/ct2/show/NCT02579317, identifier NCT02579317.
Collapse
Affiliation(s)
- Julianne L. Price
- Cardiac Neuroscience Laboratory, Center of Alcohol and Substance Use Studies, Rutgers University—New Brunswick, Piscataway, NJ, United States
- Department of Kinesiology and Health, Rutgers University—New Brunswick, Piscataway, NJ, United States
| | - Marsha E. Bates
- Cardiac Neuroscience Laboratory, Center of Alcohol and Substance Use Studies, Rutgers University—New Brunswick, Piscataway, NJ, United States
- Department of Kinesiology and Health, Rutgers University—New Brunswick, Piscataway, NJ, United States
| | - Anthony P. Pawlak
- Cardiac Neuroscience Laboratory, Center of Alcohol and Substance Use Studies, Rutgers University—New Brunswick, Piscataway, NJ, United States
| | - Sarah Grace Uhouse
- Cardiac Neuroscience Laboratory, Center of Alcohol and Substance Use Studies, Rutgers University—New Brunswick, Piscataway, NJ, United States
- Department of Psychology, Rutgers University—New Brunswick, Piscataway, NJ, United States
| | - Sabrina M. Todaro
- Cardiac Neuroscience Laboratory, Center of Alcohol and Substance Use Studies, Rutgers University—New Brunswick, Piscataway, NJ, United States
- Department of Psychology, College of Health Sciences, University of Rhode Island, South Kingstown, RI, United States
| | - Julie Morgano
- Cardiac Neuroscience Laboratory, Center of Alcohol and Substance Use Studies, Rutgers University—New Brunswick, Piscataway, NJ, United States
| | - Jennifer F. Buckman
- Cardiac Neuroscience Laboratory, Center of Alcohol and Substance Use Studies, Rutgers University—New Brunswick, Piscataway, NJ, United States
- Department of Kinesiology and Health, Rutgers University—New Brunswick, Piscataway, NJ, United States
| |
Collapse
|
27
|
Alluri S, Eisenberg SM, Grisanti LA, Tanner M, Volkow ND, Kim SW, Kil KE. Preclinical evaluation of new C-11 labeled benzo-1,4-dioxane PET radiotracers for brain α2C adrenergic receptors. Eur J Med Chem 2022; 243:114764. [DOI: 10.1016/j.ejmech.2022.114764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022]
|
28
|
Munier JJ, Marty VN, Spigelman I. Sex differences in α-adrenergic receptor function contribute to impaired hypothalamic metaplasticity following chronic intermittent ethanol exposure. Alcohol Clin Exp Res 2022; 46:1384-1396. [PMID: 35791038 PMCID: PMC9612407 DOI: 10.1111/acer.14900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/03/2022] [Accepted: 06/23/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Individuals with alcohol use disorder (AUD) exhibit maladaptive responses of the hypothalamic-pituitary-adrenal (HPA) axis to stress, which has been linked to high rates of relapse to drinking among abstinent individuals. Corticotropin-releasing factor (CRF) parvocellular neuroendocrine cells (PNCs) within the paraventricular nucleus of the hypothalamus (PVN) are critical to stress-induced HPA axis activation. Here, we investigate sex differences in synaptic transmission and plasticity in PNCs following the application of the stress-associated neurotransmitter norepinephrine (NE) in a rat model of AUD. METHODS Adult Sprague-Dawley rats were exposed to 40 days of chronic intermittent ethanol (CIE) vapor and 30 to 108 days of protracted withdrawal. We measured changes in holding current, evoked synaptic currents, and short-term glutamatergic plasticity (STP) in putative PNCs following the application of NE (10 μM) with and without the selective α1 adrenergic receptor (AR) antagonist prazosin (10 μM) or the α2AR antagonist atipamezole (10 μM). The experiments were performed using whole-cell patch clamp recordings in slices from CIE rats and air-exposed controls. RESULTS NE application caused two distinct effects: a depolarizing, inward, postsynaptic current and a reduction in amplitude of an evoked glutamatergic excitatory postsynaptic current (eEPSC). Both effects were sex- and CIE-specific. Prazosin blocked the postsynaptic inward current, while atipamezole blocked the NE-mediated suppression of eEPSCs. Additionally, STP formation was facilitated following NE application only in stress-naïve males and this response was lost in stressed animals exposed to a 30-min restraint stress following CIE exposure. Furthermore, NE + prazosin restored STP formation in stressed CIE males. CONCLUSIONS NE exerts excitatory and inhibitory effects on CRF PVN PNCs, and both effects are influenced by sex and CIE. Behavioral and hormonal responses to stress are influenced by STP formation within the PVN, which is lost following CIE and restored with the preapplication of prazosin. The selective blockade of α1AR may, therefore, ameliorate CIE-induced deficits in HPA responses to stress in a sex-specific manner.
Collapse
Affiliation(s)
- Joseph J. Munier
- Department of Molecular, Cellular, and Integrative Physiology, University of California, Los Angeles, CA, USA,Laboratory of Neuropharmacology, Section of Biosystems and Function, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Vincent N. Marty
- Laboratory of Neuropharmacology, Section of Biosystems and Function, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Igor Spigelman
- Laboratory of Neuropharmacology, Section of Biosystems and Function, School of Dentistry, University of California, Los Angeles, CA, USA
| |
Collapse
|
29
|
Involvement of the ghrelin system in the maintenance and reinstatement of cocaine-motivated behaviors: a role of adrenergic action at peripheral β1 receptors. Neuropsychopharmacology 2022; 47:1449-1460. [PMID: 34923576 PMCID: PMC9206024 DOI: 10.1038/s41386-021-01249-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 12/30/2022]
Abstract
Cocaine addiction is a significant medical and public concern. Despite decades of research effort, development of pharmacotherapy for cocaine use disorder remains largely unsuccessful. This may be partially due to insufficient understanding of the complex biological mechanisms involved in the pathophysiology of this disorder. In the present study, we show that: (1) elevation of ghrelin by cocaine plays a critical role in maintenance of cocaine self-administration and cocaine-seeking motivated by cocaine-conditioned stimuli; (2) acquisition of cocaine-taking behavior is associated with the acquisition of stimulatory effects of cocaine by cocaine-conditioned stimuli on ghrelin secretion, and with an upregulation of ghrelin receptor mRNA levels in the ventral tegmental area (VTA); (3) blockade of ghrelin signaling by pretreatment with JMV2959, a selective ghrelin receptor antagonist, dose-dependently inhibits reinstatement of cocaine-seeking triggered by either cocaine or yohimbine in behaviorally extinguished animals with a history of cocaine self-administration; (4) JMV2959 pretreatment also inhibits brain stimulation reward (BSR) and cocaine-potentiated BSR maintained by optogenetic stimulation of VTA dopamine neurons in DAT-Cre mice; (5) blockade of peripheral adrenergic β1 receptors by atenolol potently attenuates the elevation in circulating ghrelin induced by cocaine and inhibits cocaine self-administration and cocaine reinstatement triggered by cocaine. These findings demonstrate that the endogenous ghrelin system plays an important role in cocaine-related addictive behaviors and suggest that manipulating and targeting this system may be viable for mitigating cocaine use disorder.
Collapse
|
30
|
Blum K, Brodie MS, Pandey SC, Cadet JL, Gupta A, Elman I, Thanos PK, Gondre-Lewis MC, Baron D, Kazmi S, Bowirrat A, Febo M, Badgaiyan RD, Braverman ER, Dennen CA, Gold MS. Researching Mitigation of Alcohol Binge Drinking in Polydrug Abuse: KCNK13 and RASGRF2 Gene(s) Risk Polymorphisms Coupled with Genetic Addiction Risk Severity (GARS) Guiding Precision Pro-Dopamine Regulation. J Pers Med 2022; 12:1009. [PMID: 35743793 PMCID: PMC9224860 DOI: 10.3390/jpm12061009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/12/2022] [Accepted: 06/16/2022] [Indexed: 11/22/2022] Open
Abstract
Excessive alcohol intake, e.g., binge drinking, is a serious and mounting public health problem in the United States and throughout the world. Hence the need for novel insights into the underlying neurobiology that may help improve prevention and therapeutic strategies. Therefore, our group employed a darkness-induced alcohol intake protocol to define the reward deficiency domains of alcohol and other substance use disorders in terms of reward pathways' reduced dopamine signaling and its restoration via specifically-designed therapeutic compounds. It has been determined that KCNK13 and RASGRF2 genes, respectively, code for potassium two pore domain channel subfamily K member 13 and Ras-specific guanine nucleotide-releasing factor 2, and both genes have important dopamine-related functions pertaining to alcohol binge drinking. We present a hypothesis that identification of KCNK13 and RASGRF2 genes' risk polymorphism, coupled with genetic addiction risk score (GARS)-guided precision pro-dopamine regulation, will mitigate binge alcohol drinking. Accordingly, we review published reports on the benefits of this unique approach and provide data on favorable outcomes for both binge-drinking animals and drunk drivers, including reductions in alcohol intake and prevention of relapse to drinking behavior. Since driving under the influence of alcohol often leads to incarceration rather than rehabilitation, there is converging evidence to support the utilization of GARS with or without KCNK13 and RASGRF2 risk polymorphism in the legal arena, whereby the argument that "determinism" overrides the "free will" account may be a plausible defense strategy. Obviously, this type of research is tantamount to helping resolve a major problem related to polydrug abuse.
Collapse
Affiliation(s)
- Kenneth Blum
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX 78701, USA; (E.R.B.); (C.A.D.)
- Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of Provost), Western University Health Sciences, Pomona, CA 91766, USA; (D.B.); (M.F.)
- Institute of Psychology, ELTE Eötvös Loránd University, Egyetem tér 1-3, 1053 Budapest, Hungary
- Department of Psychiatry, School of Medicine, University of Vermont, Burlington, VT 05405, USA
- Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Centre, Dayton, OH 45324, USA
| | - Mark S. Brodie
- Center for Alcohol Research in Epigenetics, Departments of Physiology and Biophysics, and Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA; (M.S.B.); (S.C.P.)
| | - Subhash C. Pandey
- Center for Alcohol Research in Epigenetics, Departments of Physiology and Biophysics, and Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA; (M.S.B.); (S.C.P.)
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA;
| | - Igor Elman
- Center for Pain and the Brain (P.A.I.N Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA;
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY 14203, USA;
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Marjorie C. Gondre-Lewis
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA;
| | - David Baron
- Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of Provost), Western University Health Sciences, Pomona, CA 91766, USA; (D.B.); (M.F.)
| | - Shan Kazmi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Abdalla Bowirrat
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Marcelo Febo
- Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of Provost), Western University Health Sciences, Pomona, CA 91766, USA; (D.B.); (M.F.)
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, Long School of Medicine, University of Texas Medical Center, San Antonio, TX 78229, USA;
| | - Eric R. Braverman
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX 78701, USA; (E.R.B.); (C.A.D.)
| | - Catherine A. Dennen
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX 78701, USA; (E.R.B.); (C.A.D.)
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
31
|
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
|
32
|
Keller BN, Hajnal A, Browning KN, Arnold AC, Silberman Y. Involvement of the Dorsal Vagal Complex in Alcohol-Related Behaviors. Front Behav Neurosci 2022; 16:801825. [PMID: 35330845 PMCID: PMC8940294 DOI: 10.3389/fnbeh.2022.801825] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/19/2022] [Indexed: 12/20/2022] Open
Abstract
The neurobiological mechanisms that regulate the development and maintenance of alcohol use disorder (AUD) are complex and involve a wide variety of within and between systems neuroadaptations. While classic reward, preoccupation, and withdrawal neurocircuits have been heavily studied in terms of AUD, viable treatment targets from this established literature have not proven clinically effective as of yet. Therefore, examination of additional neurocircuitries not classically studied in the context of AUD may provide novel therapeutic targets. Recent studies demonstrate that various neuropeptides systems are important modulators of alcohol reward, seeking, and intake behaviors. This includes neurocircuitry within the dorsal vagal complex (DVC), which is involved in the control of the autonomic nervous system, control of intake of natural rewards like food, and acts as a relay of interoceptive sensory information via interactions of numerous gut-brain peptides and neurotransmitter systems with DVC projections to central and peripheral targets. DVC neuron subtypes produce a variety of neuropeptides and transmitters and project to target brain regions critical for reward such as the mesolimbic dopamine system as well as other limbic areas important for the negative reinforcing and aversive properties of alcohol withdrawal such as the extended amygdala. This suggests the DVC may play a role in the modulation of various aspects of AUD. This review summarizes the current literature on neurotransmitters and neuropeptides systems in the DVC (e.g., norepinephrine, glucagon-like peptide 1, neurotensin, cholecystokinin, thyrotropin-releasing hormone), and their potential relevance to alcohol-related behaviors in humans and rodent models for AUD research. A better understanding of the role of the DVC in modulating alcohol related behaviors may lead to the elucidation of novel therapeutic targets for drug development in AUD.
Collapse
|
33
|
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
|
34
|
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
|
35
|
Curley DE, Webb AE, Sheffler DJ, Haass-Koffler CL. Corticotropin Releasing Factor Binding Protein as a Novel Target to Restore Brain Homeostasis: Lessons Learned From Alcohol Use Disorder Research. Front Behav Neurosci 2021; 15:786855. [PMID: 34912198 PMCID: PMC8667027 DOI: 10.3389/fnbeh.2021.786855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [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: 11/13/2022] Open
Abstract
Stress is well-known to contribute to the development of many psychiatric illnesses including alcohol and substance use disorder (AUD and SUD). The deleterious effects of stress have also been implicated in the acceleration of biological age, and age-related neurodegenerative disease. The physio-pathology of stress is regulated by the corticotropin-releasing factor (CRF) system, the upstream component of the hypothalamic-pituitary-adrenal (HPA) axis. Extensive literature has shown that dysregulation of the CRF neuroendocrine system contributes to escalation of alcohol consumption and, similarly, chronic alcohol consumption contributes to disruption of the stress system. The CRF system also represents the central switchboard for regulating homeostasis, and more recent studies have found that stress and aberrations in the CRF pathway are implicated in accelerated aging and age-related neurodegenerative disease. Corticotropin releasing factor binding protein (CRFBP) is a secreted glycoprotein distributed in peripheral tissues and in specific brain regions. It neutralizes the effects of CRF by sequestering free CRF, but may also possess excitatory function by interacting with CRF receptors. CRFBP's dual role in influencing CRF bioavailability and CRF receptor signaling has been shown to have a major part in the HPA axis response. Therefore, CRFBP may represent a valuable target to treat stress-related illness, including: development of novel medications to treat AUD and restore homeostasis in the aging brain. This narrative review focuses on molecular mechanisms related to the role of CRFBP in the progression of addictive and psychiatric disorders, biological aging, and age-related neurodegenerative disease. We provide an overview of recent studies investigating modulation of this pathway as a potential therapeutic target for AUD and age-related neurodegenerative disease.
Collapse
Affiliation(s)
- Dallece E. Curley
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, United States
- Neuroscience Graduate Program, Department of Neuroscience, Brown University, Providence, RI, United States
| | - Ashley E. Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
- Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Douglas J. Sheffler
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Carolina L. Haass-Koffler
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, United States
- Carney Institute for Brain Science, Brown University, Providence, RI, United States
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, United States
| |
Collapse
|
36
|
Gutridge AM, Chakraborty S, Varga BR, Rhoda ES, French AR, Blaine AT, Royer QH, Cui H, Yuan J, Cassell RJ, Szabó M, Majumdar S, van Rijn RM. Evaluation of Kratom Opioid Derivatives as Potential Treatment Option for Alcohol Use Disorder. Front Pharmacol 2021; 12:764885. [PMID: 34803709 PMCID: PMC8596301 DOI: 10.3389/fphar.2021.764885] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose:Mitragyna speciosa extract and kratom alkaloids decrease alcohol consumption in mice at least in part through actions at the δ-opioid receptor (δOR). However, the most potent opioidergic kratom alkaloid, 7-hydroxymitragynine, exhibits rewarding properties and hyperlocomotion presumably due to preferred affinity for the mu opioid receptor (µOR). We hypothesized that opioidergic kratom alkaloids like paynantheine and speciogynine with reduced µOR potency could provide a starting point for developing opioids with an improved therapeutic window to treat alcohol use disorder. Experimental Approach: We characterized paynantheine, speciociliatine, and four novel kratom-derived analogs for their ability to bind and activate δOR, µOR, and κOR. Select opioids were assessed in behavioral assays in male C57BL/6N WT and δOR knockout mice. Key Results: Paynantheine (10 mg∙kg−1, i.p.) produced aversion in a limited conditioned place preference (CPP) paradigm but did not produce CPP with additional conditioning sessions. Paynantheine did not produce robust antinociception but did block morphine-induced antinociception and hyperlocomotion. Yet, at 10 and 30 mg∙kg−1 doses (i.p.), paynantheine did not counteract morphine CPP. 7-hydroxypaynantheine and 7-hydroxyspeciogynine displayed potency at δOR but limited µOR potency relative to 7-hydroxymitragynine in vitro, and dose-dependently decreased voluntary alcohol consumption in WT but not δOR in KO mice. 7-hydroxyspeciogynine has a maximally tolerated dose of at least 10 mg∙kg−1 (s.c.) at which it did not produce significant CPP neither alter general locomotion nor induce noticeable seizures. Conclusion and Implications: Derivatizing kratom alkaloids with the goal of enhancing δOR potency and reducing off-target effects could provide a pathway to develop novel lead compounds to treat alcohol use disorder with an improved therapeutic window.
Collapse
Affiliation(s)
- Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Soumen Chakraborty
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Balazs R Varga
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Elizabeth S Rhoda
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Alexander R French
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States
| | - Arryn T Blaine
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Quinten H Royer
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Haoyue Cui
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Jinling Yuan
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Robert J Cassell
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Drug Discovery, West Lafayette, IN, United States
| | | | - Susruta Majumdar
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States.,Purdue Institute for Drug Discovery, West Lafayette, IN, United States
| |
Collapse
|
37
|
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
|
38
|
Morley KC, Perry CJ, Watt J, Hurzeler T, Leggio L, Lawrence AJ, Haber P. New approved and emerging pharmacological approaches to alcohol use disorder: a review of clinical studies. Expert Opin Pharmacother 2021; 22:1291-1303. [PMID: 33615945 DOI: 10.1080/14656566.2021.1892641] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
introduction: The number of medications approved for AUD is small and they generally have limited efficacy. We need new pharmacotherapies for the management of AUD.Areas covered: In this review, the authors aim to synthesise literature for new approved and emerging pharmacotherapies for AUD. Recently approved medications include nalmefene, which was approved in Europe and Australia for the purposes of controlled drinking. Baclofen has also been approved in France but not in other countries. Off label medications including topiramate and gabapentin have received significant attention with multiple RCTs and meta-analyses and have widespread use in several countries including the USA. Several novel medications have emerged over the last decade but further work is required to determine their efficacy and safety for the widespread management of AUD.Expert opinion: Despite significant advances in our understanding of the neurobiological basis of factors that contribute to the development and maintenance of AUD, there have been few new AUD medications approved for almost 20 years. There are many challenges to the development and introduction of new pharmacotherapies for AUD. Strategies for improving the translational pipeline include drug repurposing and utilisation of human acute laboratory models.
Collapse
Affiliation(s)
- Kirsten C Morley
- Central Clinical School, Sydney School of Medicine, Faculty of Medicine & Health, University of Sydney, NSW, Australia.,Edith Collins Centre (Alcohol, Drugs and Toxicology), Sydney Local Health District, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Christina J Perry
- University of Melbourne, Mental Health Theme, Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Joshua Watt
- Edith Collins Centre (Alcohol, Drugs and Toxicology), Sydney Local Health District, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Tristan Hurzeler
- Central Clinical School, Sydney School of Medicine, Faculty of Medicine & Health, University of Sydney, NSW, Australia
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, USA.,Department of Neuroscience, Georgetown University Medical Center, Washington, USA.,Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, USA
| | - Andrew J Lawrence
- University of Melbourne, Mental Health Theme, Florey Institute of Neuroscience and Mental Health, Parkville, Australia.,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, USA
| | - Paul Haber
- Central Clinical School, Sydney School of Medicine, Faculty of Medicine & Health, University of Sydney, NSW, Australia.,Edith Collins Centre (Alcohol, Drugs and Toxicology), Sydney Local Health District, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.,Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, USA
| |
Collapse
|
39
|
Abstract
Bislang sind nur wenige Medikamente zur pharmakologischen Rückfallprophylaxe der Alkoholabhängigkeit zugelassen. Neben dem in Deutschland nicht mehr vertriebenen Disulfiram sind es die Opioidantagonisten Naltrexon und Nalmefen sowie das vermutlich über glutamaterge Neurone wirkende Acamprosat. Baclofen und γ‑Hydroxybutyrat (GHB) sind in einzelnen Ländern zugelassen. Wirkstoffe wie z. B. Vareniclin, Gabapentin und Topiramat können für die Rückfallprophylaxe der Alkoholabhängigkeit von Interesse sein, jedoch ist bislang keine Zulassung erfolgt. Vor dem Hintergrund der zur Revision anstehenden S3-Leitlinie zur Diagnose und Behandlung alkoholbezogener Störungen wird der heutige Kenntnisstand zur Pharmakotherapie der Alkoholabhängigkeit dargestellt.
Collapse
|
40
|
Hedges DM, Yorgason JT, Brundage JN, Wadsworth HA, Williams B, Steffensen SC, Roberto M. Corticotropin releasing factor, but not alcohol, modulates norepinephrine release in the rat central nucleus of the amygdala. Neuropharmacology 2020; 179:108293. [PMID: 32871155 DOI: 10.1016/j.neuropharm.2020.108293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/20/2022]
Abstract
Alcohol misuse and dependence is a widespread health problem. The central nucleus of the amygdala (CeA) plays important roles in both the anxiety associated with alcohol (ethanol) dependence and the increased alcohol intake that is observed during withdrawal in dependent animals. We and others have shown the essential involvement of the corticotropin releasing factor (CRF) system in alcohol's synaptic effects on the CeA and in the development of ethanol dependence. Another system that has been shown to be critically involved in the molecular underpinnings of alcohol dependence is the norepinephrine (NE) system originating in the locus coeruleus. Both the CRF and NE systems act in concert to facilitate a stress response: central amygdalar afferents release CRF in the locus coeruleus promoting widespread release of NE. In this study, we are the first to use fast-scan cyclic voltammetry to classify local electrically-evoked NE release in the CeA and to determine if acute alcohol and CRF modulate it. Evoked NE release is action potential dependent, is abolished after depletion of monoaminergic vesicles, differs pharmacologically from dopamine release, is insensitive to acute alcohol, and decreases in response to locally applied CRF. Taken together, these results indicate that NE release in the CeA is released canonically in a vesicular-dependent manner, and that while acute alcohol does not directly alter NE release, CRF decreases it. Our results suggest that CRF acts locally on NE terminals as negative feedback and potentially prevents hyperactivation of the CRF-norepinephrine stress pathway.
Collapse
Affiliation(s)
- David M Hedges
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA.
| | - Jordan T Yorgason
- Neuroscience Program, Brigham Young University, Provo, UT, 84602, USA.
| | - James N Brundage
- Neuroscience Program, Brigham Young University, Provo, UT, 84602, USA
| | | | - Benjamin Williams
- Neuroscience Program, Brigham Young University, Provo, UT, 84602, USA
| | | | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
41
|
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
|
42
|
Amaral VCS, Morais-Silva G, Laverde CF, Marin MT. Susceptibility to extinction and reinstatement of ethanol-induced conditioned place preference is related to differences in astrocyte cystine-glutamate antiporter content. Neurosci Res 2020; 170:245-254. [PMID: 32653617 DOI: 10.1016/j.neures.2020.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/22/2020] [Accepted: 07/07/2020] [Indexed: 12/25/2022]
Abstract
Individual susceptibility to alcohol effects plays an important role in the development of alcohol addiction and studies have shown that glutamate release is altered after chronic ethanol consumption. The cystine-glutamate antiporter (xCT) is a protein that regulates glutamate release. However, little is known about the relationship between xCT levels and this individual susceptibility. Thus, this study aimed to evaluate the relationship between the extinction and stress-induced reinstatement of ethanol conditioned place preference (CPP) and xCT levels in the medial prefrontal cortex (mPFC), nucleus accumbens (NAcc) and amygdala (Amy). Male Swiss mice were submitted to a CPP procedure followed by an extinction protocol and then identified as those which extinguished the CPP and those that did not. In another cohort, mice that extinguished the CPP were submitted to a protocol of stress-induced reinstatement. Immediately after the tests, brains were removed for xCT quantification. The xCT levels were significantly lower in the mPFC and NAcc of mice that did not extinguish CPP. Moreover, mice that were susceptible to stress-induced reinstatement of CPP had lower levels of xCT in the NAcc. Our results suggest that individual susceptibility to the extinction and reinstatement of ethanol CPP is related to alterations in xCT levels.
Collapse
Affiliation(s)
- Vanessa Cristiane Santana Amaral
- Laboratory of Pharmacology and Toxicology of Natural and Synthetic Products, State University of Goias, Exact and Technological Sciences Campus, Anapolis, GO, Brazil; São Paulo State University (UNESP), School of Pharmaceutical Sciences, Laboratory of Pharmacology, Araraquara, SP, Brazil; Joint Graduate Program in Physiological Sciences (PIPGCF), UFSCar/UNESP, São Carlos/Araraquara, SP, Brazil
| | - Gessynger Morais-Silva
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Laboratory of Pharmacology, Araraquara, SP, Brazil; Joint Graduate Program in Physiological Sciences (PIPGCF), UFSCar/UNESP, São Carlos/Araraquara, SP, Brazil
| | - Celina F Laverde
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Laboratory of Pharmacology, Araraquara, SP, Brazil; Joint Graduate Program in Physiological Sciences (PIPGCF), UFSCar/UNESP, São Carlos/Araraquara, SP, Brazil
| | - Marcelo T Marin
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Laboratory of Pharmacology, Araraquara, SP, Brazil; Joint Graduate Program in Physiological Sciences (PIPGCF), UFSCar/UNESP, São Carlos/Araraquara, SP, Brazil.
| |
Collapse
|
43
|
Müller TE, Fontana BD, Bertoncello KT, Franscescon F, Mezzomo NJ, Canzian J, Stefanello FV, Parker MO, Gerlai R, Rosemberg DB. Understanding the neurobiological effects of drug abuse: Lessons from zebrafish models. Prog Neuropsychopharmacol Biol Psychiatry 2020; 100:109873. [PMID: 31981718 DOI: 10.1016/j.pnpbp.2020.109873] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 01/01/2023]
Abstract
Drug abuse and brain disorders related to drug comsumption are public health problems with harmful individual and social consequences. The identification of therapeutic targets and precise pharmacological treatments to these neuropsychiatric conditions associated with drug abuse are urgently needed. Understanding the link between neurobiological mechanisms and behavior is a key aspect of elucidating drug abuse-related targets. Due to various molecular, biochemical, pharmacological, and physiological features, the zebrafish (Danio rerio) has been considered a suitable vertebrate for modeling complex processes involved in drug abuse responses. In this review, we discuss how the zebrafish has been successfully used for modeling neurobehavioral phenotypes related to drug abuse and review the effects of opioids, cannabinoids, alcohol, nicotine, and psychedelic drugs on the central nervous system (CNS). Moreover, we summarize recent advances in zebrafish-based studies and outline potential advantages and limitations of the existing zebrafish models to explore the neurochemical bases of drug abuse and addiction. Finally, we discuss how the use of zebrafish models may present fruitful approaches to provide valuable clinically translatable data.
Collapse
Affiliation(s)
- Talise E Müller
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.
| | - Barbara D Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Old St Michael's Building, Portsmouth PO1 2DT, UK
| | - Kanandra T Bertoncello
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Francini Franscescon
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Nathana J Mezzomo
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Pharmacology, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Julia Canzian
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Flavia V Stefanello
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Matthew O Parker
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Old St Michael's Building, Portsmouth PO1 2DT, UK
| | - Robert Gerlai
- Department of Psychology, University of Toronto, Mississauga, Canada; Department of Cell and Systems Biology, University of Toronto, Canada
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA.
| |
Collapse
|
44
|
Anker JJ, Kushner MG. Co-Occurring Alcohol Use Disorder and Anxiety: Bridging Psychiatric, Psychological, and Neurobiological Perspectives. Alcohol Res 2019; 40:arcr.v40.1.03. [PMID: 31886106 PMCID: PMC6927748 DOI: 10.35946/arcr.v40.1.03] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A substantial number of people who have problems with alcohol also experience strong anxiety and mood problems. This article provides an overview of the evolving perspectives of this association in the context of three related disciplines—psychiatry, psychology, and neuroscience. Psychiatric and epidemiological studies show that having either an anxiety- or alcohol-related diagnosis elevates the prospective risk for developing the other disorder. From the psychological perspective, behavioral research demonstrates that drinking to cope with negative affect is a potent marker for current and future problems with alcohol. Neuroscientific research implicates overlapping neurobiological systems and psychological processes in promoting the rise of negative affect and alcohol misuse. The psychiatric perspective that alcohol misuse and co-occurring anxiety represent neurobiologically distinct diagnostic conditions has dominated the field for many decades. However, recent research provides increasing support for the neuroscientific perspective that these conditions share underlying, mutually exacerbating, neurobiological processes.
Collapse
Affiliation(s)
- Justin J Anker
- Justin J. Anker, Ph.D., is an assistant professor in the Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota
| | - Matt G Kushner
- Matt G. Kushner, Ph.D., is a professor in the Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
45
|
Acute prazosin administration does not reduce stressor reactivity in healthy adults. Psychopharmacology (Berl) 2019; 236:3371-3382. [PMID: 31197436 PMCID: PMC6832815 DOI: 10.1007/s00213-019-05297-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/02/2019] [Indexed: 12/19/2022]
Abstract
RATIONALE Norepinephrine plays a critical role in the stress response. Clarifying the psychopharmacological effects of norepinephrine manipulation on stress reactivity in humans has important implications for basic neuroscience and treatment of stress-related psychiatric disorders, such as posttraumatic stress disorder and alcohol use disorders. Preclinical research implicates the norepinephrine alpha-1 receptor in responses to stressors. The No Shock, Predictable Shock, Unpredictable Shock (NPU) task is a human laboratory paradigm that is well positioned to test cross-species neurobiological stress mechanisms and advance experimental therapeutic approaches to clinical trials testing novel treatments for psychiatric disorders. OBJECTIVES We hypothesized that acute administration of prazosin, a noradrenergic alpha-1 antagonist, would have a larger effect on reducing stress reactivity during unpredictable, compared to predictable, stressors in the NPU task. METHODS We conducted a double-blind, placebo-controlled, crossover randomized controlled trial in which 64 healthy adults (32 female) completed the NPU task at two visits (2 mg prazosin vs. placebo). RESULTS A single acute dose of 2 mg prazosin did not reduce stress reactivity in a healthy adult sample. Neither NPU startle potentiation nor self-reported anxiety was reduced by prazosin (vs. placebo) during unpredictable (vs. predictable) stressors. CONCLUSIONS Further research is needed to determine whether this failure to translate preclinical neuroscience to human laboratory models is due to methodological factors (e.g., acute vs. chronic drug administration, brain penetration, study population) and/or suggests limited clinical utility of noradrenergic alpha-1 antagonists for treating stress-related psychiatric disorders.
Collapse
|
46
|
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
|
47
|
Witkiewitz K, Litten RZ, Leggio L. Advances in the science and treatment of alcohol use disorder. SCIENCE ADVANCES 2019; 5:eaax4043. [PMID: 31579824 PMCID: PMC6760932 DOI: 10.1126/sciadv.aax4043] [Citation(s) in RCA: 310] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/28/2019] [Indexed: 05/03/2023]
Abstract
Alcohol is a major contributor to global disease and a leading cause of preventable death, causing approximately 88,000 deaths annually in the United States alone. Alcohol use disorder is one of the most common psychiatric disorders, with nearly one-third of U.S. adults experiencing alcohol use disorder at some point during their lives. Alcohol use disorder also has economic consequences, costing the United States at least $249 billion annually. Current pharmaceutical and behavioral treatments may assist patients in reducing alcohol use or facilitating alcohol abstinence. Although recent research has expanded understanding of alcohol use disorder, more research is needed to identify the neurobiological, genetic and epigenetic, psychological, social, and environmental factors most critical in the etiology and treatment of this disease. Implementation of this knowledge in clinical practice and training of health care providers is also needed to ensure appropriate diagnosis and treatment of individuals suffering from alcohol use disorder.
Collapse
Affiliation(s)
- K. Witkiewitz
- Department of Psychology and Center on Alcoholism, Substance Abuse, and Addictions, University of New Mexico, 2650 Yale Blvd. SE, Albuquerque, NM 87106, USA
| | - R. Z. Litten
- Division of Medications Development and Division of Treatment and Recovery Research, National Institute on Alcohol Abuse and Alcoholism, 6700B Rockledge Drive, Bethesda, MD 20892-6902, USA
| | - L. Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, and National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 10 Center Drive (10CRC/15330), Bethesda, MD 21224, USA
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd., Baltimore, MD 21224, USA
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI 02912, USA
- Corresponding author.
| |
Collapse
|
48
|
Increased Ethanol Consumption and Locomotion Develop upon Ethanol Deprivation in Rats Overexpressing the Adenosine (A) 2A Receptor. Neuroscience 2019; 418:133-148. [PMID: 31449988 DOI: 10.1016/j.neuroscience.2019.08.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 08/04/2019] [Accepted: 08/17/2019] [Indexed: 02/03/2023]
Abstract
Preclinical data indicate that ethanol produces behavioral effects that can be regulated by many neurotransmitters and neuromodulators like adenosine (A). The most important receptors with respect to the rewarding effects of ethanol seem to be the A2A receptors. This study used a transgenic strategy, specifically rats overexpressing the A2A receptor, to characterize the neurobiological mechanisms of ethanol consumption as measured by intermittent access to 20% ethanol in a two-bottle choice paradigm. In this model, no change in ethanol consumption was observed in transgenic animals compared to wild type controls during the acquisition/maintenance phase. Following alcohol deprivation, only transgenic rats overexpressing the A2A receptor exhibited escalation of ethanol consumption and drank more (by ca. 90%), but not significantly, ethanol than did the wild type rats. During ethanol withdrawal, the immobility time of rats overexpressing the A2A receptor in the forced swim test was lower than that of wild type rats. Moreover, transgenic rats withdrawn from ethanol, compared to the drug-naive transgenic animals, exhibited an increase above 70% in locomotion. The results indicated that the overexpression of A2A receptors may be a risk factor for the escalation of ethanol consumption despite the reduction in depression-like signs of ethanol withdrawal.
Collapse
|
49
|
Leggio L, Falk DE, Ryan ML, Fertig J, Litten RZ. Medication Development for Alcohol Use Disorder: A Focus on Clinical Studies. Handb Exp Pharmacol 2019; 258:443-462. [PMID: 31628604 DOI: 10.1007/164_2019_295] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Compared to other medical disorders, including other brain diseases, the number of medications approved for alcohol use disorder (AUD) is very small. Disulfiram, naltrexone (oral and long-acting), and acamprosate are approved by the US Food and Drug Administration (FDA) to treat patients with AUD. These medications are also approved in other countries, including in Europe, where the European Medicines Agency (EMA) also approved nalmefene for AUD. Furthermore, baclofen was recently approved for AUD in France. These approved medications have small effect sizes, which are probably the consequence of the fact that they only work for some patients, yet a personalized approach to match the right medication with the right patient is still in its infancy. Therefore, research is needed to expand the armamentarium of medications that clinicians can use to treat their patients, as well as to better develop personalized approaches. This book chapter reviews other medications, beyond those approved by the FDA, that have shown efficacy in clinical trials, as well as medications which are still in the early stages of evaluation in human studies.
Collapse
Affiliation(s)
- Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, Bethesda, MD, USA. .,Medication Development Program, National Institute on Drug Abuse Intramural Research Program , Baltimore, MD, USA. .,Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA.
| | - Daniel E Falk
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Megan L Ryan
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Joanne Fertig
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Raye Z Litten
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| |
Collapse
|
50
|
Phosphoproteomic Analysis of the Amygdala Response to Adolescent Glucocorticoid Exposure Reveals G-Protein Coupled Receptor Kinase 2 as a Target for Reducing Motivation for Alcohol. Proteomes 2018; 6:proteomes6040041. [PMID: 30322021 PMCID: PMC6313880 DOI: 10.3390/proteomes6040041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/28/2018] [Accepted: 10/09/2018] [Indexed: 01/19/2023] Open
Abstract
Early life stress is associated with risk for developing alcohol use disorders (AUDs) in adulthood. Though the neurobiological mechanisms underlying this vulnerability are not well understood, evidence suggests that aberrant glucocorticoid and noradrenergic system functioning play a role. The present study investigated the long-term consequences of chronic exposure to elevated glucocorticoids during adolescence on the risk of increased alcohol-motivated behavior, and on amygdalar function in adulthood. A discovery-based analysis of the amygdalar phosphoproteome using mass spectrometry was employed, to identify changes in function. Adolescent corticosterone (CORT) exposure increased alcohol, but not sucrose, self-administration, and enhanced stress-induced reinstatement with yohimbine in adulthood. Phosphoproteomic analysis indicated that the amygdala phosphoproteome was significantly altered by adolescent CORT exposure, generating a list of potential novel mechanisms involved in the risk of alcohol drinking. In particular, increased phosphorylation at serines 296–299 on the α2A adrenergic receptor (α2AAR), mediated by the G-protein coupled receptor kinase 2 (GRK2), was evident after adolescent CORT exposure. We found that intra-amygdala infusion of a peptidergic GRK2 inhibitor reduced alcohol seeking, as measured by progressive ratio and stress reinstatement tests, and induced by the α2AAR antagonist yohimbine. These results suggest that GRK2 represents a novel target for treating stress-induced motivation for alcohol which may counteract alterations in brain function induced by adolescent stress exposure.
Collapse
|