1
|
Quelch D, Lingford-Hughes A, John B, Nutt D, Bradberry S, Roderique-Davies G. Promising strategies for the prevention of alcohol-related brain damage through optimised management of acute alcohol withdrawal: A focussed literature review. J Psychopharmacol 2024:2698811241294005. [PMID: 39529219 DOI: 10.1177/02698811241294005] [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] [Indexed: 11/16/2024]
Abstract
There is an increasing awareness of the link between chronic alcohol consumption and the development of cognitive, behavioural and functional deficits. Currently, preventative strategies are limited and require engagement in dedicated long-term rehabilitation and sobriety services, the availability of which is low. The acute alcohol withdrawal syndrome is an episode of neurochemical imbalance leading to autonomic dysregulation, increased seizure risk and cognitive disorientation. In addition to harm from symptoms of alcohol withdrawal (e.g. seizures), the underpinning neurochemical changes may also lead to cytotoxicity through various cellular mechanisms, which long-term, may translate to some of the cognitive impairments observed in Alcohol-Related Brain Damage (ARBD). Here we review some of the pharmacological and neurochemical mechanisms underpinning alcohol withdrawal. We discuss the cellular and pharmacological basis of various potential neuroprotective strategies that warrant further exploration in clinical populations with a view to preventing the development of ARBD. Such strategies, when integrated into the clinical management of acute alcohol withdrawal, may impact large populations of individuals, who currently face limited dedicated service delivery and healthcare resource.
Collapse
Affiliation(s)
- Darren Quelch
- Addictions Research Group, Applied Psychology Research and Innovation Group, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, UK
- Alcohol Care Team and Clinical Toxicology Service, Sandwell and West-Birmingham NHS Trust, City Hospital, Birmingham, UK
| | - Anne Lingford-Hughes
- Centre for Neuropsychopharmacology, Division of Psychiatry, Imperial College London, Hammersmith Hospital, London, UK
| | - Bev John
- Addictions Research Group, Applied Psychology Research and Innovation Group, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, UK
| | - David Nutt
- Centre for Neuropsychopharmacology, Division of Psychiatry, Imperial College London, Hammersmith Hospital, London, UK
| | - Sally Bradberry
- Addictions Research Group, Applied Psychology Research and Innovation Group, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, UK
- Alcohol Care Team and Clinical Toxicology Service, Sandwell and West-Birmingham NHS Trust, City Hospital, Birmingham, UK
| | - Gareth Roderique-Davies
- Addictions Research Group, Applied Psychology Research and Innovation Group, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, UK
| |
Collapse
|
2
|
Rodella P, Boreski D, Luz MAM, Gabriel EA, Takase LF, Chin CM. Taurine Neuroprotection and Neurogenesis Effect in Chronic Ethanol-Induced Rats. Nutrients 2024; 16:1973. [PMID: 38931326 PMCID: PMC11206532 DOI: 10.3390/nu16121973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Taurine (2-aminoethanesulfonic acid) is a non-protein β-amino acid essential for cellular homeostasis, with antioxidant, anti-inflammatory, and cytoprotective properties that are crucial for life maintenance. This study aimed to evaluate the effects of taurine administration on hippocampal neurogenesis, neuronal preservation, or reverse damage in rats exposed to forced ethanol consumption in an animal model. Wistar rats were treated with ethanol (EtOH) for a 28-day period (5% in the 1st week, 10% in the 2nd week, and 20% in the 3rd and 4th weeks). Two taurine treatment protocols (300 mg/kg i.p.) were implemented: one during ethanol consumption to analyze neuroprotection, and another after ethanol consumption to assess the reversal of ethanol-induced damage. Overall, the results demonstrated that taurine treatment was effective in protecting against deficits induced by ethanol consumption in the dentate gyrus. The EtOH+TAU group showed a significant increase in cell proliferation (145.8%) and cell survival (54.0%) compared to the EtOH+Sal group. The results also indicated similar effects regarding the reversal of ethanol-induced damage 28 days after the cessation of ethanol consumption. The EtOH+TAU group exhibited a significant increase (41.3%) in the number of DCX-immunoreactive cells compared to the EtOH+Sal group. However, this amino acid did not induce neurogenesis in the tissues of healthy rats, implying that its activity may be contingent upon post-injury stimuli.
Collapse
Affiliation(s)
- Patricia Rodella
- Laboratory for Drug Design (LAPDESF), School of Pharmaceutical Sciences, University of São Paulo State (UNESP), Araraquara 14800-903, Brazil; (P.R.); (D.B.)
| | - Diogo Boreski
- Laboratory for Drug Design (LAPDESF), School of Pharmaceutical Sciences, University of São Paulo State (UNESP), Araraquara 14800-903, Brazil; (P.R.); (D.B.)
| | - Marcus Alexandre Mendes Luz
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, Brazil; (M.A.M.L.); (E.A.G.)
| | - Edmo Atique Gabriel
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, Brazil; (M.A.M.L.); (E.A.G.)
| | - Luiz Fernando Takase
- Morphology and Pathology Department, Federal University of São Paulo of São Carlos (UFSCar), São Carlos 13565-905, Brazil;
| | - Chung Man Chin
- Laboratory for Drug Design (LAPDESF), School of Pharmaceutical Sciences, University of São Paulo State (UNESP), Araraquara 14800-903, Brazil; (P.R.); (D.B.)
- Advanced Research Center in Medicine (CEPAM), School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), Sao Jose do Rio Preto 15030-070, Brazil; (M.A.M.L.); (E.A.G.)
| |
Collapse
|
3
|
Cummins-Beebee PN, Chvilicek MM, Rothenfluh A. The Stage-Based Model of Addiction-Using Drosophila to Investigate Alcohol and Psychostimulant Responses. Int J Mol Sci 2023; 24:10909. [PMID: 37446084 PMCID: PMC10341944 DOI: 10.3390/ijms241310909] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Addiction is a progressive and complex disease that encompasses a wide range of disorders and symptoms, including substance use disorder (SUD), for which there are few therapeutic treatments. SUD is the uncontrolled and chronic use of substances despite the negative consequences resulting from this use. The progressive nature of addiction is organized into a testable framework, the neurobiological stage-based model, that includes three behavioral stages: (1) binge/intoxication, (2) withdrawal/negative affect, and (3) preoccupation/anticipation. Human studies offer limited opportunities for mechanistic insights into these; therefore, model organisms, like Drosophila melanogaster, are necessary for understanding SUD. Drosophila is a powerful model organism that displays a variety of SUD-like behaviors consistent with human and mammalian substance use, making flies a great candidate to study mechanisms of behavior. Additionally, there are an abundance of genetic tools like the GAL4/UAS and CRISPR/Cas9 systems that can be used to gain insight into the molecular mechanisms underlying the endophenotypes of the three-stage model. This review uses the three-stage framework and discusses how easily testable endophenotypes have been examined with experiments using Drosophila, and it outlines their potential for investigating other endophenotypes.
Collapse
Affiliation(s)
- Pearl N. Cummins-Beebee
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Maggie M. Chvilicek
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Adrian Rothenfluh
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
4
|
Swinford-Jackson SE, O'Brien CP, Kenny PJ, Vanderschuren LJMJ, Unterwald EM, Pierce RC. The Persistent Challenge of Developing Addiction Pharmacotherapies. Cold Spring Harb Perspect Med 2021; 11:a040311. [PMID: 32601131 PMCID: PMC8559539 DOI: 10.1101/cshperspect.a040311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
There are currently effective Food and Drug Administration (FDA)-approved therapies for alcohol, nicotine, and opioid use disorders. This article will review the development of eight compounds used in the treatment of drug addiction with an emphasis on pharmacological mechanisms and the utility of preclinical animal models of addiction in therapeutic development. In contrast to these successes, animal research has identified a number of promising medications for the treatment of psychostimulant use disorder, none of which have proven to be clinically effective. A specific example of an apparently promising pharmacotherapeutic for cocaine that failed clinically will be examined to determine whether this truly represents a challenge to the predictive validity of current models of cocaine addiction. In addition, the development of promising cocaine use disorder therapeutics derived from animal research will be reviewed, with some discussion regarding how preclinical studies might be modified to better inform clinical outcomes.
Collapse
Affiliation(s)
- Sarah E Swinford-Jackson
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Charles P O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Paul J Kenny
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Louk J M J Vanderschuren
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - Ellen M Unterwald
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, USA
| | - R Christopher Pierce
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
5
|
Yang D, Zhou Q, Labroska V, Qin S, Darbalaei S, Wu Y, Yuliantie E, Xie L, Tao H, Cheng J, Liu Q, Zhao S, Shui W, Jiang Y, Wang MW. G protein-coupled receptors: structure- and function-based drug discovery. Signal Transduct Target Ther 2021; 6:7. [PMID: 33414387 PMCID: PMC7790836 DOI: 10.1038/s41392-020-00435-w] [Citation(s) in RCA: 280] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 02/08/2023] Open
Abstract
As one of the most successful therapeutic target families, G protein-coupled receptors (GPCRs) have experienced a transformation from random ligand screening to knowledge-driven drug design. We are eye-witnessing tremendous progresses made recently in the understanding of their structure-function relationships that facilitated drug development at an unprecedented pace. This article intends to provide a comprehensive overview of this important field to a broader readership that shares some common interests in drug discovery.
Collapse
Affiliation(s)
- Dehua Yang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Qingtong Zhou
- School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
| | - Viktorija Labroska
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Shanshan Qin
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Sanaz Darbalaei
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yiran Wu
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Elita Yuliantie
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Linshan Xie
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Houchao Tao
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Qing Liu
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Wenqing Shui
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| | - Yi Jiang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.
| | - Ming-Wei Wang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China. .,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China. .,School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China. .,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China. .,School of Pharmacy, Fudan University, 201203, Shanghai, China.
| |
Collapse
|
6
|
Demery-Poulos C, Chambers JM. Genetic variation in alcoholism and opioid addiction susceptibility and treatment: a pharmacogenomic approach. AIMS MOLECULAR SCIENCE 2021. [DOI: 10.3934/molsci.2021016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>Alcohol and opioid abuse have pervasive and detrimental consequences from the individual to societal level. The extent of genetic contribution to alcoholism has been studied for decades, yielding speculative and often inconsistent results since the previous discovery of two pharmacokinetic variants strongly protective against alcoholism. The neurobiology of addiction involves innumerate genes with combinatorial and epistatic interactions, creating a difficult landscape for concrete conclusions. In contrast, pharmacogenomic variation in the treatment of alcoholism yields more immediate clinical utility, while also emphasizing pathways crucial to the progression of addiction. An improved understanding of genetic predisposition to alcohol abuse has inherent significance for opioid addiction and treatment, as the two drugs induce the same reward pathway. This review outlines current knowledge, treatments, and research regarding genetic predisposition to alcoholism, focusing on pharmacodynamic variation within the dopaminergic system and shared implications for opioid abuse. Multifaceted and highly polygenic, the phenotype of addiction seems to grow more complex as new research extends the scope of its impact on the brain, body, and progeny.</p>
</abstract>
Collapse
|
7
|
Volkow ND, Blanco C. The changing opioid crisis: development, challenges and opportunities. Mol Psychiatry 2021; 26:218-233. [PMID: 32020048 PMCID: PMC7398847 DOI: 10.1038/s41380-020-0661-4] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 01/02/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022]
Abstract
The current opioid epidemic is one of the most severe public health crisis in US history. Responding to it has been difficult due to its rapidly changing nature and the severity of its associated outcomes. This review examines the origin and evolution of the crisis, the pharmacological properties of opioids, the neurobiology of opioid use and opioid use disorder (OUD), medications for opioid use disorder (MOUD), and existing and promising approaches to prevention. The results of the review indicate that the opioid epidemic is a complex, evolving phenomenon that involves neurobiological vulnerabilities and social determinants of health. Successfully addressing the epidemic will require advances in basic science, development of more acceptable and effective treatments, and implementation of public health approaches, including prevention. The advances achieved in addressing the current crisis should also serve to advance the science and treatment of other substance use disorders.
Collapse
Affiliation(s)
| | - Carlos Blanco
- National Institute on Drug Abuse, Bethesda, MD, 20892, USA.
| |
Collapse
|
8
|
Kokane SS, Perrotti LI. Sex Differences and the Role of Estradiol in Mesolimbic Reward Circuits and Vulnerability to Cocaine and Opiate Addiction. Front Behav Neurosci 2020; 14:74. [PMID: 32508605 PMCID: PMC7251038 DOI: 10.3389/fnbeh.2020.00074] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
Although both men and women become addicted to drugs of abuse, women transition to addiction faster, experience greater difficulties remaining abstinent, and relapse more often than men. In both humans and rodents, hormonal cycles are associated with females' faster progression to addiction. Higher concentrations and fluctuating levels of ovarian hormones in females modulate the mesolimbic reward system and influence reward-directed behavior. For example, in female rodents, estradiol (E2) influences dopamine activity within the mesolimbic reward system such that drug-directed behaviors that are normally rewarding and reinforcing become enhanced when circulating levels of E2 are high. Therefore, neuroendocrine interactions, in part, explain sex differences in behaviors motivated by drug reward. Here, we review sex differences in the physiology and function of the mesolimbic reward system in order to explore the notion that sex differences in response to drugs of abuse, specifically cocaine and opiates, are the result of molecular neuroadaptations that differentially develop depending upon the hormonal state of the animal. We also reconsider the notion that ovarian hormones, specifically estrogen/estradiol, sensitize target neurons thereby increasing responsivity when under the influence of either cocaine or opiates or in response to exposure to drug-associated cues. These adaptations may ultimately serve to guide the motivational behaviors that underlie the factors that cause women to be more vulnerable to cocaine and opiate addiction than men.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Psychology, University of Texas at Arlington, Arlington, TX, United States
| | - Linda I Perrotti
- Department of Psychology, University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
9
|
Kokane SS, Armant RJ, Bolaños-Guzmán CA, Perrotti LI. Overlap in the neural circuitry and molecular mechanisms underlying ketamine abuse and its use as an antidepressant. Behav Brain Res 2020; 384:112548. [PMID: 32061748 PMCID: PMC7469509 DOI: 10.1016/j.bbr.2020.112548] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/18/2020] [Accepted: 02/07/2020] [Indexed: 02/09/2023]
Abstract
Ketamine, a dissociative anesthetic and psychedelic compound, has revolutionized the field of psychopharmacology by showing robust, and rapid-acting antidepressant activity in patients suffering from major depressive disorder (MDD), suicidal tendencies, and treatment-resistant depression (TRD). Ketamine's efficacy, however, is transient, and patients must return to the clinic for repeated treatment as they experience relapse. This is cause for concern because ketamine is known for its abuse liability, and repeated exposure to drugs of abuse often leads to drug abuse/dependence. Though the mechanism(s) underlying its antidepressant activity is an area of current intense research, both clinical and preclinical evidence shows that ketamine's effects are mediated, at least in part, by molecular adaptations resulting in long-lasting synaptic changes in mesolimbic brain regions known to regulate natural and drug reward. This review outlines our limited knowledge of ketamine's neurobiological and biochemical underpinnings mediating its antidepressant effects and correlates them to its abuse potential. Depression and addiction share overlapping neural circuitry and molecular mechanisms, and though speculative, repeated use of ketamine for the treatment of depression could lead to the development of substance use disorder/addiction, and thus should be tempered with caution. There is much that remains to be known about the long-term effects of ketamine, and our lack of understanding of neurobiological mechanisms underlying its antidepressant effects is a clear limiting factor that needs to be addressed systematically before using repeated ketamine in the treatment of depressed patients.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Psychology, The University of Texas at Arlington, United States
| | - Ross J Armant
- Department of Psychology, The University of Texas at Arlington, United States
| | - Carlos A Bolaños-Guzmán
- Department of Psychological and Brain Sciences, Institute for Neuroscience, Texas A&M University, College Station, TX 77840, United States
| | - Linda I Perrotti
- Department of Psychology, The University of Texas at Arlington, United States.
| |
Collapse
|
10
|
Robins MT, Heinricher MM, Ryabinin AE. From Pleasure to Pain, and Back Again: The Intricate Relationship Between Alcohol and Nociception. Alcohol Alcohol 2020; 54:625-638. [PMID: 31509854 DOI: 10.1093/alcalc/agz067] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
AIMS A close and bidirectional relationship between alcohol consumption and pain has been previously reported and discussed in influential reviews. The goal of the present narrative review is to provide an update on the developments in this field in order to guide future research objectives. METHODS We evaluated both epidemiological and neurobiological literature interrogating the relationship between alcohol use and pain for the presence of significant effects. We outlined studies on interactions between alcohol use and pain using both self-reports and objective experimental measures and discussed potential underlying mechanisms of these interactions. RESULTS Epidemiological, preclinical and clinical literature point to three major interactions between alcohol use and pain: (a) alcohol use leading to hyperalgesia, (b) alcohol use moderating pain and hyperalgesia and (c) chronic pain as a risk factor predisposing to alcohol relapse. Neurobiological studies using animal models to assess these interactions have transitioned from mostly involuntary modes of experimenter-controlled alcohol administration to self-administration procedures, and increasingly indicate that neuronal circuits implicated in both withdrawal and anticipation stages of alcohol use disorder also have a role in chronic pain. Mechanistically, alterations in GABA, glutamate, the corticotropin-releasing factor system, endogenous opioids and protein kinase C appear to play crucial roles in this maladaptive overlap. CONCLUSIONS Many of the principles explaining the interactions between alcohol and pain remain on a strong foundation, but continuing progress in modeling these interactions and underlying systems will provide a clearer basis for understanding, and ultimately treating, the damaging aspects of this interaction.
Collapse
Affiliation(s)
- Meridith T Robins
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Mary M Heinricher
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.,Department of Neurological Surgery, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| |
Collapse
|
11
|
Abstract
Despite a number of approved medications for alcohol use disorder (AUD), this chronic relapsing disease still produces a considerable global burden, with both health-related and financial consequences. While clinical trials are a critical step in drug development, human laboratory studies provide the field with means of screening pharmacotherapy for more nuanced aspects of AUD. Specifically, studies employing alcohol administration techniques (e.g., alcohol challenge and self-administration) are able to investigate potential drugs with respect to their ability to alter various responses to alcohol administration or alter alcohol consumption in laboratory settings. This chapter reviews methodological designs and provides updates from alcohol administration studies used to screen for potential AUD pharmacotherapy over the past decade. These recent studies have supported the efficacy of approved drugs, identified some promising novel drugs, and investigated other drugs that appear ineffective in AUD treatment. Yet, few drugs are explored using the different variants of alcohol administration methods, and using the different methods has provided inconsistent results for the same drug. Future research would aid advancement in the field by testing medication with various methodologies and refining recently developed techniques.
Collapse
|
12
|
Leggio L, Chick J. From ‘The Red Journal’, for the Retirement of Editor-in-Chief Philippe De Witte: A Fond and Grateful Farewell. Alcohol Alcohol 2020; 55:1-2. [DOI: 10.1093/alcalc/agaa008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
13
|
Hong SI, Peyton L, Chern Y, Choi DS. Novel Adenosine Analog, N6-(4-Hydroxybenzyl)-Adenosine, Dampens Alcohol Drinking and Seeking Behaviors. J Pharmacol Exp Ther 2019; 371:260-267. [PMID: 31409667 DOI: 10.1124/jpet.119.261529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
Adenosine signaling is associated with ethanol-related behaviors. We previously found that adenosine A2A receptor (A2AR) activation dampens ethanol drinking behaviors in equilibrative nucleoside transporter 1 (ENT1) knockout mice, and A2AR inhibition augments reward-seeking behavior in wild-type mice. The novel adenosine analog N6-(4-hydroxybenzyl)-adenosine (NHBA), which is isolated from the rhizomes of Gastrodia elata, activates A2AR and inhibits ENT1. Here, we examined the effects of NHBA on ethanol drinking in the two-bottle choice test and operant ethanol seeking behaviors. We selected mice exhibiting high ethanol drinking behavior in the two-bottle choice test. NHBA (0.1 mg/kg, i.p.) reduced ethanol drinking behavior in a limited-access 3-hour drinking session in high-consumption ethanol drinking mice, and NHBA (0.1 mg/kg, i.p.) did not alter locomotor activity in the open-field test. Operant conditioning with 10% ethanol and 10% sucrose (10E10S) reward increased zone entries and time spent in the ethanol zone, while NHBA (0.1 mg/kg, i.p.) dampened ethanol zone preference in the Y-maze. Furthermore, NHBA (0.1 mg/kg, i.p.) devalued 10E10S and 10% ethanol (10E) reward after operant conditioning with 10E10S and 10E. Taken together, NHBA through A2AR activation and ENT1 modulation may dampen ethanol drinking and seeking behaviors, suggesting that NHBA is a potential therapeutic agent for treating alcohol use disorder. SIGNIFICANCE STATEMENT: Our work highlights that A2AR activation and ENT1 inhibition by a novel adenosine analog isolated from Gastrodia elata, N6-(4-hydroxybenzyl)-adenosine, decreases ethanol drinking and seeking behaviors. We suggest that NHBA is a potential therapeutic agent to treat alcohol use disorder.
Collapse
Affiliation(s)
- Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Yijuang Chern
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics (S.-I.H., L.P., D.-S.C.), Neuroscience Program (D.-S.C.), and Department of Psychiatry and Psychology (D.-S.C.), Mayo Clinic College of Medicine, Rochester, Minnesota; and Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.C.)
| |
Collapse
|
14
|
Lee G, Zhou Y. NMDAR Hypofunction Animal Models of Schizophrenia. Front Mol Neurosci 2019; 12:185. [PMID: 31417356 PMCID: PMC6685005 DOI: 10.3389/fnmol.2019.00185] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) hypofunction hypothesis has been proposed to help understand the etiology and pathophysiology of schizophrenia. This hypothesis was based on early observations that NMDAR antagonists could induce a full range of symptoms of schizophrenia in normal human subjects. Accumulating evidence in humans and animal studies points to NMDAR hypofunctionality as a convergence point for various symptoms of schizophrenia. Here we review animal models of NMDAR hypofunction generated by pharmacological and genetic approaches, and how they relate to the pathophysiology of schizophrenia. In addition, we discuss the limitations of animal models of NMDAR hypofunction and their potential utility for therapeutic applications.
Collapse
Affiliation(s)
| | - Yi Zhou
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
15
|
Pezzilli R, Caputo F, Testino G, Patussi V, Greco G, Macciò L, Rossin MR, Mioni D, Balbinot P, Gandin C, Zanesini F, Frulloni L, Aricò S, Bottaro LC, Pellicano R, Scafato E. Alcohol-related chronic exocrine pancreatic insufficiency: diagnosis and therapeutic management. A proposal for treatment by the Italian Association for the Study of the Pancreas (AISP) and the Italian Society of Alcohology (SIA). Minerva Med 2019; 110:425-438. [PMID: 30938130 DOI: 10.23736/s0026-4806.19.06043-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Current estimates of the prevalence of chronic pancreatitis, one of the most common causes of exocrine pancreatic insufficiency, are in the range of 3-10 per 100,000 people in many parts of the world. Alcohol consumption is a very important risk factor for exocrine pancreatic insufficiency and is involved in nearly half of all cases. The main hypothesis regarding the role of chronic alcohol consumption in pancreatitis is that there must be additional environmental or genetic risk factors involved for ongoing damage to occur. Treatment of patients with alcohol-related exocrine pancreatic insufficiency is complex, as the patient has two concomitant pathologies, alcohol-use disorder (AUD) and exocrine pancreatic insufficiency/chronic pancreatitis. Alcohol abstinence is the starting point for treatment, although even this along with the most advanced therapies allow only a slowdown in progression rather than restoration of function. This position paper of the Italian Association for the Study of the Pancreas and the Italian Society of Alcohology provides an overview of the pathogenesis of alcohol-related pancreatitis and discuss diagnostic issues. Treatment options for both exocrine pancreatic insufficiency/chronic pancreatitis (with a focus on pancreatic enzyme replacement therapy) and AUD (acamprosate, disulfiram, oral naltrexone, long-acting injectable naltrexone, sodium oxybate, nalmefene, baclofen, and psychosocial interventions) are also reviewed.
Collapse
Affiliation(s)
- Raffaele Pezzilli
- Pancreas Unit, Department of Gastroenterology, Polyclinic of Sant'Orsola, Bologna, Italy - .,Italian Association for the Study of the Pancreas, Rome, Italy -
| | - Fabio Caputo
- Department of Internal Medicine, SS. Annunziata Hospital, Cento, Ferrara, Italy.,G. Fontana Center for the Study and Multidisciplinary Treatment of Alcohol Addiction, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Gianni Testino
- Regional Alcohol Center of Liguria Region, ASL3, San Martino Hospital, Genoa, Italy
| | | | - Giovanni Greco
- Department of Mental Health and Pathological Dependency, AUSL Romagna, SerT Ravenna, Ravenna, Italy
| | | | | | - Davide Mioni
- Casa di Cura Parco dei Tigli, Villa Di Teolo, Padua, Italy
| | - Patrizia Balbinot
- Regional Alcohol Center of Liguria Region, ASL3, San Martino Hospital, Genoa, Italy
| | - Claudia Gandin
- National Observatory on Alcohol, National Institute of Health, Rome, Italy
| | | | - Luca Frulloni
- Department of Medicine, Pancreas Institute, University of Verona, Verona, Italy
| | - Sarino Aricò
- Unit of Gastroenterology, Mauriziano Hospital, Turin, Italy
| | | | | | - Emanuele Scafato
- National Observatory on Alcohol, National Institute of Health, Rome, Italy
| | | |
Collapse
|
16
|
Affiliation(s)
- Daniel Fuster
- From the Internal Medicine Service, Addiction Unit, Hospital Universitari Germans Trias i Pujol, Universitat Autonoma de Barcelona, Badalona, Spain (D.F.); and the Clinical Addiction Research and Education Unit, Section of General Internal Medicine, Department of Medicine, Boston Medical Center and Boston University School of Medicine, the Grayken Center for Addiction, Boston Medical Center, and the Department of Community Health Sciences, Boston University School of Public Health - all in Boston ( J.H.S.)
| | - Jeffrey H Samet
- From the Internal Medicine Service, Addiction Unit, Hospital Universitari Germans Trias i Pujol, Universitat Autonoma de Barcelona, Badalona, Spain (D.F.); and the Clinical Addiction Research and Education Unit, Section of General Internal Medicine, Department of Medicine, Boston Medical Center and Boston University School of Medicine, the Grayken Center for Addiction, Boston Medical Center, and the Department of Community Health Sciences, Boston University School of Public Health - all in Boston ( J.H.S.)
| |
Collapse
|
17
|
Acamprosate's ethanol intake-reducing effect is associated with its ability to increase dopamine. Pharmacol Biochem Behav 2018; 175:101-107. [PMID: 30266455 DOI: 10.1016/j.pbb.2018.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/04/2018] [Accepted: 09/25/2018] [Indexed: 12/23/2022]
Abstract
Previous studies indicate that the anti-craving substance acamprosate modulates nucleus accumbens (nAc) dopamine levels via a dopamine-controlling nAc-VTA-nAc neurocircuitry. It was demonstrated that glycine receptors in the nAc are involved both in the dopamine-elevating effect and the ethanol intake-reducing effect of the drug. Here we wanted to explore the interaction of ethanol and acamprosate on nAc dopamine and investigate whether dopaminergic transmission may be related to the ethanol intake-reducing effects. In three separate studies we investigated nAc extracellular dopamine levels by means of in vivo microdialysis after administration of acamprosate and ethanol in 1) naïve rats, 2) rats pre-treated with acamprosate for two days or 3) ethanol medium- and high-preferring rats receiving ten days of acamprosate pre-treatment. In the first two studies, acamprosate elevated dopamine and simultaneously prevented ethanol from further increasing dopamine output. In the third study, long-term acamprosate pre-treatment produced a loss of the ethanol intake-reducing as well as the dopamine-elevating effects of acamprosate, and the dopamine elevating property of ethanol was restored. We suggest that acamprosate may partly substitute for the dopamine-elevating effect of ethanol but once tolerance develops to this effect, the ability to decrease ethanol intake is lost.
Collapse
|
18
|
Pradhan G, Melugin PR, Wu F, Fang HM, Weber R, Kroener S. Calcium chloride mimics the effects of acamprosate on cognitive deficits in chronic alcohol-exposed mice. Psychopharmacology (Berl) 2018; 235:2027-2040. [PMID: 29679288 PMCID: PMC10766324 DOI: 10.1007/s00213-018-4900-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/09/2018] [Indexed: 12/22/2022]
Abstract
RATIONALE Acamprosate (calcium-bis N-acetylhomotaurinate) is the leading medication approved for the maintenance of abstinence, shown to reduce craving and relapse in animal models and human alcoholics. Acamprosate can improve executive functions that are impaired by chronic intermittent ethanol (CIE) exposure. Recent work has suggested that acamprosate's effects on relapse prevention are due to its calcium component, which raises the question whether its pro-cognitive effects are similarly mediated by calcium. OBJECTIVES This study examined the effects of acamprosate on alcohol-induced behavioral deficits and compared them with the effects of the sodium salt version of N-acetylhomotaurinate or calcium chloride, respectively. METHODS We exposed mice to alcohol via three cycles of CIE and measured changes in alcohol consumption in a limited-access paradigm. We then compared the effects of acamprosate and calcium chloride (applied subchronically for 3 days during withdrawal) in a battery of cognitive tasks that have been shown to be affected by chronic alcohol exposure. RESULTS CIE-treated animals showed deficits in attentional set-shifting and deficits in novel object recognition. Alcohol-treated animals showed no impairments in social novelty detection and interaction, or delayed spontaneous alternation. Both acamprosate and calcium chloride ameliorated alcohol-induced cognitive deficits to comparable extents. In contrast, the sodium salt version of N-acetylhomotaurinate did not reverse the cognitive deficits. CONCLUSIONS These results add evidence to the notion that acamprosate produces its anti-relapse effects through its calcium moiety. Our results also suggest that improved regulation of drug intake by acamprosate after withdrawal might at least in part be related to improved cognitive function.
Collapse
Affiliation(s)
- Grishma Pradhan
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd, BSB14, Richardson, TX, 75080, USA
| | - Patrick R Melugin
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd, BSB14, Richardson, TX, 75080, USA
| | - Fei Wu
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd, BSB14, Richardson, TX, 75080, USA
- Institute of Neurobiology, Jining Medical University, Jining, China
| | - Hannah M Fang
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd, BSB14, Richardson, TX, 75080, USA
| | - Rachel Weber
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd, BSB14, Richardson, TX, 75080, USA
| | - Sven Kroener
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd, BSB14, Richardson, TX, 75080, USA.
| |
Collapse
|
19
|
de Guglielmo G, Conlisk DE, Barkley-Levenson AM, Palmer AA, George O. Inhibition of Glyoxalase 1 reduces alcohol self-administration in dependent and nondependent rats. Pharmacol Biochem Behav 2018; 167:36-41. [PMID: 29505808 PMCID: PMC5866249 DOI: 10.1016/j.pbb.2018.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/01/2018] [Accepted: 03/01/2018] [Indexed: 01/12/2023]
Abstract
Previous studies showed that the glyoxalase 1 (Glo1) gene modulates anxiety-like behavior, seizure susceptibility, depression-like behavior, and alcohol drinking in the drinking-in-the-dark paradigm in nondependent mice. Administration of the small-molecule GLO1 inhibitor S-bromobenzylglutathione cyclopentyl diester (pBBG) decreased alcohol drinking in nondependent mice, suggesting a possible therapeutic strategy. However, the preclinical therapeutic efficacy of pBBG in animal models of alcohol dependence remains to be demonstrated. We tested the effect of pBBG (7.5 and 25 mg/kg) on operant alcohol self-administration in alcohol-dependent and nondependent rats. Wistar rats were trained to self-administer 10% alcohol (v/v) and made dependent by chronic intermittent passive exposure to alcohol vapor for 5 weeks. Pretreatment with pBBG dose-dependently reduced alcohol self-administration in both nondependent and dependent animals, without affecting water self-administration. pBBG treatment was more effective in dependent rats than in nondependent rats. These data extend previous findings that implicated Glo1 in alcohol drinking in nondependent mice by showing even more profound effects in alcohol-dependent rats. These results suggest that the pharmacological inhibition of GLO1 is a relevant therapeutic target for the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Giordano de Guglielmo
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Dana E Conlisk
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Olivier George
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
20
|
Lee KM, Coelho MA, Class MA, Szumlinski KK. mGlu5-dependent modulation of anxiety during early withdrawal from binge-drinking in adult and adolescent male mice. Drug Alcohol Depend 2018; 184:1-11. [PMID: 29324247 PMCID: PMC6371787 DOI: 10.1016/j.drugalcdep.2017.10.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/26/2017] [Accepted: 10/29/2017] [Indexed: 10/18/2022]
Abstract
Binge alcohol-drinking elicits symptoms of negative affect such as anxiety upon cessation, which is a source of negative reinforcement for perpetuating this pattern of alcohol abuse. Binge-induced anxiety during early (24 h) withdrawal is associated with increased expression of metabotropic glutamate receptor 5 (mGlu5) within the nucleus accumbens shell (AcbSh) of adult male mice, but was unchanged in anxiety-resilient adolescents. Herein, we determined the role of mGlu5 signaling in withdrawal-induced anxiety via pharmacological manipulation using the mGlu5 negative allosteric modulator MTEP and the positive allosteric modulator CDPPB. Adult (PND 56) and adolescent (PND 28) male C57BL/6J mice binge-drank for 14 days under 3-bottle-choice procedures for 2 h/day; control animals drank water only. Approximately 24 h following the final alcohol presentation, animals were treated with 30 mg/kg IP MTEP, CDPPB, or vehicle and then tested, thirty minutes later, for behavioral signs of anxiety. Vehicle-treated binge-drinking adults exhibited hyperanxiety in all paradigms, while vehicle-treated binge-drinking adolescents did not exhibit withdrawal-induced anxiety. In adults, 30 mg/kg MTEP decreased alcohol-induced anxiety across paradigms, while 3 mg/kg MTEP was anxiolytic in adult water controls. CDPPB was modestly anxiogenic in both alcohol- and water-drinking mice. Adolescent animals showed minimal response to either CDPPB or MTEP, suggesting that anxiety in adolescence may be mGlu5-independent. These results demonstrate a causal role for mGlu5 in withdrawal-induced anxiety in adults and suggest age-related differences in the behavioral pharmacology of the negative reinforcing properties of alcohol.
Collapse
Affiliation(s)
- Kaziya M. Lee
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, 93106-9660, USA
| | - Michal A. Coelho
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, 93106-9660, USA
| | - MacKayla A. Class
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, 93106-9660, USA,Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, 93106-9625, USA
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, 93106-9660, USA,Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, 93106-9625, USA,Corresponding author at: University of California Santa Barbara, Santa Barbara, CA, 93106-9660, USA. (K.K. Szumlinski)
| |
Collapse
|
21
|
Pujol CN, Paasche C, Laprevote V, Trojak B, Vidailhet P, Bacon E, Lalanne L. Cognitive effects of labeled addictolytic medications. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:306-332. [PMID: 28919445 DOI: 10.1016/j.pnpbp.2017.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/11/2017] [Accepted: 09/11/2017] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Alcohol, tobacco, and illegal drug usage is pervasive throughout the world, and abuse of these substances is a major contributor to the global disease burden. Many pharmacotherapies have been developed over the last 50years to target addictive disorders. While the efficacy of these pharmacotherapies is largely recognized, their cognitive impact is less known. However, all substance abuse disorders are known to promote cognitive disorders like executive dysfunction and memory impairment. These impairments are critical for the maintenance of addictive behaviors and impede cognitive behavioral therapies that are regularly administered in association with pharmacotherapies. It is also unknown if addictolytic medications have an impact on preexisting cognitive disorders, and if this impact is modulated by the indication of prescription, i.e. abstinence, reduction or substitution, or by the specific action of the medication. METHOD We reviewed the cognitive effects of labeled medications for tobacco addiction (varenicline, bupropion, nicotine patch and nicotine gums), alcohol addiction (naltrexone, nalmefene, baclofen, disulfiram, sodium oxybate, acamprosate), and opioid addiction (methadone, buprenorphine) in human studies. Studies were selected following MOOSE guidelines for systematic reviews of observational studies, using the keywords [Cognition] and [Cognitive disorders] and [treatment] for each medication. RESULTS 971 articles were screened and 77 studies met the inclusion criteria and were reported in this review (for alcohol abuse, n=21, for tobacco n=22, for opioid n=34. However, very few comparative clinical trials have explored the chronic effects of addictolytic medications on cognition in addictive behaviors, and there are no clinical trials on the cognitive impact of nalmefene in patients suffering from alcohol use disorders. DISCUSSION Although some medications seem to enhance cognition in patients suffering from cognitive disorders, others could promote cognitive impairments, and our work highlights a lack of literature on this subject. In conclusion, more comparative clinical trials are needed to better understand the cognitive impact of addictolytic medications.
Collapse
Affiliation(s)
- Camille Noélie Pujol
- Department of Neurosciences, Institute for Functional Genomics, INSERM U-661, CNRS UMR-5203, 34094 Montpellier, France
| | - Cecilia Paasche
- INSERM 1114, Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Vincent Laprevote
- Centre Psychothérapique de Nancy, Laxou, F-54520, France.; EA 7298, INGRES, Université de Lorraine, Vandoeuvre-lès-, Nancy F-54000, France; CHU Nancy, Maison des Addictions, Nancy, F-54000, France.
| | - Benoit Trojak
- Department of Psychiatry and Addictology, University Hospital of Dijon, France; EA 4452, LPPM, University of Burgundy, France.
| | - Pierre Vidailhet
- INSERM 1114, Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France; Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France..
| | - Elisabeth Bacon
- INSERM 1114, Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France.
| | - Laurence Lalanne
- INSERM 1114, Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France; Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France..
| |
Collapse
|
22
|
Abstract
SummaryThe relationship between alcohol use and cognitive impairment has been notoriously difficult to disentangle. We present what is known about cognitive impairment associated with alcohol use/misuse, covering the spectrum from mild and subtle cognitive change through to severe alcohol-related brain damage, including Wernicke-Korsakoff syndrome. We highlight aids to the diagnosis and management of these conditions, and emphasise the benefits of prompt treatment on outcome. We also review progress in understanding their neurobiology. Suggestions for possible service configuration based on both our clinical practice and national guidelines are given.Learning Objectives•Gain an understanding of the spectrum of clinical presentations found in alcohol-related brain damage.•Understand that the aetiology of these conditions is complex and not solely due to the neurotoxic effects of alcohol.•Be better able to plan for the rehabilitation of individuals with established alcohol-related brain damage in your local service area.
Collapse
|
23
|
Söderpalm B, Lidö HH, Ericson M. The Glycine Receptor-A Functionally Important Primary Brain Target of Ethanol. Alcohol Clin Exp Res 2017; 41:1816-1830. [PMID: 28833225 DOI: 10.1111/acer.13483] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/15/2017] [Indexed: 12/27/2022]
Abstract
Identification of ethanol's (EtOH) primary molecular brain targets and determination of their functional role is an ongoing, important quest. Pentameric ligand-gated ion channels, that is, the nicotinic acetylcholine receptor, the γ-aminobutyric acid type A receptor, the 5-hydroxytryptamine3 , and the glycine receptor (GlyR), are such targets. Here, aspects of the structure and function of these receptors and EtOH's interaction with them are briefly reviewed, with special emphasis on the GlyR and the importance of this receptor and its ligands for EtOH pharmacology. It is suggested that GlyRs are involved in (i) the dopamine-activating effect of EtOH, (ii) regulating EtOH intake, and (iii) the relapse preventing effect of acamprosate. Exploration of the GlyR subtypes involved and efforts to develop subtype specific agonists or antagonists may offer new pharmacotherapies for alcohol use disorders.
Collapse
Affiliation(s)
- Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Helga H Lidö
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
24
|
Mason BJ. Emerging pharmacotherapies for alcohol use disorder. Neuropharmacology 2017; 122:244-253. [PMID: 28454983 PMCID: PMC5643030 DOI: 10.1016/j.neuropharm.2017.04.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 01/29/2023]
Abstract
The identification of different stages within the alcohol use disorder (AUD) cycle that are linked to neurocircuitry changes in pathophysiology associated with the negative emotional states of abstinence has provided a view of medication development for AUD that emphasizes changes in the brain reward and stress systems. Alcohol use disorder can be defined as a chronic relapsing disorder that involves compulsive alcohol seeking and taking, loss of control over alcohol intake, and emergence of a negative emotional state during abstinence. The focus of early medications development was to block the motivation to seek alcohol in the binge/intoxication stage. More recent work has focused on reversing the motivational dysregulations associated with the withdrawal/negative affect and preoccupation/anticipation stages during protracted abstinence. Advances in our understanding of the neurocircuitry and neuropharmacological mechanisms that are involved in the development and maintenance of the withdrawal/negative affect stage using validated animal models have provided viable targets for future medications. Another major advance has been proof-of-concept testing of potential therapeutics and clinical validation of relevant pharmacological targets using human laboratory models of protracted abstinence. This review focuses on future targets for medication development associated with reversal of the loss of reward function and gain in brain stress function that drive negative reinforcement in the withdrawal/negative affect stage of addiction. Basic research has identified novel neurobiological targets associated with the withdrawal/negative affect stage and preoccupation/anticipation stage, with a focus on neuroadaptive changes within the extended amygdala that account for the transition to dependence and vulnerability to relapse. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Barbara J Mason
- The Pearson Center on Alcoholism and Addiction Research, Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, TPC-5 La Jolla, CA 92037 USA.
| |
Collapse
|
25
|
Hanak C, Benoit J, Fabry L, Hein M, Verbanck P, de Witte P, Walter H, Dexter DT, Ward RJ. Changes in Pro-Inflammatory Markers in Detoxifying Chronic Alcohol Abusers, Divided by Lesch Typology, Reflect Cognitive Dysfunction. Alcohol Alcohol 2017; 52:529-534. [DOI: 10.1093/alcalc/agx043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 06/12/2017] [Indexed: 12/14/2022] Open
|
26
|
The galanin-3 receptor antagonist, SNAP 37889, suppresses alcohol drinking and morphine self-administration in mice. Neuropharmacology 2017; 118:1-12. [DOI: 10.1016/j.neuropharm.2017.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/17/2017] [Accepted: 03/02/2017] [Indexed: 02/07/2023]
|
27
|
A Phase 2, Double-Blind, Placebo-Controlled Randomized Trial Assessing the Efficacy of ABT-436, a Novel V1b Receptor Antagonist, for Alcohol Dependence. Neuropsychopharmacology 2017; 42:1012-1023. [PMID: 27658483 PMCID: PMC5506792 DOI: 10.1038/npp.2016.214] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/30/2016] [Accepted: 09/08/2016] [Indexed: 12/20/2022]
Abstract
Alcohol use disorder has been linked to dysregulation of the brain stress systems, producing a negative emotional state leading to chronic relapsing behavior. Vasopressin receptors appear to have a regulatory role in stress, anxiety, and alcohol. This study evaluated the novel compound, ABT-436, a V1b receptor antagonist, in alcohol-dependent participants in a 12-week clinical trial. Men and women (n=150) who met criteria for DSM-IV alcohol dependence were recruited across four sites. Participants received double-blind ABT-436 or placebo, and a computerized behavioral intervention. ABT-436 was titrated to 800 mg/day during weeks 2-12. Although the primary outcome, percentage of heavy drinking days, was lower in participants receiving ABT-436 compared with placebo, this difference was not statistically significant (31.3 vs 37.6, respectively; p=0.172; d=0.20). However, participants receiving ABT-436 had significantly greater percentage of days abstinent than those receiving placebo (51.2 vs 41.6, respectively; p=0.037; d=0.31). No significant differences were found between treatment groups on any other measures of drinking, alcohol craving, or alcohol-related consequences. Smokers receiving ABT-436 smoked significantly fewer cigarettes per week than those receiving placebo (p=0.046). ABT-436 was well tolerated, with diarrhea (mild-to-moderate severity) being the most common side effect. In subgroup analyses, participants with relatively higher baseline levels of stress responded better to ABT-436 than placebo on select drinking outcomes, suggesting there may be value in testing medications targeting the vasopressin receptor in high stress, alcohol-dependent patients.
Collapse
|
28
|
Orrico A, Martí-Prats L, Cano-Cebrián MJ, Granero L, Polache A, Zornoza T. Pre-Clinical Studies with D-Penicillamine as a Novel Pharmacological Strategy to Treat Alcoholism: Updated Evidences. Front Behav Neurosci 2017; 11:37. [PMID: 28326026 PMCID: PMC5339308 DOI: 10.3389/fnbeh.2017.00037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 02/21/2017] [Indexed: 11/29/2022] Open
Abstract
Ethanol, as other drugs of abuse, is able to activate the ventral tegmental area dopamine (VTA-DA) neurons leading to positively motivational alcohol-seeking behavior and use, and, ultimately to ethanol addiction. In the last decades, the involvement of brain-derived acetaldehyde (ACD) in the ethanol actions in the mesolimbic pathway has been widely demonstrated. Consistent published results have provided a mechanistic support to the use of ACD inactivating agents to block the motivational and reinforcing properties of ethanol. Hence, in the last years, several pre-clinical studies have been performed in order to analyze the effects of the sequestering ACD agents in the prevention of ethanol relapse-like drinking behavior as well as in chronic alcohol consumption. In this sense, one of the most explored interventions has been the administration of D-Penicillamine (DP). These pre-clinical studies, that we critically summarize in this article, are considered a critical step for the potential development of a novel pharmacotherapeutic strategy for alcohol addiction treatment that could improve the outcomes of current ones. Thus, on one hand, several experimental findings provide the rationale for using DP as a novel therapeutic intervention alone and/or in combination to prevent relapse into alcohol seeking and consumption. On the other hand, its effectiveness in reducing voluntary ethanol consumption in long-term experienced animals still remains unclear. Finally, this drug offers the additional advantage that has already been approved for use in humans, hence it could be easily implemented as a new therapeutic intervention for relapse prevention in alcoholism.
Collapse
Affiliation(s)
- Alejandro Orrico
- Área de Investigación en Vacunas, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO) Valencia, Spain
| | | | - María J Cano-Cebrián
- Department of Pharmacy and Pharmacy Technology and Parasitology, University of Valencia Valencia, Spain
| | - Luis Granero
- Department of Pharmacy and Pharmacy Technology and Parasitology, University of Valencia Valencia, Spain
| | - Ana Polache
- Department of Pharmacy and Pharmacy Technology and Parasitology, University of Valencia Valencia, Spain
| | - Teodoro Zornoza
- Department of Pharmacy and Pharmacy Technology and Parasitology, University of Valencia Valencia, Spain
| |
Collapse
|
29
|
Abstract
Genetic factors contribute to more than 50% of the variation in the vulnerability to alcohol dependence (AD). Although significant advances have been made in medications for AD, these medications do not work for all people. Precise tailoring of medicinal strategies for individual alcoholic patients is needed to achieve optimal outcomes. This review updates the most promising information on genetic variants in AD, which may be useful for improving diagnostic, therapeutic, and monitoring strategies. We describe genetic candidates of various neurotransmitter and enzyme systems. In addition to biological and allelic associations with AD, genetic effects on AD-related phenotypes and treatment responses have also been described. Gene-gene and gene-environment interactions have been considered. Potential applications of genomewide and epigenetic approaches for identifying genetic biomarkers of AD have been discussed. Overall, the application of genetic findings in precision medicine for AD will likely involve an integrated approach that distinguishes effect sizes of specific genetic predictors with regard to sex, pharmacotherapy, ethnicity, and AD-related aspects and considers gene-gene and gene-environment interactions. Our work may pave the way toward more precise treatment for AD that could ultimately improve clinical management and interventions.
Collapse
|
30
|
Addolorato G, Mirijello A, Barrio P, Gual A. Treatment of alcohol use disorders in patients with alcoholic liver disease. J Hepatol 2016; 65:618-30. [PMID: 27155530 DOI: 10.1016/j.jhep.2016.04.029] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/24/2016] [Accepted: 04/25/2016] [Indexed: 12/18/2022]
Abstract
Alcohol use disorders (AUDs) is one of the leading causes of disease and disability in almost all European countries. Among the alcohol-related diseases, alcoholic liver disease (ALD) is the most common. At present, alcohol is the most frequent cause of liver cirrhosis in the Western world. The cornerstone of treatment for ALD is achieving total alcohol abstinence and preventing relapse; medical and surgical treatments for ALD are limited when drinking continues. This narrative review summarizes current treatments for AUDs with a particular emphasis to the treatment of AUDs in patients with ALD. Medical management, psychosocial and pharmacological interventions are analyzed, underlying limits and options in AUD patients. Finally, this review discusses the most appropriate setting for the management of AUD patients with advanced liver disease as well as the indications for liver transplantation in AUD patients.
Collapse
Affiliation(s)
- Giovanni Addolorato
- Alcohol Use Disorders Unit, Department of Internal Medicine, Gastroenterology and Hepatology, Catholic University of Rome, Italy.
| | - Antonio Mirijello
- Alcohol Use Disorders Unit, Department of Internal Medicine, Gastroenterology and Hepatology, Catholic University of Rome, Italy; Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, FG, Italy
| | - Pablo Barrio
- Department of Psychiatry, Neurosciences Institute, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Antoni Gual
- Department of Psychiatry, Neurosciences Institute, Hospital Clínic, IDIBAPS, Barcelona, Spain.
| |
Collapse
|
31
|
Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry 2016; 3:760-773. [PMID: 27475769 PMCID: PMC6135092 DOI: 10.1016/s2215-0366(16)00104-8] [Citation(s) in RCA: 1947] [Impact Index Per Article: 216.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/09/2016] [Accepted: 03/11/2016] [Indexed: 12/17/2022]
Abstract
Drug addiction represents a dramatic dysregulation of motivational circuits that is caused by a combination of exaggerated incentive salience and habit formation, reward deficits and stress surfeits, and compromised executive function in three stages. The rewarding effects of drugs of abuse, development of incentive salience, and development of drug-seeking habits in the binge/intoxication stage involve changes in dopamine and opioid peptides in the basal ganglia. The increases in negative emotional states and dysphoric and stress-like responses in the withdrawal/negative affect stage involve decreases in the function of the dopamine component of the reward system and recruitment of brain stress neurotransmitters, such as corticotropin-releasing factor and dynorphin, in the neurocircuitry of the extended amygdala. The craving and deficits in executive function in the so-called preoccupation/anticipation stage involve the dysregulation of key afferent projections from the prefrontal cortex and insula, including glutamate, to the basal ganglia and extended amygdala. Molecular genetic studies have identified transduction and transcription factors that act in neurocircuitry associated with the development and maintenance of addiction that might mediate initial vulnerability, maintenance, and relapse associated with addiction.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA.
| | - Nora D Volkow
- National Institute on Drug Abuse, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
32
|
Kwako LE, Momenan R, Litten RZ, Koob GF, Goldman D. Addictions Neuroclinical Assessment: A Neuroscience-Based Framework for Addictive Disorders. Biol Psychiatry 2016; 80:179-89. [PMID: 26772405 PMCID: PMC4870153 DOI: 10.1016/j.biopsych.2015.10.024] [Citation(s) in RCA: 234] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 02/06/2023]
Abstract
This article proposes a heuristic framework for the Addictions Neuroclinical Assessment that incorporates key functional domains derived from the neurocircuitry of addiction. We review how addictive disorders (ADs) are presently diagnosed and the need for new neuroclinical measures to differentiate patients who meet clinical criteria for addiction to the same agent while differing in etiology, prognosis, and treatment response. The need for a better understanding of the mechanisms provoking and maintaining addiction, as evidenced by the limitations of current treatments and within-diagnosis clinical heterogeneity, is articulated. In addition, recent changes in the nosology of ADs, challenges to current classification systems, and prior attempts to subtype individuals with ADs are described. Complementary initiatives, including the Research Domain Criteria project, that have established frameworks for the neuroscience of psychiatric disorders are discussed. Three domains-executive function, incentive salience, and negative emotionality-tied to different phases in the cycle of addiction form the core functional elements of ADs. Measurement of these domains in epidemiologic, genetic, clinical, and treatment studies will provide the underpinnings for an understanding of cross-population and temporal variation in addictions, shared mechanisms in addictive disorders, impact of changing environmental influences, and gene identification. Finally, we show that it is practical to implement such a deep neuroclinical assessment using a combination of neuroimaging and performance measures. Neuroclinical assessment is key to reconceptualizing the nosology of ADs on the basis of process and etiology, an advance that can lead to improved prevention and treatment.
Collapse
Affiliation(s)
- Laura E Kwako
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland.
| | - Reza Momenan
- Section on Brain Electrophysiology and Imaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Raye Z Litten
- Division of Intramural Clinical and Biological Research; Division of Treatment and Recovery Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - George F Koob
- Office of the Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - David Goldman
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
33
|
Abstract
Despite increased awareness, prevention campaigns, and tighter laws and regulations, alcohol consumption caused an average of 88,000 deaths per year and a burden of $249 billion to the U.S economy in 2010. Only three FDA-approved drugs, disulfiram, naltrexone and acamprosate are available for the treatment of alcohol use disorder. These drugs are only effective modestly and patient compliance is a serious issue because of several adverse side effects, necessitating the developments of newer drugs. The current research drug development efforts remains in the identification new alcohol targets and elucidating the molecular mechanism of its action is needed for effective intervention. In addition, current treatment could be improved by testing the existing medications for comorbid conditions using the patient’s genetic background information.
Collapse
Affiliation(s)
- Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, US
| |
Collapse
|
34
|
Masternak S, Skoczeń N, Dziwota E, Olajossy M. Is it time for baclofen to be included in the official recommendations concerning the treatment of alcoholism? CURRENT PROBLEMS OF PSYCHIATRY 2016. [DOI: 10.1515/cpp-2016-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractAlcohol dependence and its treatment is not an exactly resolved problem. Based on the EZOP [Epidemiology of Mental Disorders and Accessibility of Mental Health Care] survey, which included a regular analysis of the incidence of mental disorders in the population of adult Polish citizens, we were able to estimate that the problem of alcohol abuse in any period of life affects even 10.9% of the population aged 18-64 years, and those addicted represent 2.2% of the country’s population. The typical symptoms of alcohol dependence according to ICD-10, include alcohol craving, impaired ability to control alcohol consumption, withdrawal symptoms which appear when a heavy drinker stops drinking, alternating alcohol tolerance, growing neglect of other areas of life, and persistent alcohol intake despite clear evidence of its destructive effect on life. At the moment, the primary method of alcoholism treatment is psychotherapy. It aims to change the patient’s habits, behaviours, relationships, or the way of thinking. It seems that psychotherapy is irreplaceable in the treatment of alcoholism, but for many years now attempts have been made to increase the effectiveness of alcoholism treatment with pharmacological agents. In this article we will try to provide a description of medications which help patients sustain abstinence in alcoholism therapy with particular emphasis on baclofen.
Collapse
Affiliation(s)
- Sebastian Masternak
- II Department of Psychiatry and Psychiatric Rehabilitation, Medical University of Lublin Poland
| | - Nikodem Skoczeń
- II Department of Psychiatry and Psychiatric Rehabilitation, Medical University of Lublin Poland
| | - Ewelina Dziwota
- II Department of Psychiatry and Psychiatric Rehabilitation, Medical University of Lublin Poland
| | - Marcin Olajossy
- II Department of Psychiatry and Psychiatric Rehabilitation, Medical University of Lublin Poland
| |
Collapse
|
35
|
Abstract
Acamprosate (Campral(®), Aotal(®), Regtect(®)) is one of a limited number of pharmacological treatment options approved as an adjunct to psychosocial interventions to facilitate the maintenance of abstinence in alcohol-dependent patients. It has been used in Europe, the USA and other countries for many years and was recently approved for this indication in Japan. In several randomized, double-blind, placebo-controlled trials (without active comparators), acamprosate in conjunction with psychosocial therapy for 3-12 months was generally significantly better than placebo plus psychosocial interventions in improving various key outcomes, including the proportion of patients who maintained complete abstinence from alcohol (complete abstinence rate), the mean cumulative abstinence duration, the percentage of alcohol-free days and the median time to first drink. Acamprosate as an adjunct to psychosocial interventions also demonstrated efficacy in some randomized, active-comparator trials of similar duration. Although results were not always consistent across individual trials, overall findings were generally favourable for acamprosate in a recent meta-analysis, which showed that alcohol-consumption outcomes were similarly improved with acamprosate or naltrexone. Acamprosate is generally well tolerated, has a low propensity for drug interactions and may be used without dosage adjustment in patients with mild to moderate hepatic impairment, although dosage adjustments or contraindications are recommended in patients with renal impairment. Thus, the use of acamprosate as an adjunct to psychosocial interventions in alcohol-dependent patients provides modest but potentially valuable improvements in alcohol-consumption outcomes and is generally well tolerated.
Collapse
Affiliation(s)
- Greg L Plosker
- Springer, Private Bag 65901, Mairangi Bay, 0754, Auckland, New Zealand,
| |
Collapse
|
36
|
Rosato V, Abenavoli L, Federico A, Masarone M, Persico M. Pharmacotherapy of alcoholic liver disease in clinical practice. Int J Clin Pract 2016; 70:119-31. [PMID: 26709723 DOI: 10.1111/ijcp.12764] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIMS Alcohol is the most commonly used addictive substance and alcoholic liver disease (ALD) is a major cause of chronic liver disease worldwide, responsible for 47.9% of all liver chronic deaths. Despite ALD has a significant burden on the health, few therapeutic advances have been made in the last 40 years, particularly in the long-term management of these patients. METHODS we searched in PubMed, Scopus, Google Scholar, and MEDLINE databases to identify relevant English language publications focused on long-term therapy of ALD. RESULTS From the huge literature on this topic, including about 755 studies, 75 were selected as eligible including clinical trials and meta-analysis. CONCLUSIONS Abstinence remains the cornerstone of ALD therapy but it is also the most difficult therapeutic target to achieve and the risk of recidivism is very high at any time. Several drugs (disulfiram, naltrexone, acamprosate, sodium oxybate) have proven to be effective to prevent alcohol relapse and increase the abstinence, although the psychotherapeutic support remains crucial. Baclofen seems to be effective to improve abstinence, showing an excellent safety and tolerability. ALD is often complicated by a state of malnutrition, which is related to a worst mortality. A nutritional therapy may improve survival in cirrhotic patients, reversing muscle wasting, weight loss and specific nutritional deficiencies. While in aggressive forms of alcoholic hepatitis are recommended specific drug treatments, including glucocorticoids or pentoxifylline, for the long-term treatment of ALD, specific treatments aimed at stopping the progression of fibrosis are not yet approved, but there are some future perspective in this field, including probiotics and antibiotics, caspase inhibitors, osteopontin and endocannabinoids.
Collapse
Affiliation(s)
- V Rosato
- Internal Medicine and Hepatology Department, Second University of Naples, Naples, Italy
| | - L Abenavoli
- Department of Health Science, University Magna Graecia, Catanzaro, Italy
| | - A Federico
- Gastroenterology and Endoscopy Unit, Second University of Naples, Naples, Italy
| | - M Masarone
- Internal Medicine and Hepatology Unit, University of Salerno, Baronissi, Italy
| | - M Persico
- Internal Medicine and Hepatology Unit, University of Salerno, Baronissi, Italy
| |
Collapse
|
37
|
Gitto S, Golfieri L, Caputo F, Grandi S, Andreone P. Multidisciplinary View of Alcohol Use Disorder: From a Psychiatric Illness to a Major Liver Disease. Biomolecules 2016; 6:11. [PMID: 26784248 PMCID: PMC4808805 DOI: 10.3390/biom6010011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 12/16/2015] [Accepted: 12/27/2015] [Indexed: 12/20/2022] Open
Abstract
Alcohol use disorder is a significant health problem being a cause of increased morbidity and mortality worldwide. Alcohol-related illness has a relevant economic impact on the society and a negative influence on the life of patients and their family members. Psychosocial support might be useful in the management of people affected by alcohol use disorder since psychiatric and pharmaceutical approaches show some limits. In fact, many drugs are accessible for the treatment of alcohol disorder, but only Baclofen is functional as an anti-craving drug in patients with advanced liver disease. The alcohol-related liver damage represents the most frequent cause of advanced liver disease in Europe, and it is the main cause of death among adults with high alcohol consumption. The multidisciplinary action of clinical-psychologists, psychiatrics and hepatologists, is essential in the management of patients with alcohol liver disease especially in the case of liver transplantation. In general, the multidisciplinary approach is necessary in prevention, in framing patients and in the treatment. More resources should be used in prevention and research with the main aim of decreasing the harmful alcohol consumption.
Collapse
Affiliation(s)
- Stefano Gitto
- Department of Gastroenterology, Azienda Ospedaliero-Universitaria & University of Modena and Reggio Emilia, Modena 41124, Italy.
| | - Lucia Golfieri
- Department of Psychology, University of Bologna, Bologna 40138, Italy.
| | - Fabio Caputo
- Department of Internal Medicine, SS Annunziata Hospital, Cento, Ferrara 44011, Italy.
- "G. Fontana" Centre for the Study and Multidisciplinary Treatment of Alcohol Addiction, Department of Clinical Medicine, University of Bologna, Bologna 40138, Italy.
| | - Silvana Grandi
- Department of Psychology, University of Bologna, Bologna 40138, Italy.
| | - Pietro Andreone
- Department of Medical and Surgical Sciences, University of Bologna and Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola Malpighi, Via Massarenti 9, Bologna 40138, Italy.
| |
Collapse
|
38
|
Herman MA, Varodayan FP, Oleata CS, Luu G, Kirson D, Heilig M, Ciccocioppo R, Roberto M. Glutamatergic transmission in the central nucleus of the amygdala is selectively altered in Marchigian Sardinian alcohol-preferring rats: Alcohol and CRF effects. Neuropharmacology 2015; 102:21-31. [PMID: 26519902 DOI: 10.1016/j.neuropharm.2015.10.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/22/2015] [Accepted: 10/22/2015] [Indexed: 11/25/2022]
Abstract
The CRF system of the central nucleus of the amygdala (CeA) is important for the processing of anxiety, stress, and effects of acute and chronic ethanol. We previously reported that ethanol decreases evoked glutamate transmission in the CeA of Sprague Dawley rats and that ethanol dependence alters glutamate release in the CeA. Here, we examined the effects of ethanol, CRF and a CRF1 receptor antagonist on spontaneous and evoked glutamatergic transmission in CeA neurons from Wistar and Marchigian Sardinian Preferring (msP) rats, a rodent line genetically selected for excessive alcohol drinking and characterized by heightened activity of the CRF1 system. Basal spontaneous and evoked glutamate transmission in CeA neurons from msP rats was increased compared to Wistar rats. Ethanol had divergent effects, either increasing or decreasing spontaneous glutamate release in the CeA of Wistar rats. This bidirectional effect was retained in msP rats, but the magnitude of the ethanol-induced increase in glutamate release was significantly smaller. The inhibitory effect of ethanol on evoked glutamatergic transmission was similar in both strains. CRF also either increased or decreased spontaneous glutamate release in CeA neurons of Wistar rats, however, in msP rats CRF only increased glutamate release. The inhibitory effect of CRF on evoked glutamatergic transmission was also lost in neurons from msP rats. A CRF1 antagonist produced only minor effects on spontaneous glutamate transmission, which were consistent across strains, and no effects on evoked glutamate transmission. These results demonstrate that the genetically altered CRF system of msP rats results in alterations in spontaneous and stimulated glutamate signaling in the CeA that may contribute to both the anxiety and drinking behavioral phenotypes.
Collapse
Affiliation(s)
- Melissa A Herman
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Florence P Varodayan
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher S Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Dean Kirson
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Markus Heilig
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Clinical and Translational Sciences, National Institutes of Health, Bldg.10-CRC/Rm. 1-5330, 10 Center Drive, Bethesda, MD 20892-1108, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
39
|
Koob GF, Mason BJ. Existing and Future Drugs for the Treatment of the Dark Side of Addiction. Annu Rev Pharmacol Toxicol 2015; 56:299-322. [PMID: 26514207 DOI: 10.1146/annurev-pharmtox-010715-103143] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The identification of a heuristic framework for the stages of the addiction cycle that are linked to neurocircuitry changes in pathophysiology includes the binge/intoxication stage, the withdrawal/negative affect stage, and the preoccupation/anticipation (craving) stage, which represent neuroadaptations in three neurocircuits (basal ganglia, extended amygdala, and frontal cortex, respectively). The identification of excellent and validated animal models, the development of human laboratory models, and an enormous surge in our understanding of neurocircuitry and neuropharmacological mechanisms have provided a revisionist view of addiction that emphasizes the loss of brain reward function and gain of stress function that drive negative reinforcement (the dark side of addiction) as a key to compulsive drug seeking. Reversing the dark side of addiction not only explains much of the existing successful pharmacotherapies for addiction but also points to vast new opportunities for future medications to alleviate this major source of human suffering.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037; ,
| | - Barbara J Mason
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037; ,
| |
Collapse
|
40
|
Chumakov I, Nabirotchkin S, Cholet N, Milet A, Boucard A, Toulorge D, Pereira Y, Graudens E, Traoré S, Foucquier J, Guedj M, Vial E, Callizot N, Steinschneider R, Maurice T, Bertrand V, Scart-Grès C, Hajj R, Cohen D. Combining two repurposed drugs as a promising approach for Alzheimer's disease therapy. Sci Rep 2015; 5:7608. [PMID: 25566747 PMCID: PMC5378993 DOI: 10.1038/srep07608] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/19/2014] [Indexed: 02/08/2023] Open
Abstract
Alzheimer disease (AD) represents a major medical problem where mono-therapeutic interventions demonstrated only a limited efficacy so far. We explored the possibility of developing a combinational therapy that might prevent the degradation of neuronal and endothelial structures in this disease. We argued that the distorted balance between excitatory (glutamate) and inhibitory (GABA/glycine) systems constitutes a therapeutic target for such intervention. We found that a combination of two approved drugs – acamprosate and baclofen – synergistically protected neurons and endothelial structures in vitro against amyloid-beta (Aβ) oligomers. The neuroprotective effects of these drugs were mediated by modulation of targets in GABA/glycinergic and glutamatergic pathways. In vivo, the combination alleviated cognitive deficits in the acute Aβ25–35 peptide injection model and in the mouse mutant APP transgenic model. Several patterns altered in AD were also synergistically normalised. Our results open up the possibility for a promising therapeutic approach for AD by combining repurposed drugs.
Collapse
Affiliation(s)
- Ilya Chumakov
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | - Nathalie Cholet
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Aude Milet
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Aurélie Boucard
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Damien Toulorge
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Yannick Pereira
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Esther Graudens
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Sory Traoré
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Julie Foucquier
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Mickael Guedj
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Emmanuel Vial
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | | | - Tangui Maurice
- 1] Université de Montpellier 2, 34095 Montpellier, France; Inserm, U710, 34095 Montpellier, France; EPHE, 75017 Paris, France [2] Amylgen, 2196 bd de la Lironde, 34980 Montferrier-sur-Lez, France
| | - Viviane Bertrand
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | - Rodolphe Hajj
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Daniel Cohen
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| |
Collapse
|
41
|
Six-month outcome in bipolar spectrum alcoholics treated with acamprosate after detoxification: a retrospective study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2014; 11:12983-96. [PMID: 25514151 PMCID: PMC4276657 DOI: 10.3390/ijerph111212983] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/25/2014] [Accepted: 12/05/2014] [Indexed: 11/30/2022]
Abstract
Background: Glutamate system is modified by ethanol and contributes both to the euphoric and the dysphoric consequences of intoxication, but there is now growing evidence that the glutamatergic system also plays a central role in the neurobiology and treatment of mood disorders, including major depressive disorders and bipolar disorders. We speculate that, using acamprosate, patients with bipolar depression (BIP-A) can take advantage of the anti-glutamate effect of acamprosate to “survive” in treatment longer than peers suffering from non-bipolar depression (NBIP-A) after detoxification. Method: We retrospectively evaluated the efficacy of a long-term (six-month) acamprosate treatment, after alcohol detoxification, in 41 patients (19 males and 22 females), who could be classified as depressed alcoholics, while taking into account the presence/absence of bipolarity. Results: During the period of observation most NBIP-A patients relapsed, whereas a majority of BIP-A patients were still in treatment at the end of their period of observation. The cumulative proportion of ‘surviving’ patients was significantly higher in BIP-A patients, but this finding was not related to gender or to other demographic or clinically investigated characteristics. The treatment time effect was significant in both subgroups. The treatment time-group effect was significant (and significantly better) for bipolar patients on account of changes in the severity of their illness. Limitations: Retrospective methodology and the lack of DSM criteria in diagnosing bipolarity. Conclusions: Bipolarity seems to be correlated with the efficacy of acamprosate treatment in inducing patients to refrain from alcohol use after detoxification (while avoiding relapses) in depressed alcoholics. Placebo-controlled clinical trials are now warranted to check the validity of this hypothesis.
Collapse
|
42
|
Peana AT, Muggironi G, Bennardini F. Change of cystine/glutamate antiporter expression in ethanol-dependent rats. Front Neurosci 2014; 8:311. [PMID: 25339860 PMCID: PMC4186264 DOI: 10.3389/fnins.2014.00311] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/14/2014] [Indexed: 01/31/2023] Open
Abstract
Background: Some drugs of abuse down regulate the expression of cystine/glutamate (xCT) antiporter in the nucleus accumbens (Acb) after extinction or withdrawal. The altered level of xCT exchanger in Acb, a structure involved in ethanol reinforcement, may contribute to the pathological glutamatergic signaling, linked to addiction. We hypothesized that the expression of xCT may be changed in Acb and whole brain also in non-dependent (occasional drinkers), ethanol-dependent rats, as well as, during ethanol withdrawal. Methods: Wistar rats were made ethanol-dependent by chronic exposure to an alcoholic milk beverage (from 2.4 to 7.2% v/v ethanol). Ethanol non-dependent rats were exposed to a similar, but non-alcoholic liquid diet and self-administered ethanol (10%) twice a week. Withdrawal in ethanol-dependent rats was studied at 12 h after the last ethanol-enriched diet exposure. Immediately after the measurement of somatic signs of withdrawal, Western blot analysis with a polyclonal antibody against xCT was carried out in a naïve control group, non-dependent and ethanol-dependent rats as well as withdrawal rats, in order to study the level of xCT expression in Acb and whole brain. Results: Non-dependent rats self-administered an average dose of 1.21 ± 0.02 g/kg per session (30 min). Daily ethanol consumption during chronic exposure to the alcoholic beverage ranged from 6.30 ± 0.16 to 13.99 ± 0.66 g/kg. Ethanol dependent rats after suspension of the ethanol-enriched diet have shown significant somatic signs of withdrawal. Western blotting analysis of Acb lysates revealed that xCT was over expressed in ethanol-dependent rats whereas in whole brain preparations xCT was over expressed in both non-dependent and ethanol-dependent rats compared to control group. On the contrary, xCT expression during withdrawal was down regulated in Acb and restored to control level in whole brain preparations. Conclusions: The changes of xCT expression in both Acb and whole brain following ethanol dependence and withdrawal indicate that xCT might represent a novel therapeutic target for the treatment of ethanol addiction.
Collapse
Affiliation(s)
- Alessandra T Peana
- Department of Chemistry and Pharmacy, University of Sassari Sassari, Italy
| | - Giulia Muggironi
- Department of Chemistry and Pharmacy, University of Sassari Sassari, Italy
| | | |
Collapse
|
43
|
Targeting the glutamatergic system to treat pathological gambling: current evidence and future perspectives. BIOMED RESEARCH INTERNATIONAL 2014; 2014:109786. [PMID: 25013755 PMCID: PMC4075088 DOI: 10.1155/2014/109786] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/22/2014] [Indexed: 12/14/2022]
Abstract
Pathological gambling or gambling disorder has been defined by the DSM-5 as a behavioral addiction. To date, its pathophysiology is not completely understood and there is no FDA-approved treatment for gambling disorders. Glutamate is the principal excitatory neurotransmitter in the nervous system and it has been recently involved in the pathophysiology of addictive behaviors. In this paper, we review the current literature on a class of drugs that act as modulating glutamate system in PG. A total of 19 studies have been included, according to inclusion and exclusion criteria. Clinical trial and case series using glutamatergic drugs (N-acetylcysteine, memantine, amantadine, topiramate, acamprosate, baclofen, gabapentin, pregabalin, and modafinil) will be presented to elucidate the effectiveness on gambling behaviors and on the related clinical dimensions (craving, withdrawal, and cognitive symptoms) in PG patients. The results have been discussed to gain more insight in the pathophysiology and treatment of PG. In conclusion, manipulation of glutamatergic neurotransmission appears to be promising in developing improved therapeutic agents for the treatment of gambling disorders. Further studies are required. Finally, we propose future directions and challenges in this research area.
Collapse
|
44
|
Rubio MD, Drummond JB, Meador-Woodruff JH. Glutamate receptor abnormalities in schizophrenia: implications for innovative treatments. Biomol Ther (Seoul) 2014; 20:1-18. [PMID: 24116269 PMCID: PMC3792192 DOI: 10.4062/biomolther.2012.20.1.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/25/2011] [Indexed: 01/18/2023] Open
Abstract
Schizophrenia is a devastating psychiatric illness that afflicts 1% of the population worldwide, resulting in substantial impact to patients, their families, and health care delivery systems. For many years, schizophrenia has been felt to be associated with dysregulated dopaminergic neurotransmission as a key feature of the pathophysiology of the illness. Although numerous studies point to dopaminergic abnormalities in schizophrenia, dopamine dysfunction cannot completely account for all of the symptoms seen in schizophrenia, and dopamine-based treatments are often inadequate and can be associated with serious side effects. More recently, converging lines of evidence have suggested that there are abnormalities of glutamate transmission in schizophrenia. Glutamatergic neurotransmission involves numerous molecules that facilitate glutamate release, receptor activation, glutamate reuptake, and other synaptic activities. Evidence for glutamatergic abnormalities in schizophrenia primarily has implicated the NMDA and AMPA subtypes of the glutamate receptor. The expression of these receptors and other molecules associated with glutamate neurotransmission has been systematically studied in the brain in schizophrenia. These studies have generally revealed region- and molecule-specific changes in glutamate receptor transcript and protein expression in this illness. Given that glutamatergic neurotransmission has been implicated in the pathophysiology of schizophrenia, recent drug development efforts have targeted the glutamate system. Much effort to date has focused on modulation of the NMDA receptor, although more recently other glutamate receptors and transporters have been the targets of drug development. These efforts have been promising thus far, and ongoing efforts to develop additional drugs that modulate glutamatergic neurotransmission are underway that may hold the potential for novel classes of more effective treatments for this serious psychiatric illness.
Collapse
Affiliation(s)
- Maria D Rubio
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294-0021, USA
| | | | | |
Collapse
|
45
|
Haass-Koffler CL, Leggio L, Kenna GA. Pharmacological approaches to reducing craving in patients with alcohol use disorders. CNS Drugs 2014; 28:343-60. [PMID: 24573997 PMCID: PMC3990000 DOI: 10.1007/s40263-014-0149-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Research on the concept of craving may lead to better understanding of the biobehavioural circuitries that contribute to the complexity of alcohol use disorders (AUDs). The experiences described as craving or desire to drink are often associated with physical responses such as increased salivation and heart rate, and alteration of stress hormones, as well as psychological responses such as anxiety and depression. Greater craving has been associated with an increased probability of alcohol relapse. Reversal of craving, which is understood as a symptom of protracted abstinence, offers the possibility of preventing relapses and treating alcoholism. Various medications have been studied to establish whether they are able to reduce craving; however, the results obtained from clinical studies have been inconsistent. Here, we review the interdisciplinary models developed to evaluate craving, then the different approaches used to assess and measure craving and, finally, the medications utilized and tested to lessen craving in patients suffering from AUDs.
Collapse
Affiliation(s)
| | - Lorenzo Leggio
- Center for Alcohol & Addiction Studies, Brown University, Providence, Rhode Island, USA
- Intramural Research Program, National Institute on Drug Abuse (NIDA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - George A. Kenna
- Center for Alcohol & Addiction Studies, Brown University, Providence, Rhode Island, USA
- Department of Psychiatry and Human Behavior, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
46
|
Kallupi M, Varodayan FP, Oleata CS, Correia D, Luu G, Roberto M. Nociceptin/orphanin FQ decreases glutamate transmission and blocks ethanol-induced effects in the central amygdala of naive and ethanol-dependent rats. Neuropsychopharmacology 2014; 39:1081-92. [PMID: 24169802 PMCID: PMC3957102 DOI: 10.1038/npp.2013.308] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/08/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022]
Abstract
The central nucleus of the amygdala (CeA) mediates several addiction-related processes and nociceptin/orphanin FQ (nociceptin) regulates ethanol intake and anxiety-like behaviors. Glutamatergic synapses, in the CeA and throughout the brain, are very sensitive to ethanol and contribute to alcohol reinforcement, tolerance, and dependence. Previously, we reported that in the rat CeA, acute and chronic ethanol exposures significantly decrease glutamate transmission by both pre- and postsynaptic actions. In this study, using electrophysiological techniques in an in vitro CeA slice preparation, we investigated the effects of nociceptin on glutamatergic transmission and its interaction with acute ethanol in naive and ethanol-dependent rats. We found that nociceptin (100-1000 nM) diminished basal-evoked compound glutamatergic receptor-mediated excitatory postsynaptic potentials (EPSPs) and spontaneous and miniature EPSCs (s/mEPSCs) by mainly decreasing glutamate release in the CeA of naive rats. Notably, nociceptin blocked the inhibition induced by acute ethanol (44 mM) and ethanol blocked the nociceptin-induced inhibition of evoked EPSPs in CeA neurons of naive rats. In neurons from chronic ethanol-treated (ethanol-dependent) rats, the nociceptin-induced inhibition of evoked EPSP amplitude was not significantly different from that in naive rats. Application of [Nphe1]Nociceptin(1-13)NH2, a nociceptin receptor (NOP) antagonist, revealed tonic inhibitory activity of NOP on evoked CeA glutamatergic transmission only in ethanol-dependent rats. The antagonist also blocked nociceptin-induced decreases in glutamatergic responses, but did not affect ethanol-induced decreases in evoked EPSP amplitude. Taken together, these studies implicate a potential role for the nociceptin system in regulating glutamatergic transmission and a complex interaction with ethanol at CeA glutamatergic synapses.
Collapse
Affiliation(s)
- Marsida Kallupi
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Florence P Varodayan
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher S Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Diego Correia
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Pharmacology, Universidade Federal do Paraná, Jardim das Américas, Curitiba, Paraná, Brazil
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
47
|
Acamprosate produces its anti-relapse effects via calcium. Neuropsychopharmacology 2014; 39:783-91. [PMID: 24081303 PMCID: PMC3924515 DOI: 10.1038/npp.2013.264] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 09/16/2013] [Accepted: 09/17/2013] [Indexed: 11/08/2022]
Abstract
Alcoholism is one of the most prevalent neuropsychiatric diseases, having an enormous health and socioeconomic impact. Along with a few other medications, acamprosate (Campral-calcium-bis (N-acetylhomotaurinate)) is clinically used in many countries for relapse prevention. Although there is accumulated evidence suggesting that acamprosate interferes with the glutamate system, the molecular mode of action still remains undefined. Here we show that acamprosate does not interact with proposed glutamate receptor mechanisms. In particular, acamprosate does not interact with NMDA receptors or metabotropic glutamate receptor group I. In three different preclinical animal models of either excessive alcohol drinking, alcohol-seeking, or relapse-like drinking behavior, we demonstrate that N-acetylhomotaurinate by itself is not an active psychotropic molecule. Hence, the sodium salt of N-acetylhomotaurinate (i) is ineffective in alcohol-preferring rats to reduce operant responding for ethanol, (ii) is ineffective in alcohol-seeking rats in a cue-induced reinstatement paradigm, (iii) and is ineffective in rats with an alcohol deprivation effect. Surprisingly, calcium salts produce acamprosate-like effects in all three animal models. We conclude that calcium is the active moiety of acamprosate. Indeed, when translating these findings to the human situation, we found that patients with high plasma calcium levels due to acamprosate treatment showed better primary efficacy parameters such as time to relapse and cumulative abstinence. We conclude that N-acetylhomotaurinate is a biologically inactive molecule and that the effects of acamprosate described in more than 450 published original investigations and clinical trials and 1.5 million treated patients can possibly be attributed to calcium.
Collapse
|
48
|
Caputo F, Vignoli T, Grignaschi A, Cibin M, Addolorato G, Bernardi M. Pharmacological management of alcohol dependence: from mono-therapy to pharmacogenetics and beyond. Eur Neuropsychopharmacol 2014; 24:181-91. [PMID: 24182622 DOI: 10.1016/j.euroneuro.2013.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/06/2013] [Accepted: 10/09/2013] [Indexed: 01/01/2023]
Abstract
Almost 10% of the world's population is affected by alcohol use disorders, and the treatment of alcohol dependence (AD) still remains a challenge. Patients with AD can differ in many traits. Three drugs (disulfiram, naltrexone, and acamprosate) have been approved by the FDA for the treatment of AD, and in some European countries sodium oxybate is also approved for this purpose. Combined pharmacological therapy has not provided such convincing results. Considering the fact that the "ideal" and effective drug for all types of alcoholic patients does not exists, the future challenge will be to identify a personalized approach. Recent data has shown that this objective can be achieved by investigating the genetic variability of the patient. Moreover, the use of replacement molecules can probably be considered an advantageous therapeutic opportunity (i.e. sodium oxybate). In addition, reduction of alcohol consumption is increasingly accepted as a viable treatment goal, and the use of nalmefene "as-needed" (a pharmacological approach similar to naltrexone, but, possibly, with lower hepatotoxicity) may help in the treatment of AD. Thus, it is important to stress that a pharmacological approach to treat AD should be preceded by the definition of patient characteristics; this may help in the choice of the most appropriate drug and it can be done more easily when more pharmacological options approved for the treatment of AD are also available.
Collapse
Affiliation(s)
- Fabio Caputo
- Department of Internal Medicine, SS Annunziata Hospital, Cento, Ferrara, Italy; "G. Fontana" Centre for the Study and Multidisciplinary Treatment of Alcohol Addiction, Department of Clinical Medicine, University of Bologna, Italy.
| | - Teo Vignoli
- Unit for Addiction Treatment, Department of Mental Health, Lugo, Ravenna, Italy
| | - Alice Grignaschi
- "G. Fontana" Centre for the Study and Multidisciplinary Treatment of Alcohol Addiction, Department of Clinical Medicine, University of Bologna, Italy
| | - Mauro Cibin
- Department of Addictive Behaviours, Dolo, Venice, Italy
| | | | - Mauro Bernardi
- "G. Fontana" Centre for the Study and Multidisciplinary Treatment of Alcohol Addiction, Department of Clinical Medicine, University of Bologna, Italy
| |
Collapse
|
49
|
Wackernah RC, Minnick MJ, Clapp P. Alcohol use disorder: pathophysiology, effects, and pharmacologic options for treatment. Subst Abuse Rehabil 2014; 5:1-12. [PMID: 24648792 PMCID: PMC3931699 DOI: 10.2147/sar.s37907] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alcohol use disorders (AUD) continue to be a concerning health issue worldwide. Harmful alcohol use leads to 2.5 million deaths annually worldwide. Multiple options exist for the management of dependence on alcohol, not all of which are approved by drug-regulating agencies. Current practice in treating AUD does not reflect the diversity of pharmacologic options that have potential to provide benefit, and guidance for clinicians is limited. Few medications are approved for treatment of AUD, and these have exhibited small and/or inconsistent effects in broad patient populations with diverse drinking patterns. The need for continued research into the treatment of this disease is evident in order to provide patients with more specific and effective options. This review describes the neurobiological mechanisms of AUD that are amenable to treatment and drug therapies that target pathophysiological conditions of AUD to reduce drinking. In addition, current literature on pharmacologic (both approved and non-approved) treatment options for AUD offered in the United States and elsewhere are reviewed. The aim is to inform clinicians regarding the options for alcohol abuse treatment, keeping in mind that not all treatments are completely successful in reducing craving or heavy drinking or increasing abstinence.
Collapse
Affiliation(s)
- Robin C Wackernah
- Department of Pharmacy Practice, School of Pharmacy, Rueckert-Hartman College for Health Professions, Regis University, Denver, CO, USA
| | - Matthew J Minnick
- Department of Pharmacy Practice, School of Pharmacy, Rueckert-Hartman College for Health Professions, Regis University, Denver, CO, USA
| | - Peter Clapp
- Department of Pharmaceutical Sciences, School of Pharmacy, Rueckert-Hartman College for Health Professions, Regis University, Denver, CO, USA
| |
Collapse
|
50
|
Sommer DB, Stacy MA. What’s in the pipeline for the treatment of Parkinson’s disease? Expert Rev Neurother 2014; 8:1829-39. [DOI: 10.1586/14737175.8.12.1829] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|