1
|
Li Y, Yang L, Hao D, Chen Y, Ye-Lin Y, Li CSR, Li G. Functional Networks of Reward and Punishment Processing and Their Molecular Profiles Predicting the Severity of Young Adult Drinking. Brain Sci 2024; 14:610. [PMID: 38928610 PMCID: PMC11201596 DOI: 10.3390/brainsci14060610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/15/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Alcohol misuse is associated with altered punishment and reward processing. Here, we investigated neural network responses to reward and punishment and the molecular profiles of the connectivity features predicting alcohol use severity in young adults. We curated the Human Connectome Project data and employed connectome-based predictive modeling (CPM) to examine how functional connectivity (FC) features during wins and losses are associated with alcohol use severity, quantified by Semi-Structured Assessment for the Genetics of Alcoholism, in 981 young adults. We combined the CPM findings and the JuSpace toolbox to characterize the molecular profiles of the network connectivity features of alcohol use severity. The connectomics predicting alcohol use severity appeared specific, comprising less than 0.12% of all features, including medial frontal, motor/sensory, and cerebellum/brainstem networks during punishment processing and medial frontal, fronto-parietal, and motor/sensory networks during reward processing. Spatial correlation analyses showed that these networks were associated predominantly with serotonergic and GABAa signaling. To conclude, a distinct pattern of network connectivity predicted alcohol use severity in young adult drinkers. These "neural fingerprints" elucidate how alcohol misuse impacts the brain and provide evidence of new targets for future intervention.
Collapse
Affiliation(s)
- Yashuang Li
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, 100 Pingleyuan, Beijing 100124, China; (Y.L.)
| | - Lin Yang
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, 100 Pingleyuan, Beijing 100124, China; (Y.L.)
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
- BJUT-UPV Joint Research Laboratory in Biomedical Engineering, 46022 Valencia, Spain
| | - Dongmei Hao
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, 100 Pingleyuan, Beijing 100124, China; (Y.L.)
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
- BJUT-UPV Joint Research Laboratory in Biomedical Engineering, 46022 Valencia, Spain
| | - Yu Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA (C.-S.R.L.)
| | - Yiyao Ye-Lin
- BJUT-UPV Joint Research Laboratory in Biomedical Engineering, 46022 Valencia, Spain
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, 46022 Valencia, Spain
| | - Chiang-Shan Ray Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA (C.-S.R.L.)
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06520, USA
- Wu Tsai Institute, Yale University, New Haven, CT 06511, USA
| | - Guangfei Li
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, 100 Pingleyuan, Beijing 100124, China; (Y.L.)
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
- BJUT-UPV Joint Research Laboratory in Biomedical Engineering, 46022 Valencia, Spain
| |
Collapse
|
2
|
Arnold ME, Schank JR. Sex differences in neuronal activation in the cortex and midbrain during quinine-adulterated alcohol intake. Alcohol Alcohol 2024; 59:agae031. [PMID: 38742547 PMCID: PMC11091839 DOI: 10.1093/alcalc/agae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
AIMS Continued alcohol consumption despite negative consequences is a core symptom of alcohol use disorder. This is modeled in mice by pairing negative stimuli with alcohol, such as adulterating alcohol solution with quinine. Mice consuming alcohol under these conditions are considered to be engaging in aversion-resistant intake. Previously, we have observed sex differences in this behavior, with females more readily expressing aversion-resistant consumption. We also identified three brain regions that exhibited sex differences in neuronal activation during quinine-alcohol drinking: ventromedial prefrontal cortex (vmPFC), posterior insular cortex (PIC), and ventral tegmental area (VTA). Specifically, male mice showed increased activation in vmPFC and PIC, while females exhibited increased activation in VTA. In this study, we aimed to identify what specific type of neurons are activated in these regions during quinine-alcohol drinking. METHOD We assessed quinine-adulterated alcohol intake using the two-bottle choice procedure. We also utilized RNAscope in situ hybridization in the three brain regions that previously exhibited a sex difference to examine colocalization of Fos, glutamate, GABA, and dopamine. RESULT Females showed increased aversion-resistant alcohol consumption compared to males. We also found that males had higher colocalization of glutamate and Fos in vmPFC and PIC, while females had greater dopamine and Fos colocalization in the VTA. CONCLUSIONS Collectively, these experiments suggest that glutamatergic output from the vmPFC and PIC may have a role in suppressing, and dopaminergic activity in the VTA may promote, aversion-resistant alcohol consumption. Future experiments will examine neuronal circuits that contribute to sex differences in aversion resistant consumption.
Collapse
Affiliation(s)
- Miranda E Arnold
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Drive, Athens, GA 30602, United States
| | - Jesse R Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Drive, Athens, GA 30602, United States
| |
Collapse
|
3
|
Morris J, Boness CL, Burton R. (Mis)understanding alcohol use disorder: Making the case for a public health first approach. Drug Alcohol Depend 2023; 253:111019. [PMID: 37952353 PMCID: PMC11061885 DOI: 10.1016/j.drugalcdep.2023.111019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 11/14/2023]
Abstract
'Alcohol use disorder' (AUD) is used by several contemporary conceptualizations to identify, treat and prevent problems associated with alcohol use. Such conceptualizations encompass diagnostic classifications and broader frameworks for policy and practice. However, current AUD concepts are subject to multiple tensions and limitations in capturing and responding to the complex and heterogeneous nature of alcohol problems. Further, public understandings of alcohol problems are heavily divergent from professional AUD concepts and remain embedded within an 'alcoholism' master narrative in which disease model stereotypes come with multiple costs for prevention and 'recovery'. The persistence of a problematic 'alcoholism' paradigm reflects the coalescing of multiple forces including the cognitive appeal of reductionism, motives to stigmatize and 'other', and an over-emphasis on AUD as an individually located biomedical problem. Public misperceptions of AUD as a matter of the individual, the individual's essence, and misconceived notions of responsibility and control have been bolstered by industry interests and the ascension of neuroscience and genetics, in turn diverting attention from the importance of the environmental and commercial determinants of health and the effectiveness of under-utilized public health policies. We call for multiple stakeholders to support efforts to prioritize a public health first approach to advancing AUD research, policy and treatment in order to make significant advances in AUD prevention and treatment. We offer several recommendations to assist in shifting public understanding and scientific limitations in AUD concepts and responses.
Collapse
Affiliation(s)
- James Morris
- London South Bank University, Centre for Addictive Behaviours Research, UK.
| | - Cassandra L Boness
- University of New Mexico, Center on Alcohol, Substance use, And Addictions, USA
| | - Robyn Burton
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| |
Collapse
|
4
|
Maher EE, Strzelecki AM, Weafer JJ, Gipson CD. The importance of translationally evaluating steroid hormone contributions to substance use. Front Neuroendocrinol 2023; 69:101059. [PMID: 36758769 PMCID: PMC10182261 DOI: 10.1016/j.yfrne.2023.101059] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Clinically, women appear to be more susceptible to certain aspects of substance use disorders (SUDs). The steroid hormones 17β-estradiol (E2) and progesterone (Pg) have been linked to women-specific drug behaviors. Here, we review clinical and preclinical studies investigating how cycling ovarian hormones affect nicotine-, cocaine-, and opioid-related behaviors. We also highlight gaps in the literature regarding how synthetic steroid hormone use may influence drug-related behaviors. In addition, we explore how E2 and Pg are known to interact in brain reward pathways and provide evidence of how these interactions may influence drug-related behaviors. The synthesis of this review demonstrates the critical need to study women-specific factors that may influence aspects of SUDs, which may play important roles in addiction processes in a sex-specific fashion. It is important to understand factors that impact women's health and may be key to moving the field forward toward more efficacious and individualized treatment strategies.
Collapse
Affiliation(s)
- Erin E Maher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Ashley M Strzelecki
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Jessica J Weafer
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
5
|
Morris J, Boness CL, Witkiewitz K. Should we promote alcohol problems as a continuum? Implications for policy and practice. DRUGS (ABINGDON, ENGLAND) 2023; 31:271-281. [PMID: 38682086 PMCID: PMC11052541 DOI: 10.1080/09687637.2023.2187681] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/01/2023] [Indexed: 04/03/2023]
Abstract
The highly heterogeneous nature of alcohol use and problems has presented significant challenges to those attempting to understand, treat or prevent what is commonly termed alcohol use disorder (AUD). However, any attempts to capture this complex phenomenon, including the various current criterion of AUD, come with a number of limitations. One particular limitation has been how alcohol problems are represented or understood in ways which do not capture the broad spectrum of alcohol use and harms and the many potential routes to prevention, treatment, and recovery. One possible response to this has been proposed as more explicitly framing or conceptualizing a continuum model of alcohol use and harms. In this commentary, we attempt to identify the key implications of a continuum model for policy and practice, examining the historical and current context of alcohol problem classifications and models. We argue a continuum model of alcohol use and problems holds a number of advantages for advancing public health goals, but also some potential limitations, both of which require further examination.
Collapse
Affiliation(s)
- J Morris
- Centre for Addictive Behaviours Research, School of Applied Sciences, London South Bank University, United Kingdom
| | - C L Boness
- Center on Alcohol, Substance use, And Addictions, University of New Mexico, Albuquerque, New Mexico
| | - K Witkiewitz
- Center on Alcohol, Substance use, And Addictions, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
6
|
Binge-like Alcohol Exposure in Adolescence: Behavioural, Neuroendocrine and Molecular Evidence of Abnormal Neuroplasticity… and Return. Biomedicines 2021; 9:biomedicines9091161. [PMID: 34572345 PMCID: PMC8470908 DOI: 10.3390/biomedicines9091161] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 01/02/2023] Open
Abstract
Binge alcohol consumption among adolescents affects the developing neural networks underpinning reward and stress processing in the nucleus accumbens (NAc). This study explores in rats the long-lasting effects of early intermittent exposure to intoxicating alcohol levels at adolescence, on: (1) the response to natural positive stimuli and inescapable stress; (2) stress-axis functionality; and (3) dopaminergic and glutamatergic neuroadaptation in the NAc. We also assess the potential effects of the non-intoxicating phytocannabinoid cannabidiol, to counteract (or reverse) the development of detrimental consequences of binge-like alcohol exposure. Our results show that adolescent binge-like alcohol exposure alters the sensitivity to positive stimuli, exerts social and novelty-triggered anxiety-like behaviour, and passive stress-coping during early and prolonged withdrawal. In addition, serum corticosterone and hypothalamic and NAc corticotropin-releasing hormone levels progressively increase during withdrawal. Besides, NAc tyrosine hydroxylase levels increase at late withdrawal, while the expression of dopamine transporter, D1 and D2 receptors is dynamically altered during binge and withdrawal. Furthermore, the expression of markers of excitatory postsynaptic signaling—PSD95; Homer-1 and -2 and the activity-regulated spine-morphing proteins Arc, LIM Kinase 1 and FOXP1—increase at late withdrawal. Notably, subchronic cannabidiol, during withdrawal, attenuates social- and novelty-induced aversion and passive stress-coping and rectifies the hyper-responsive stress axis and NAc dopamine and glutamate-related neuroplasticity. Overall, the exposure to binge-like alcohol levels in adolescent rats makes the NAc, during withdrawal, a locus minoris resistentiae as a result of perturbations in neuroplasticity and in stress-axis homeostasis. Cannabidiol holds a promising potential for increasing behavioural, neuroendocrine and molecular resilience against binge-like alcohol harmful effects.
Collapse
|
7
|
Windisch KA, Morochnik M, Reed B, Kreek MJ. Nalmefene, a mu opioid receptor antagonist/kappa opioid receptor partial agonist, potentiates cocaine motivation but not intake with extended access self-administration in adult male mice. Neuropharmacology 2021; 192:108590. [PMID: 33974940 DOI: 10.1016/j.neuropharm.2021.108590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 11/30/2022]
Abstract
The mu opioid receptor antagonist/kappa opioid receptor (KOR) partial agonist nalmefene (NMF), a close structural analog of naltrexone (NTX), has been shown to reduce cocaine reward in preclinical models. Given the greater KOR potency and improved bioavailability compared to NTX, NMF may be a promising pharmacotherapeutic for cocaine use disorder (CUD). Here we examine the effects of NMF pretreatment on chronic daily extended access (4h) cocaine intravenous self-administration (IVSA) in adult male C57Bl/6J mice. METHODS separate groups of mice had daily 4h cocaine IVSA sessions (0.25 or 0.5 mg/kg/inf, FR1) for 14 days. Starting on day 8, mice were pretreated with NMF (0, 1, or 10 mg/kg) 30m before each session. A separate group of mice acquired cocaine IVSA [seven days FR1 then four FR3 of 4h daily sessions (0.5 mg/kg/inf)] prior to a single progressive ratio 3 session to examine the effect of 1 mg/kg NMF on cocaine motivation. RESULTS No significant effect of NMF pretreatment on cocaine intake was observed. Acute pretreatment of 1 mg/kg NMF significantly potentiated cocaine motivation as measured by progressive ratio breakpoint. CONCLUSIONS NMF did not significantly attenuate cocaine intake and increased motivation for cocaine suggesting that NMF may not be suitable for non-abstinent CUD patients. Further research is needed with KOR selective partial or full agonists to determine their effect on cocaine reinforcement.
Collapse
Affiliation(s)
- Kyle A Windisch
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| | - Michelle Morochnik
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Brian Reed
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
8
|
Could Agomelatine Play a Role to Treat Anhedonia in Heroin Dependence? ADDICTIVE DISORDERS & THEIR TREATMENT 2021. [DOI: 10.1097/adt.0000000000000245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
9
|
Kalafateli AL, Aranäs C, Jerlhag E. Activation of the amylin pathway modulates cocaine-induced activation of the mesolimbic dopamine system in male mice. Horm Behav 2021; 127:104885. [PMID: 33166561 DOI: 10.1016/j.yhbeh.2020.104885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022]
Abstract
Besides food intake reduction, activation of the amylin pathway by salmon calcitonin (sCT), an amylin and calcitonin receptor agonist, inhibits alcohol-mediated behaviors in rodents. This involves brain areas processing reward, i.e. the laterodorsal (LDTg), ventral tegmental area (VTA) and nucleus accumbens (NAc). However, the effects of stimulation of the amylin pathway on behaviors caused by cocaine and the brain areas involved in these processes have not yet been investigated. We therefore explored in male mice, the effects of systemic administration of sCT on cocaine-induced locomotor stimulation, dopamine release in the NAc and cocaine reward, as well as reward-dependent memory of cocaine, in the conditioned place preference (CPP) paradigm. Moreover, the outcome of systemic sCT and cocaine co-administration for five days on locomotor activity was investigated. Lastly, the impact of sCT infusions into the LDTg, VTA, NAc shell or core on cocaine-evoked locomotor stimulation was explored. We found that sCT attenuated cocaine-induced locomotor stimulation and accumbal dopamine release, without altering cocaine's rewarding properties or reward-dependent memory retrieval in the CPP paradigm. Five days of cocaine administration caused locomotor stimulation in mice pre-treated with vehicle, but not with sCT. In mice infused with vehicle into the aforementioned reward-related areas, cocaine caused locomotor stimulation, a response that was not evident following sCT infusions. The current findings suggest a novel role for the amylinergic pathway as regulator of cocaine-evoked activation of the mesolimbic dopamine system, opening the way for the investigation of the amylin signalling in the modulation of other drugs of abuse.
Collapse
Affiliation(s)
- Aimilia Lydia Kalafateli
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cajsa Aranäs
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
10
|
Demirel G, Guzel E, Creighton CJ, Ozturk YE, Kucuk C, Asliyuksek H, Yurdun T. MDMA Abuse in Relation to MicroRNA Variation in Human Brain Ventral Tegmental Area and Nucleus Accumbens. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 18:1989-1999. [PMID: 32184864 PMCID: PMC7059052 DOI: 10.22037/ijpr.2019.15097.12874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
3,4-methylenedioxymethamphetamine (MDMA) is one of the most widespread illegal drugs, that have been used particularly by young people in the 15-34 age group. MicroRNAs (miRNAs) are endogenously synthesized, non-coding, and small RNAs that post-transcriptionally regulate their target genes' expression by inhibiting protein translation or degradation. miRNAs are increasingly implicated in drug-related gene expressions and functions. Notably, there are no reports of miRNA variation in the human brain in MDMA abuse. We here present a miRNA profiling study - the first such study, to the best of our knowledge - into the post-mortem human brains of a sample of people with MDMA abuse, along with non-drug dependent controls. The miRNA profiling of nucleus accumbens (NAc) and ventral tegmental areas (VTA) was performed by microarray analysis. Subsequently, two candidate miRNA putative biomarkers were selected according to significant regional differential expression (miR-1202 and miR-7975), using quantitative reverse-transcription PCR (qRT-PCR). We showed that the expression level of miR-7975 was significantly lower in the VTA regions of the 30 MDMA users, as compared with the 30 control samples. Another significantly deregulated miR-1202 was down-regulated in the NAc regions of 30 MDMA samples in comparison to the control samples. Alteration of these miRNAs can potentially serve as novel biomarkers for MDMA abuse, and warrant further research in independent and larger samples of patients.
Collapse
Affiliation(s)
- Goksun Demirel
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Biruni University, Istanbul, Turkey.,Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Çukurova University, Adana, Turkey.,G D and E G. These authors contributed equally to this work
| | - Esra Guzel
- Department of Molecular Biology and Genetics, Institute of Health Sciences, University of Health Sciences, Istanbul, Turkey.,G D and E G. These authors contributed equally to this work
| | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Cancer Center Division of Biostatistics, Houston, Texas
| | - Yeter Erol Ozturk
- Chemistry Department, Council of Forensic Medicine, Istanbul, Turkey
| | | | | | - Türkan Yurdun
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| |
Collapse
|
11
|
Shyness, alcohol use disorders and ‘hangxiety’: A naturalistic study of social drinkers. PERSONALITY AND INDIVIDUAL DIFFERENCES 2019. [DOI: 10.1016/j.paid.2018.10.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
12
|
Gómez-Coronado N, Sethi R, Bortolasci CC, Arancini L, Berk M, Dodd S. A review of the neurobiological underpinning of comorbid substance use and mood disorders. J Affect Disord 2018; 241:388-401. [PMID: 30145509 DOI: 10.1016/j.jad.2018.08.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/09/2018] [Accepted: 08/10/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND There is evidence that substance use disorders and other mental disorders may have shared biological mechanisms. However, the neurobiological basis of this comorbidity remains only partially explained. This review describes the historical evolution of the dual disorders concept and approach, and reviews the existing literature on neurobiological findings specifically regarding comorbid substance use and mood disorders. METHODS Searches were conducted using PubMed and Scopus in December 2017. A Boolean search was performed using combinations of "dual diagnosis" or "dual disorder" or "depression" or "bipolar" or "affective disorder" or "mood disorder" and "substance use" or "substance abuse" and "neurobiology" or "functional neuroimaging" or "genetics" or "neurotransmitters" or "neuroendocrinology" in the title or abstract, or as keywords, using no language restriction. RESULTS 32 studies met the inclusion criteria. We found robust evidence for involvement of the neurotransmitters dopamine, GABA and glutamate and their receptors, as well as by the central corticotrophin-releasing hormone, hypothalamic-pituitary-adrenal axis activation, oxidative stress and inflammation. Recent studies focusing on neuroimaging and genetics have not shown consistent results. LIMITATIONS Only two search tools were used; most identified studies excluded the population of interest (comorbid mood and substance abuse disorders). CONCLUSIONS The neurobiological relevance for the occurrence of comorbid mood and substance abuse disorders has not been fully elucidated. Considering the high levels of individuals who experience comorbidity in these areas as well as the negative associated outcomes, this is clearly an area that requires further in-depth investigation. Furthermore, findings from this area can help to inform drug abuse prevention and intervention efforts, and especially how they relate to populations with psychiatric symptoms.
Collapse
Affiliation(s)
- Nieves Gómez-Coronado
- Unidad de Gestión Clínica Salud Mental, Hospital Universitario Virgen del Rocío, Sevilla, Spain; Department of Psychiatry, Hospital San Agustín ORL, Dos Hermanas, Sevilla, Spain
| | - Rickinder Sethi
- London Health Sciences Centre, Western University, London, Canada
| | - Chiara Cristina Bortolasci
- Centre for Molecular and Medical Research, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Lauren Arancini
- IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia; University Hospital Geelong, Barwon Health, Geelong VIC, Australia; Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia; Orygen, The National Centre for Excellence in Youth Mental Health, Parkville, VIC, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Seetal Dodd
- IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia; University Hospital Geelong, Barwon Health, Geelong VIC, Australia; Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia; Orygen, The National Centre for Excellence in Youth Mental Health, Parkville, VIC, Australia.
| |
Collapse
|
13
|
Impact of neuroimmune activation induced by alcohol or drug abuse on adolescent brain development. Int J Dev Neurosci 2018; 77:89-98. [PMID: 30468786 DOI: 10.1016/j.ijdevneu.2018.11.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/19/2018] [Accepted: 11/19/2018] [Indexed: 12/27/2022] Open
Abstract
Evidence obtained in recent decades has demonstrated that the brain still matures in adolescence. Changes in neural connectivity occur in different regions, including cortical and subcortical structures, which undergo modifications in white and gray matter densities. These alterations concomitantly occur in some neurotransmitter systems and hormone secretion, which markedly influence the refinement of certain brain areas and neural circuits. The immaturity of the adolescent brain makes it more vulnerable to the effects of alcohol and drug abuse, whose use can trigger long-term behavioral dysfunction. This article reviews the action of alcohol and drug abuse (cannabis, cocaine, opioids, amphetamines, anabolic androgenic steroids) in the adolescent brain, and their impact on both cognition and behavioral dysfunction, including predisposition to drug abuse in later life. It also discusses recent evidence that indicates the role of the neuroimmune system response and neuroinflammation as mechanisms that participate in many actions of ethanol and drug abuse in adolescence, including the neurotoxicity and alterations in neurocircuitry that contribute to the dysfunctional behaviors associated with addiction. The new data suggest the therapeutic potential of anti-inflammatory targets to prevent the long-term consequences of drug abuse in adolescence.
Collapse
|
14
|
Pascual M, Montesinos J, Guerri C. Role of the innate immune system in the neuropathological consequences induced by adolescent binge drinking. J Neurosci Res 2017; 96:765-780. [PMID: 29214654 DOI: 10.1002/jnr.24203] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/25/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022]
Abstract
Adolescence is a critical stage of brain maturation in which important plastic and dynamic processes take place in different brain regions, leading to development of the adult brain. Ethanol drinking in adolescence disrupts brain plasticity and causes structural and functional changes in immature brain areas (prefrontal cortex, limbic system) that result in cognitive and behavioral deficits. These changes, along with secretion of sexual and stress-related hormones in adolescence, may impact self-control, decision making, and risk-taking behaviors that contribute to anxiety and initiation of alcohol consumption. New data support the participation of the neuroimmune system in the effects of ethanol on the developing and adult brain. This article reviews the potential pathological bases that underlie the effects of alcohol on the adolescent brain, such as the contribution of genetic background, the perturbation of epigenetic programming, and the influence of the neuroimmune response. Special emphasis is given to the actions of ethanol in the innate immune receptor toll-like receptor 4 (TLR4), since recent studies have demonstrated that by activating the inflammatory TLR4/NFκB signaling response in glial cells, binge drinking of ethanol triggers the release of cytokines/chemokines and free radicals, which exacerbate the immune response that causes neuroinflammation/neural damage as well as short- and long-term neurophysiological, cognitive, and behavioral dysfunction. Finally, potential treatments that target the neuroimmune response to treat the neuropathological and behavioral consequences of adolescent alcohol abuse are discussed.
Collapse
Affiliation(s)
- María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, Valencia, Spain
| | - Jorge Montesinos
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, Valencia, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, Valencia, Spain
| |
Collapse
|
15
|
Zamroziewicz M, Raskin SA, Tennen H, Austad CS, Wood RM, Fallahi CR, Dager AD, Sawyer B, Leen S, Pearlson GD. Effects of drinking patterns on prospective memory performance in college students. Neuropsychology 2017; 31:191-199. [PMID: 27841457 PMCID: PMC5280574 DOI: 10.1037/neu0000313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE Traditional college students are at a critical juncture in the development of prospective memory (PM). Their brains are vulnerable to the effects of alcohol. METHOD There were 123 third and fourth year college students, 19-23 years old, who completed the Self-Rating Effects of Alcohol (SREA), Modified Timeline Follow-back (TFLB), Brief Young Adult Alcohol Consequences Scale (BYAACS), and Alcohol Effects Questionnaire (AEQ) once per month on a secure online database, as reported elsewhere (Dager et al., 2013). Data from the 6 months immediately before memory testing were averaged. In a single testing session participants were administered the Mini International Neuropsychiatric Interview-Diagnostic and Statistical Manual for Mental Disorders-Fourth Edition-Text Revision (MINI-DSM-IV-TR), measures of PM (event-based and time-based), and retrospective memory (RM). Based on the average score of six consecutive monthly responses to the SREA, TLFB, and AEQ, students were classified as nondrinkers, light drinkers, or heavy drinkers (as defined previously; Dager et al., 2013). Alcohol-induced amnesia (blackout) was measured with the BYAACS. RESULTS We found a relationship between these alcohol use classifications and time-based PM, such that participants who were classified as heavier drinkers were more likely to forget to perform the time-based PM task. We also found that self-reported alcohol-induced amnesia (blackouts) during the month immediately preceding memory testing was associated with lower performance on the event-based PM task. Participants' ability to recall the RM tasks suggested the PM items were successfully encoded even when they were not carried out, and we observed no relationship between alcohol use and RM performance. CONCLUSION Heavy alcohol use in college students may be related to impairments in PM. (PsycINFO Database Record
Collapse
Affiliation(s)
| | | | - Howard Tennen
- Department of Community Medicine and Health Care, University of Connecticut School of Medicine
| | - Carol S Austad
- Department of Psychology, Central Connecticut State University
| | - Rebecca M Wood
- Department of Psychology, Central Connecticut State University
| | | | | | | | | | | |
Collapse
|
16
|
Van't Veer A, Smith KL, Cohen BM, Carlezon WA, Bechtholt AJ. Kappa-opioid receptors differentially regulate low and high levels of ethanol intake in female mice. Brain Behav 2016; 6:e00523. [PMID: 27688945 PMCID: PMC5036438 DOI: 10.1002/brb3.523] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/21/2016] [Accepted: 05/28/2016] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Studies in laboratory animals and humans indicate that endogenous opioids play an important role in regulating the rewarding value of various drugs, including ethanol (EtOH). Indeed, opioid antagonists are currently a front-line treatment for alcoholism in humans. Although roles for mu- and delta-opioid receptors have been characterized, the contribution of kappa-opioid receptors (KORs) is less clear. There is evidence that changes in KOR system function can decrease or increase EtOH drinking, depending on test conditions. For example, female mice lacking preprodynorphin - the precursor to the endogenous KOR ligand dynorphin - have reduced EtOH intake. Considering that KORs can regulate dopamine (DA) transmission, we hypothesized that KORs expressed on DA neurons would play a prominent role in EtOH intake in females. METHODS We used a Cre/loxP recombination strategy to ablate KORs throughout the body or specifically on dopamine uptake transporter (DAT)-expressing neurons to investigate the role of KORs on preference for and intake of EtOH (2-bottle choice), the transition from moderate to excessive EtOH drinking (intermittent EtOH access), and binge EtOH drinking (drinking in the dark [DID]). RESULTS KOR deletion decreased preference for EtOH, although this effect was less pronounced when EtOH intake increased beyond relatively low levels. DISCUSSION Our findings indicate that KOR activation increases EtOH drinking via effects mediated, at least in part, by KORs on DA neurons. While the mechanisms of this regulation remain unknown, previous work suggests that alterations in negative reinforcement processes or sensitivity to the sensory properties of EtOH can affect preference and intake.
Collapse
Affiliation(s)
- Ashlee Van't Veer
- Department of Psychiatry Harvard Medical School McLean Hospital Belmont MA USA; National Institute of Neurological Disorders and Stroke National Institutes of Health Bethesda MD USA
| | - Karen L Smith
- Department of Psychiatry Harvard Medical School McLean Hospital Belmont MA USA
| | - Bruce M Cohen
- Department of Psychiatry Harvard Medical School McLean Hospital Belmont MA USA
| | - William A Carlezon
- Department of Psychiatry Harvard Medical School McLean Hospital Belmont MA USA
| | - Anita J Bechtholt
- Department of Psychiatry Harvard Medical School McLean Hospital Belmont MA USA; National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Bethesda MD USA
| |
Collapse
|
17
|
Brumback T, Castro N, Jacobus J, Tapert S. Effects of Marijuana Use on Brain Structure and Function: Neuroimaging Findings from a Neurodevelopmental Perspective. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 129:33-65. [PMID: 27503447 DOI: 10.1016/bs.irn.2016.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Marijuana, behind only tobacco and alcohol, is the most popular recreational drug in America with prevalence rates of use rising over the past decade. A wide range of research has highlighted neurocognitive deficits associated with marijuana use, particularly when initiated during childhood or adolescence. Neuroimaging, describing alterations to brain structure and function, has begun to provide a picture of possible mechanisms associated with the deleterious effects of marijuana use. This chapter provides a neurodevelopmental framework from which recent data on brain structural and functional abnormalities associated with marijuana use is reviewed. Based on the current data, we provide aims for future studies to more clearly delineate the effects of marijuana on the developing brain and to define underlying mechanisms of the potential long-term negative consequences of marijuana use.
Collapse
Affiliation(s)
- T Brumback
- University of California, San Diego, La Jolla, CA, United States; VA San Diego Healthcare System, San Diego, CA, United States.
| | - N Castro
- University of California, San Diego, La Jolla, CA, United States
| | - J Jacobus
- University of California, San Diego, La Jolla, CA, United States; VA San Diego Healthcare System, San Diego, CA, United States
| | - S Tapert
- University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
18
|
Abstract
Psychostimulants are a diverse group of substances that cause an increase in psychomotor activity at least in part through their actions on catecholaminergic systems including the dopaminergic mesolimbic pathways. Animal models used to study addiction are based on the psychomotor stimulant theory of addiction. The basics of this theory are that the reinforcing effects and the addition liabilities of the drugs can be predicted from their ability to induce psychomotor activation. This approach focuses on the ability of the drugs to directly control the animal's behavior and to induce psychomotor stimulation, and is consistent with the behavioral definition of addiction and behavioral sensitization. Animal experiments have the advantage over clinical studies of lower variation and fewer confounding effects.
Collapse
|
19
|
Oliveto A, Mancino M, Sanders N, Cargile C, Benjamin Guise J, Bickel W, Brooks Gentry W. Effects of prototypic calcium channel blockers in methadone-maintained humans responding under a naloxone discrimination procedure. Eur J Pharmacol 2013; 715:424-35. [PMID: 23524089 DOI: 10.1016/j.ejphar.2013.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 03/07/2013] [Accepted: 03/07/2013] [Indexed: 11/28/2022]
Abstract
Accumulating evidence suggests that L-type calcium channel blockers (CCBs) attenuate the expression of opioid withdrawal and the dihydropyridine L-type CCB isradipine has been shown to block the behavioral effects of naloxone in opioid-maintained humans. This study determined whether two prototypic L-type CCBs with differing chemical structures, the benzothiazepine diltiazem and the phenylalkamine verapamil, attenuate the behavioral effects of naloxone in methadone-maintained humans trained to distinguish between low-dose naloxone (0.15 mg/70 kg, i.m.) and placebo under an instructed novel-response drug discrimination procedure. Once discrimination was acquired, diltiazem (0, 30, 60, 120 mg) and verapamil (0, 30, 60, 120 mg), alone and combined with the training dose of naloxone, were tested. Diltiazem alone produced 33-50% naloxone- and novel-appropriate responding at 30 and 60 mg and essentially placebo-appropriate responding at 120 mg. Verapamil alone produced 20-40% naloxone- and 0% novel-appropriate responding. Diltiazem at 60 mg decreased several ratings associated with positive mood and increased VAS ratings of "Bad Drug Effects" relative to placebo, whereas verapamil increased ratings associated with euphoria. When administered with naloxone, diltiazem produced 94-100% naloxone-appropriate-responding with 6% novel-appropriate responding at 60 mg (n=3). When administered with naloxone, verapamil produced 60-80% naloxone- and 0% novel-appropriate responding (n=5). Diltiazem decreased diastolic blood pressure and heart rate whereas verapamil decreased ratings of arousal relative to placebo. These results suggest that CCBs with different chemical structures can be differentiated behaviorally, and that diltiazem and verapamil do not attenuate the discriminative stimulus effects of naloxone in humans at the doses tested.
Collapse
Affiliation(s)
- Alison Oliveto
- Department of Psychiatry and Behavioral Sciences, University of Arkansas for Medical Sciences, 4301 W Markham St., Little Rock, AR 72205, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Marie N, Noble F. Dépendance aux drogues : avancées de la neurobiologie et perspectives thérapeutiques. Presse Med 2012; 41:1259-70. [DOI: 10.1016/j.lpm.2012.07.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/31/2012] [Indexed: 02/08/2023] Open
|
21
|
Sirohi S, Bakalkin G, Walker BM. Alcohol-induced plasticity in the dynorphin/kappa-opioid receptor system. Front Mol Neurosci 2012; 5:95. [PMID: 23060746 PMCID: PMC3459013 DOI: 10.3389/fnmol.2012.00095] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 08/29/2012] [Indexed: 01/02/2023] Open
Abstract
Alcoholism is a chronic relapsing disorder characterized by continued alcohol use despite numerous adverse consequences. Alcohol has been shown to interact with numerous neurotransmitter systems to exert its pharmacological effects. The endogenous opioid system (EOS) has been strongly implicated in the positive and negative reinforcing effects of alcohol. Traditionally recognized as dysphoric/anhedonic in nature, the dynorphin/kappa-opioid receptor (DYN/KOR) system has recently received considerable attention due to evidence suggesting that an upregulated DYN/KOR system may be a critical contributor to the complex factors that result in escalated alcohol consumption once dependent. The present review will discuss alcohol-induced plasticity in the DYN/KOR system and how these neuroadaptations could contribute to excessive alcohol seeking and consumption.
Collapse
Affiliation(s)
- Sunil Sirohi
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University Pullman, WA, USA
| | | | | |
Collapse
|
22
|
Involvement of nucleus accumbens dopamine D1 receptors in ethanol drinking, ethanol-induced conditioned place preference, and ethanol-induced psychomotor sensitization in mice. Psychopharmacology (Berl) 2012; 222:141-53. [PMID: 22222864 DOI: 10.1007/s00213-011-2630-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 12/19/2011] [Indexed: 12/24/2022]
Abstract
RATIONALE Dopamine D1 receptor (D1R) signaling has been associated to ethanol consumption and reward in laboratory animals. OBJECTIVES Here, we hypothesize that this receptor, which is located within the nucleus accumbens (NAc) neurons, modulates alcohol reward mechanisms. METHODS To test this hypothesis, we measured alcohol consumption and ethanol-induced psychomotor sensitization and conditioned place preference (CPP) in mice that received bilateral microinjections of small interference RNA (siRNA)-expressing lentiviral vectors (LV-siD1R) producing D1R knock-down. The other group received control (LV-Mock) viral vectors into the NAc. RESULTS There were no differences in the total fluid consumed and also no differences in the amount of ethanol consumed between groups prior to surgery. However, after surgery, the LV-siD1R group consumed less ethanol than the control group. This difference was not associated to taste neophobia. In addition, results have shown that down-regulation of endogenous D1R using viral-mediated siRNA in the NAc significantly decreased ethanol-induced behavioral sensitization as well as acquisition, but not expression, of ethanol-induced place preference. CONCLUSIONS We conclude that decreased D1R expression into the NAc led to reduced ethanol rewarding properties, thereby leading to lower voluntary ethanol consumption. Together, these findings demonstrate that the D1 receptor pathway within the NAc controls ethanol reward and intake.
Collapse
|
23
|
Conceptualizing withdrawal-induced escalation of alcohol self-administration as a learned, plasticity-dependent process. Alcohol 2012; 46:339-48. [PMID: 22459874 DOI: 10.1016/j.alcohol.2012.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/11/2012] [Accepted: 01/24/2012] [Indexed: 11/24/2022]
Abstract
This article represents one of five contributions focusing on the topic "Plasticity and neuroadaptive responses within the extended amygdala in response to chronic or excessive alcohol exposure" that were developed by awardees participating in the Young Investigator Award Symposium at the "Alcoholism and Stress: A Framework for Future Treatment Strategies" conference in Volterra, Italy on May 3-6, 2011 that was organized/chaired by Drs. Antonio Noronha and Fulton Crews and sponsored by the National Institute on Alcohol Abuse and Alcoholism. This review discusses the dependence-induced neuroadaptations in affective systems that provide a basis for negative reinforcement learning and presents evidence demonstrating that escalated alcohol consumption during withdrawal is a learned, plasticity-dependent process. The review concludes by identifying changes within extended amygdala dynorphin/kappa-opioid receptor systems that could serve as the foundation for the occurrence of negative reinforcement processes. While some evidence contained herein may be specific to alcohol dependence-related learning and plasticity, much of the information will be of relevance to any addictive disorder involving negative reinforcement mechanisms. Collectively, the information presented within this review provides a framework to assess the negative reinforcing effects of alcohol in a manner that distinguishes neuroadaptations produced by chronic alcohol exposure from the actual plasticity that is associated with negative reinforcement learning in dependent organisms.
Collapse
|
24
|
Pascual M, Do Couto BR, Alfonso-Loeches S, Aguilar MA, Rodriguez-Arias M, Guerri C. Changes in histone acetylation in the prefrontal cortex of ethanol-exposed adolescent rats are associated with ethanol-induced place conditioning. Neuropharmacology 2012; 62:2309-19. [PMID: 22349397 DOI: 10.1016/j.neuropharm.2012.01.011] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 01/04/2012] [Accepted: 01/15/2012] [Indexed: 01/08/2023]
Abstract
Alcohol drinking during adolescence can induce long-lasting effects on the motivation to consume alcohol. Abnormal plasticity in reward-related processes might contribute to the vulnerability of adolescents to drug addiction. We have shown that binge-like ethanol treatment in adolescent rats induces alterations in the dopaminergic system and causes histone modifications in brain reward regions. Considering that histone acetylation regulates transcriptional activity and contributes to drug-induced alterations in gene expression and behavior, we addressed the hypothesis that ethanol is capable of inducing transcriptional changes by histone modifications in specific gene promoters in adolescent brain reward regions, and whether these events are associated with acquisition of place conditioning. After treating juvenile and adult rats with intermittent ethanol administration, we found that ethanol treatment upregulates histone acetyl transferase (HAT) activity in adolescent prefrontal cortex and increases histone (H3 or H4) acetylation and H3(K4) dimethylation in the promoter region of cFos, Cdk5 and FosB. Inhibition of histone deacetylase by sodium butyrate before ethanol injection enhances both up-regulation of HAT activity and histone acetylation of cFos, Cdk5 and FosB. Furthermore, co-administration of sodium butyrate with ethanol prolongs the extinction of conditioned place aversion and increased the reinstatement effects of ethanol in ethanol-treated adolescents, but not in ethanol-treated adult rats. These results indicate that ethanol exposure during adolescence induces chromatin remodeling, changes histone acetylation and methylation, and modify the effects of ethanol on place conditioning. They also suggest that epigenetic mechanisms might open up avenues to new treatments for binge drinking-induced drug addiction during adolescence.
Collapse
Affiliation(s)
- María Pascual
- Department of Cell Pathology, Príncipe Felipe Research Center, Avda. Autopista del Saler, 16, 46012 Valencia, Spain
| | | | | | | | | | | |
Collapse
|
25
|
Pohorecky LA, Sweeny A. Amphetamine modifies ethanol intake of psychosocially stressed male rats. Pharmacol Biochem Behav 2012; 101:417-26. [PMID: 22285324 DOI: 10.1016/j.pbb.2012.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 01/10/2012] [Accepted: 01/14/2012] [Indexed: 11/19/2022]
Abstract
Studies of socially housed rodents have provided significant information regarding the consequences of exposure to stressors. Psychosocial stressors are known to alter the ingestion of ethanol and the activity of the dopaminergic neuronal system. Since both stressors and ethanol are known to affect the function of dopaminergic neurons, we employed amphetamine to assess the role of this neural system on the ingestion of ethanol by psychosocially stressed male rats. Male rats housed two per cage were designated as dominant or subdominant rats based on evaluations of agonistic behavior and body weight changes. The dyad-housed rats and a group of single-housed rats were sequentially assessed for ethanol intake after injections of saline or amphetamine (0.3, 0.9 or 2.7 mg/kg i.p.) both prior to dyad housing and subsequently again during dyad-housing. Prior to dyad housing ethanol intake of future subdominant rats was higher than that of future dominant rats. Dyad-housing significantly increased ethanol intake of dominant rats. Pre-dyad the highest dose of amphetamine potently depressed ethanol ingestion. Sensitivity to amphetamine's depressant effect on ethanol intake was higher at the dyad test in all subjects, most prominently in single-housed rats. In contrast to the single-housed rats, the dyad-housed rats displayed saccharin anhedonia. It can be concluded that dopaminergic system modulates, at least partially, the psychosocial stress-induced changes in ethanol intake. Furthermore, the level of ethanol ingestion at the pre-dyad test was predictive of future hierarchical status.
Collapse
|
26
|
Canales JJ. Deficient plasticity in the hippocampus and the spiral of addiction: focus on adult neurogenesis. Curr Top Behav Neurosci 2012; 15:293-312. [PMID: 22976276 DOI: 10.1007/7854_2012_230] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Addiction is a complex neuropsychiatric disorder which causes disruption at multiple levels, including cognitive, emotional, and behavioral domains. Traditional biological theories of addiction have focused on the mesolimbic dopamine pathway and the nucleus accumbens as anatomical substrates mediating addictive-like behaviors. More recently, we have begun to recognize the engagement and dynamic influence of a much broader circuitry which encompasses the frontal cortex, the amygdala, and the hippocampus. In particular, neurogenesis in the adult hippocampus has become a major focus of attention due to its ability to influence memory, motivation, and affect, all of which are disrupted in addiction. First, I summarize toxicological data that reveal strongly suppressive effects of drug exposure on adult hippocampal neurogenesis. Then, I discuss the impact of deficient neurogenesis on learning and memory function, stress responsiveness and affective behavior, as they relate to addiction. Finally, I examine recent behavioral observations that implicate neurogenesis in the adult hippocampus in the emergence and maintenance of addictive behavior. The evidence reviewed here suggests that deficient neurogenesis is associated with several components of the downward spiraling loop that characterizes addiction, including elevated sensitivity to drug-induced reward and reinforcement, enhanced neurohormonal responsiveness, emergence of a negative affective state, memory impairment, and inflexible behavior.
Collapse
Affiliation(s)
- Juan J Canales
- Behavioural Neuroscience, Department of Psychology, The University of Canterbury, Private Bag 4800, 8140, Christchurch, New Zealand,
| |
Collapse
|
27
|
Bahi A, Dreyer JL. Involvement of tissue plasminogen activator “tPA” in ethanol-induced locomotor sensitization and conditioned-place preference. Behav Brain Res 2012; 226:250-8. [DOI: 10.1016/j.bbr.2011.09.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 08/25/2011] [Accepted: 09/12/2011] [Indexed: 12/26/2022]
|
28
|
Alfonso-Loeches S, Guerri C. Molecular and behavioral aspects of the actions of alcohol on the adult and developing brain. Crit Rev Clin Lab Sci 2011; 48:19-47. [PMID: 21657944 DOI: 10.3109/10408363.2011.580567] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The brain is one of the major target organs of alcohol actions. Alcohol abuse can lead to alterations in brain structure and functions and, in some cases, to neurodegeneration. Cognitive deficits and alcohol dependence are highly damaging consequences of alcohol abuse. Clinical and experimental studies have demonstrated that the developing brain is particularly vulnerable to alcohol, and that drinking during gestation can lead to a range of physical, learning and behavioral defects (fetal alcohol spectrum disorders), with the most dramatic presentation corresponding to fetal alcohol syndrome. Recent findings also indicate that adolescence is a stage of brain maturation and that heavy drinking at this stage can have a negative impact on brain structure and functions causing important short- and long-term cognitive and behavioral consequences. The effects of alcohol on the brain are not uniform; some brain areas or cell populations are more vulnerable than others. The prefrontal cortex, the hippocampus, the cerebellum, the white matter and glial cells are particularly susceptible to the effects of ethanol. The molecular actions of alcohol on the brain are complex and involve numerous mechanisms and signaling pathways. Some of the mechanisms involved are common for the adult brain and for the developing brain, while others depend on the developmental stage. During brain ontogeny, alcohol causes irreversible alterations to the brain structure. It also impairs several molecular, neurochemical and cellular events taking place during normal brain development, including alterations in both gene expression regulation and the molecules involved in cell-cell interactions, interference with the mitogenic and growth factor response, enhancement of free radical formation and derangements of glial cell functions. However, in both adult and adolescent brains, alcohol damages specific brain areas through mechanisms involving excitotoxicity, free radical formation and neuroinflammatory damage resulting from activation of the innate immune system mediated by TLR4 receptors. Alcohol also acts on specific membrane proteins, such as neurotransmitter receptors (e.g. NMDA, GABA-A), ion channels (e.g. L-type Ca²⁺ channels, GIRKs), and signaling pathways (e.g. PKA and PKC signaling). These effects might underlie the wide variety of behavioral effects induced by ethanol drinking. The neuroadaptive changes affecting neurotransmission systems which are more sensitive to the acute effects of alcohol occur after long-term alcohol consumption. Alcohol-induced maladaptations in the dopaminergic mesolimbic system, abnormal plastic changes in the reward-related brain areas and genetic and epigenetic factors may all contribute to alcohol reinforcement and alcohol addiction. This manuscript reviews the mechanisms by which ethanol impacts the adult and the developing brain, and causes both neural impairments and cognitive and behavioral dysfunctions. The identification and the understanding of the cellular and molecular mechanisms involved in ethanol toxicity might contribute to the development of treatments and/or therapeutic agents that could reduce or eliminate the deleterious effects of alcohol on the brain.
Collapse
|
29
|
Morzorati SL, Marunde RL, Downey D. Limited access to ethanol increases the number of spontaneously active dopamine neurons in the posterior ventral tegmental area of nondependent P rats. Alcohol 2010; 44:257-64. [PMID: 20682193 DOI: 10.1016/j.alcohol.2010.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 01/28/2010] [Accepted: 02/10/2010] [Indexed: 10/19/2022]
Abstract
Microdialysis experiments in alcohol-preferring (P) rats have shown that chronic ethanol exposure increases extracellular levels of dopamine (DA) in the nucleus accumbens. Because DA neuronal activity contributes to the regulation of DA overflow in terminal regions, we hypothesized that posterior ventral tegmental area (VTA) DA neuronal activity (firing frequency, burst activity, and/or the number of spontaneously active DA neurons) would be increased in P rats consuming ethanol compared with P rats consuming only water. In vivo electrophysiological techniques were used to evaluate the activity of single DA neurons in the posterior VTA. Our findings show that voluntary ethanol intake by nondependent P rats significantly increased the number of spontaneously active DA neurons in the posterior VTA compared with P rats that consumed only water. Firing frequency and burst activity did not differ between the two groups. These results suggest that adaptive changes occur in the mesolimbic DA system of nondependent P rats to increase the excitability of posterior VTA DA neurons and enhance DA release from nerve terminals in the nucleus accumbens.
Collapse
|
30
|
Vaz de Lima FB, Andriolo RB, da Silveira DX. Dopaminergic antagonists for alcohol dependence. Hippokratia 2010. [DOI: 10.1002/14651858.cd008460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | - Régis B Andriolo
- Universidade Federal de São Paulo; Emergency Medicine and Evidence Based Medicine; Rua Pedro de Toledo 598 São Paulo Brazil 04039-001
| | - Dartiu X da Silveira
- Federal University of Sao Paulo; Department of Psychiatry; Rua dos Otonis, 887 Sao Paulo Sao Paulo Brazil 04601-040
| |
Collapse
|
31
|
Guerri C, Pascual M. Mechanisms involved in the neurotoxic, cognitive, and neurobehavioral effects of alcohol consumption during adolescence. Alcohol 2010; 44:15-26. [PMID: 20113871 DOI: 10.1016/j.alcohol.2009.10.003] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 09/29/2009] [Accepted: 10/01/2009] [Indexed: 01/13/2023]
Abstract
Studies over the last decade demonstrate that adolescence is a brain maturation period from childhood to adulthood. Plastic and dynamic processes drive adolescent brain development, creating flexibility that allows the brain to refine itself, specialize, and sharpen its functions for specific demands. Maturing connections enable increased communication among brain regions, allowing greater integration and complexity. Compelling evidence has shown that the developing brain is vulnerable to the damaging effects of ethanol. It is possible to infer, therefore, that alcohol exposure during the critical adolescent developmental stages could disrupt the brain plasticity and maturation processes, resulting in behavioral and cognitive deficits. Recent neuroimaging studies have provided evidence of the impact of human adolescent drinking in brain structure and functions. Findings in experimental animals have also given new insight into the potential mechanisms of the toxic effects of ethanol on both adolescent brain maturation and the short- and long-term cognitive consequences of adolescent drinking. Adolescence is also characterized by the rapid maturation of brain systems mediating reward and by changes in the secretion of stress-related hormones, events that might participate in the increasing in anxiety and the initiation pattern of alcohol and drug consumption. Studies in human adolescents demonstrate that drinking at early ages can enhance the likelihood of developing alcohol-related problems. Experimental evidence suggests that early exposure to alcohol sensitizes the neurocircuitry of addiction and affects chromatin remodeling, events that could induce abnormal plasticity in reward-related learning processes that contribute to adolescents' vulnerability to drug addiction. In this article, we review the potential mechanisms by which ethanol impacts brain development and lead to brain impairments and cognitive and behavioral dysfunctions as well as the neurobiological and neurochemical processes underlying the adolescent-specific vulnerability to drug addiction.
Collapse
Affiliation(s)
- Consuelo Guerri
- Department of Cellular Pathology, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | | |
Collapse
|
32
|
Bacher I, Wu B, Shytle DR, George TP. Mecamylamine - a nicotinic acetylcholine receptor antagonist with potential for the treatment of neuropsychiatric disorders. Expert Opin Pharmacother 2010; 10:2709-21. [PMID: 19874251 DOI: 10.1517/14656560903329102] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mecamylamine (Inversine), the first orally available antihypertensive agent launched in the 1950s, is rarely used today for hypertension because of its widespread ganglionic side effects at antihypertensive doses (25 - 90 mg/day). However, more recent clinical studies suggest that mecamylamine is effective at much lower doses for blocking the central and peripheral effects of nicotine. Pharmacologically, mecamylamine has been well characterized as a nonselective and noncompetitive antagonist of nicotinic acetylcholine receptors (nAChRs). Because mecamylamine easily crosses the blood - brain barrier at relatively low doses (2.5 - 10 mg), it has been used by several research groups over the past two decades investigating the role of central nAChRs in the etiology and treatment of various neuropsychiatric disorders, including addiction disorders, Tourette's syndrome, schizophrenia and various cognitive and mood disorders. Two independent Phase II clinical trials recently confirmed mecamylamine's hypothesized antidepressant activity and suggest that it may be effective as an augmentation pharmacotherapy for SSRI treatment resistant major depression. These areas of investigation for mecamylamine are reviewed and recommendations for future research directions are proposed.
Collapse
Affiliation(s)
- Ingrid Bacher
- University of Toronto, Department of Psychiatry, Faculty of Medicine, Toronto, Canada.
| | | | | | | |
Collapse
|
33
|
Neurobiological substrates of cue-elicited craving and anhedonia in recently abstinent opioid-dependent males. Drug Alcohol Depend 2009; 99:183-92. [PMID: 18823721 DOI: 10.1016/j.drugalcdep.2008.07.012] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 07/02/2008] [Accepted: 07/24/2008] [Indexed: 01/08/2023]
Abstract
AIM Drug-related stimuli may induce craving in addicted patients, prompting drug-seeking behaviour. In addition, studies have shown addicted patients to be less sensitive to pleasant, but non-drug-related, stimuli; a condition generally referred to as anhedonia. The neural correlates of cue-induced craving and anhedonia in opioid-dependent patients are, however, not well understood. We studied brain activation patterns following visual presentation of neutral, pleasant and heroin-related cues. METHODS Detoxified opioid-dependent males (n=12) and healthy male control subjects (n=17) underwent functional magnetic resonance imaging (fMRI) while subjects viewed neutral, pleasant and heroin-related images. In addition, subjective cue-elicited craving (OCDUS and DDQ) and anhedonia (SHAPS) were measured. RESULTS Opioid-dependent subjects, but not control subjects, showed significant increases in activation in hippocampal region and subcortical limbic structures in response to heroin-related stimuli with a significant group x stimulus interaction effect for the subthalamic nucleus (STN). Control subjects, but not opioid-dependent subjects, showed significant increases in activation of anterior frontal areas and basal ganglia while viewing pleasant images with a significant group x stimulus interaction effect for bilateral anterior prefrontal cortex. Regression analyses showed a positive association between cue-elicited craving and ventral tegmental area (VTA) activation in response to heroin-related stimuli in heroin-dependent patients. In addition, a negative correlation was found between self-reported anhedonia and medial prefrontal regions in both groups. CONCLUSIONS Our findings suggest that the VTA is prominently involved in cue-induced opioid craving for heroin stimuli, in addition to mesolimbic and mesocortical pathways as identified in previous research. The present study also provides further evidence for the involvement of the STN in reward processing. Finally, our data support the presence of reduced brain activation in heroin-dependent patients in response to pleasant (non-drug-related) stimuli.
Collapse
|
34
|
Pascual M, Boix J, Felipo V, Guerri C. Repeated alcohol administration during adolescence causes changes in the mesolimbic dopaminergic and glutamatergic systems and promotes alcohol intake in the adult rat. J Neurochem 2008; 108:920-31. [PMID: 19077056 DOI: 10.1111/j.1471-4159.2008.05835.x] [Citation(s) in RCA: 252] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Adolescence is a developmental period which the risk of drug and alcohol abuse increases. Since mesolimbic dopaminergic system undergoes developmental changes during adolescence, and this system is involved in rewarding effects of drugs of abuse, we addressed the hypothesis that ethanol exposure during juvenile/adolescent period over-activates mesolimbic dopaminergic system inducing adaptations which can trigger long-term enduring behavioural effects of alcohol abuse. We treated juvenile/adolescent or adult rats with ethanol (3 g/kg) for two-consecutive days at 48-h intervals over 14-day period. Here we show that intermittent ethanol treatment during the juvenile/adolescence period alters subsequent ethanol intake. In vivo microdialysis demonstrates that ethanol elicits a similar prolonged dopamine response in the nucleus accumbens of both adolescent and adult animals pre-treated with multiple doses of ethanol, although the basal dopamine levels were higher in ethanol-treated adolescents than in adult-treated animals. Repeated ethanol administration also down-regulates the expression of DRD2 and NMDAR2B phosphorylation in prefrontal cortex of adolescent animals, but not of adult rats. Finally, ethanol treatment during adolescence changes the acetylation of histones H3 and H4 in frontal cortex, nucleus accumbens and striatum, suggesting chromatin remodelling changes. In summary, our findings demonstrate the sensitivity of adolescent brain to ethanol effects on dopaminergic and glutamatergic neurotransmission, and suggest that abnormal plasticity in reward-related processes and epigenetic mechanisms could contribute to the vulnerability of adolescents to alcohol addiction.
Collapse
|
35
|
Andersen SL, Teicher MH. Desperately driven and no brakes: developmental stress exposure and subsequent risk for substance abuse. Neurosci Biobehav Rev 2008; 33:516-24. [PMID: 18938197 DOI: 10.1016/j.neubiorev.2008.09.009] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 09/15/2008] [Accepted: 09/17/2008] [Indexed: 11/29/2022]
Abstract
Adverse life events are associated with a wide range of psychopathology, including an increased risk for substance abuse. In this review, we focus on the inter-relationship between exposure to adversity and brain development, and relate this to enhanced windows of vulnerability. This review encompasses clinical and preclinical data, drawing evidence from epidemiological studies, morphometric and functional imaging studies, and molecular biology and genetics. The interaction of exposure during a sensitive period and maturational events produces a cascade that leads to the initiation of substance use at younger ages, and increases the likelihood of addiction by adolescence or early adulthood. A stress-incubation/corticolimbic dysfunction model is proposed based on the interplay of stress exposure, development stage, and neuromaturational events that may explain the seeking of specific classes of drugs later in life. Three main factors contribute to this age-based progression of increased drug use: (1) a sensitized stress response system; (2) sensitive periods of vulnerability; and (3) maturational processes during adolescence. Together, these factors may explain why exposure to early adversity increases risk to abuse substances during adolescence.
Collapse
Affiliation(s)
- Susan L Andersen
- Developmental Biopsychiatry Research Program, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA.
| | | |
Collapse
|
36
|
Riherd DN, Galindo DG, Krause LR, Mayfield RD. Ethanol potentiates dopamine uptake and increases cell surface distribution of dopamine transporters expressed in SK-N-SH and HEK-293 cells. Alcohol 2008; 42:499-508. [PMID: 18579334 DOI: 10.1016/j.alcohol.2008.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 04/29/2008] [Accepted: 04/29/2008] [Indexed: 11/15/2022]
Abstract
Ethanol increases dopaminergic release in the reward and reinforcement areas of the brain. The primary protein responsible for terminating dopamine (DA) neurotransmission is the plasma membrane-bound dopamine transporter (DAT). In vitro electrophysiological and biochemical studies in Xenopus laevis oocytes have previously shown ethanol potentiates DAT function and increases transporter-binding sites. The potentiating effect of ethanol on the transporter is eliminated in Xenopus oocytes by the DAT mutation glycine 130 to threonine. However, ethanol's action on DAT functional regulation has yet to be examined in mammalian cell expression systems. To further understand the molecular mechanisms of ethanol's action on DAT, we determined the direct mechanistic action of short-term (< or =2 h) ethanol exposure on transporter function and cell surface distribution in non-neuronal human embryonic kidney cells-293 (HEK-293) and neuronal SK-N-SH neuroblastoma cells expressing the transporter. Wild-type or G130T mutant DAT were overexpressed in HEK-293 and SK-N-SH cells. Ethanol potentiated DAT mediated [(3)H]DA uptake in a dose (25, 50, 100 mM), but not time dependent manner in cells expressing wild-type DAT. Ethanol-induced potentiation of uptake was significantly reduced in cells expressing the G130T mutant. Analysis of DA uptake kinetic parameters indicates 100-mM ethanol exposure increased [(3)H]DA uptake velocity (V(max)), while affinity for DA (K(m)) remained unchanged. The effect of ethanol on wild-type DAT surface expression was measured by biotinylation cell surface labeling. DAT surface expression increased 40%-50% after 1-h, 100-mM ethanol exposure. These studies show ethanol potentiates DAT functional regulation in both neuronal and non-neuronal cells, suggesting a direct mechanistic action of ethanol on transporter trafficking in mammalian systems. Our findings demonstrate ethanol's action on DAT function and regulation is consistent across multiple model systems.
Collapse
Affiliation(s)
- D Nicole Riherd
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | | | | | | |
Collapse
|
37
|
Papale L, Andersen M, Perry J, Tufik S. Participation of the cholinergic system in the ethanol-induced suppression of paradoxical sleep in rats. Braz J Med Biol Res 2008; 41:782-8. [DOI: 10.1590/s0100-879x2008000900007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 08/20/2008] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | - J.C. Perry
- Universidade Federal de São Paulo, Brasil
| | - S. Tufik
- Universidade Federal de São Paulo, Brasil
| |
Collapse
|
38
|
Pharmacothérapies actuelles et immunothérapie dans l’addiction à la cocaïne. Presse Med 2008; 37:689-98. [DOI: 10.1016/j.lpm.2007.08.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Accepted: 08/29/2007] [Indexed: 11/18/2022] Open
|
39
|
alpha1-noradrenergic receptor antagonism blocks dependence-induced increases in responding for ethanol. Alcohol 2008; 42:91-7. [PMID: 18358987 DOI: 10.1016/j.alcohol.2007.12.002] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 11/28/2007] [Accepted: 12/10/2007] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to test the hypothesis that blockade of alpha1-adrenergic receptors may suppress the excessive ethanol consumption associated with acute withdrawal in ethanol-dependent rats. Following the acquisition and stabilization of operant ethanol self-administration in male Wistar rats, dependence was induced in half the animals by subjecting them to a 4-week intermittent vapor exposure period in which animals were exposed to ethanol vapor for 14h/day. Subsequent to dependence induction, the effect of alpha1-noradrenergic receptor antagonist prazosin (0.0, 0.25, 0.5, 1, 1.5, and 2.0mg/kg IP) was tested on operant responding for ethanol in vapor-exposed and control rats during acute withdrawal. In ethanol-dependent animals, prazosin significantly suppressed responding at the 1.5 and 2.0mg/kg doses, whereas only the 2.0mg/kg dose was effective in nondependent animals, identifying an increase in the sensitivity to prazosin in dependent animals. Conversely, at the lowest dose tested (0.25mg/kg), prazosin increased responding in nondependent animals, which is consistent with the effect of anxiolytics on ethanol self-administration in nondependent animals. None of the doses tested reliably affected concurrent water self-administration. These results suggest the involvement of the noradrenergic system in the excessive alcohol drinking seen during acute withdrawal in ethanol-dependent rats.
Collapse
|
40
|
Nocjar C, Hammonds M, Shim S. Chronic lithium treatment magnifies learning in rats. Neuroscience 2007; 150:774-88. [DOI: 10.1016/j.neuroscience.2007.09.063] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 09/26/2007] [Accepted: 09/27/2007] [Indexed: 01/05/2023]
|
41
|
George AK, Balarama Kaimal S, Paulose CS. Decreased dopamine D(2) receptor function in cerebral cortex and brain stem: their role in hepatic ALDH regulation in ethanol treated rats. Mol Cell Biochem 2007; 304:181-8. [PMID: 17530188 DOI: 10.1007/s11010-007-9498-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Accepted: 04/27/2007] [Indexed: 10/23/2022]
Abstract
Ethanol exerts numerous pharmacological effects through its interaction with various neurotransmitters. The dopaminergic pathway is associated with cognitive, endocrine, and motor functions, and reinforcement of addictive substances or behaviours. Aldehyde dehydrogenase (ALDH) is a vital enzyme involved with alcohol metabolism and detoxification. In the present study, we investigated the role of cerebral cortex and brain stem dopamine D(2) receptors in the functional regulation on ALDH enzyme activity, in ethanol administrated rats. Two groups of rats were selected viz. control and alcoholic. Cerebral cortex, brain stem and the liver dopamine content was decreased significantly (P < 0.05, 0.05, 0.001, respectively) and homovanillic acid/dopamine (HVA/DA) ratio has significantly increased (P < 0.05, 0.001 and 0.001), respectively in ethanol treated rats when compared to control. Scatchard analysis of [(3)H]YM-09151-2 binding to synaptic membrane preparations of cerebral cortex and brain stem showed a significant decrease (P < 0.001, 0.05, respectively) in B (max) in ethanol treated rats compared to control and the K (d) also decreased significantly (P < 0.05). The ALDH analysis showed a significant increase (P < 0.05) in V (max) in cerebral cortex, plasma and liver of experimental rats when compared with control without having significant change in brain stem but with decreased K (m) (P < 0.001). Our results suggest that decreased function of dopamine mediated through DA D(2) receptor in the cerebral cortex and brain stem enhanced the brain, plasma and liver ALDH activity in ethanol treated rats. This ALDH regulation has significance to correct alcoholics from addiction due to allergic reaction observed in aldehyde accumulation.
Collapse
Affiliation(s)
- Akash K George
- Molecular Neurobiology and Cell Biology Unit, Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology, Cochin, 682022 Kerala, India
| | | | | |
Collapse
|
42
|
Amato JL, Bankson MG, Yamamoto BK. Prior exposure to chronic stress and MDMA potentiates mesoaccumbens dopamine release mediated by the 5-HT(1B) receptor. Neuropsychopharmacology 2007; 32:946-54. [PMID: 16885935 DOI: 10.1038/sj.npp.1301174] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
(+) 3,4,-Methylenedioxymethamphetamine (MDMA) is an abused drug that acutely releases serotonin (5-HT) and dopamine (DA) but produces long-term damage to 5-HT terminals. MDMA-induced DA release has been shown to be dampened by 5-HT. Although stress also activates the mesolimbic DA pathway, it is unknown if chronic stress after exposure to neurotoxic doses of MDMA will augment MDMA-induced DA release in the nucleus accumbens shell (NAcc(sh)). Rats were pretreated with MDMA (10 mg/kg x 4, intraperitoneal (i.p.)). After 7 days, rats were subjected to 10 days of chronic unpredictable stress. DA release in the NAcc(sh) and 5-HT in the ventral tegmental area (VTA) were measured after a challenge injection of MDMA (5 mg/kg, i.p.). The combination of pretreatment with MDMA+stress decreased basal concentrations of 5-HT in the VTA and DA in the NAcc(sh) and enhanced MDMA-stimulated DA release in the NAcc(sh). Pretreatment with MDMA or stress alone blunted MDMA-induced 5-HT release in the VTA. The augmentation of MDMA-induced DA release in rats pretreated with MDMA+chronic stress was attenuated by perfusion of the 5-HT(1B) antagonist, GR127935 into the VTA before the MDMA challenge injection. These results suggest that prior exposure to both MDMA and stress can produce a long-term augmentation in mesolimbic DA transmission and enhanced drug abuse vulnerability that is mediated, in part, by the 5-HT(1B) receptor in the VTA.
Collapse
Affiliation(s)
- Jennifer L Amato
- Laboratory of Neurochemistry, Department of Pharmacology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | |
Collapse
|
43
|
Golovko AI, Golovko SI, Leontieva LV. The neurochemistry of the psychological dependence syndrome in addictive diseases of chemical etiology. NEUROCHEM J+ 2007. [DOI: 10.1134/s1819712407010011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
44
|
Pettinati HM, O'Brien CP, Rabinowitz AR, Wortman SP, Oslin DW, Kampman KM, Dackis CA. The status of naltrexone in the treatment of alcohol dependence: specific effects on heavy drinking. J Clin Psychopharmacol 2006; 26:610-25. [PMID: 17110818 DOI: 10.1097/01.jcp.0000245566.52401.20] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND In almost 2 decades of naltrexone research for treating alcoholism, there have been 29 published randomized placebo-controlled trials of opioid antagonists, primarily naltrexone, for the treatment of alcohol dependence. The present review builds on prior systematic reviews while maximizing the number of included studies to date, for the purpose of resolving inconsistencies in naltrexone's reported efficacy across trials. Clinical trial results in this article are evaluated by the type of outcome measure used to determine naltrexone's treatment advantage, that is, measures related to reducing heavy drinking versus those related to increasing abstinence. METHODS We conducted a Medline search to identify double-blind studies from 1990 to the present (2006) that evaluated the use of anopiate antagonist for the treatment of alcohol dependence. There were 29 studies identified, representing 5997 alcohol-dependent patients, which met our study inclusion criteria for this review. Studies were evaluated in this review on 4 prespecified drinking outcomes-2 related to "any drinking" and 2 related to "heavy or excessive drinking." RESULTS In the treatment of alcohol dependence, we found that 19 (70%) of 27 clinical trials that measured reductions in "heavy or excessive drinking" demonstrated an advantage for prescribing naltrexone over placebo, whereas only 9 (36%) of 25 clinical trials that measured abstinence or "any drinking" found an advantage for medication over placebo. CONCLUSION The majority of double-blind clinical trials in the literature favored prescribing naltrexone for alcohol dependence to reduce heavy drinking. This finding is consistent with our understanding of naltrexone's mechanism of action of decreasing excessive drinking by reducing the reward associated with drinking alcohol. Thus, we conclude that outcome measures related to heavy or excessive drinking are most relevant to defining naltrexone's therapeutic effects. Factors influencing naltrexone response (treatment adherence and distinct patient subgroups) are also discussed.
Collapse
Affiliation(s)
- Helen M Pettinati
- Center for the Study of Addictions, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6178, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Ivanov IS, Schulz KP, Palmero RC, Newcorn JH. Neurorobiology and evidence-based biological treatments for substance abuse disorders. CNS Spectr 2006; 11:864-77. [PMID: 17075558 DOI: 10.1017/s1092852900015017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Behavioral patterns of addiction include compulsive drug-seeking, persistent abuse of substances despite the often dire consequences on social functioning and physical health, and the high probability of relapse even after prolonged drug-free periods. The recent focus on the biological basis of addiction has provided evidence to support the hypothesis that behavioral manifestations for addiction are influenced by biological factors, and biological factors often produce behavioral changes that can further increase risk. The current understanding of the role of the dopaminergic, glutamatergic, Upsilon-aminobutyric acidergic, and opioid receptor systems in the pathophysiology of addiction as well as the clinical implications of these systems for new and emerging treatments will be discussed. This article will also review the pharmacologic agents used in the treatment of substance abuse disorders and presents evidence-based data for their safety, efficacy, and feasibility of use in different patient populations.
Collapse
Affiliation(s)
- Iliyan S Ivanov
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | |
Collapse
|
46
|
Ito M, Nagai T, Kamei H, Nakamichi N, Nabeshima T, Takuma K, Yamada K. Involvement of Tissue Plasminogen Activator-Plasmin System in Depolarization-Evoked Dopamine Release in the Nucleus Accumbens of Mice. Mol Pharmacol 2006; 70:1720-5. [PMID: 16908600 DOI: 10.1124/mol.106.022467] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tissue plasminogen activator (tPA), a serine protease, catalyzes the conversion of plasminogen to plasmin. In the present study, we investigated the role of the tPA-plasmin system in depolarization-evoked dopamine (DA) and acetylcholine (ACh) release in the nucleus accumbens (NAc) and hippocampus, respectively, of mice, by using in vivo microdialysis. Microinjection of either tPA or plasmin significantly potentiated 40 mM KCl-induced DA release without affecting basal DA levels. In contrast, plasminogen activator inhibitor-1 dose-dependently reduced 60 mM KCl-induced DA release. The 60 mM KCl-evoked DA release in the NAc was markedly diminished in tPA-deficient (tPA-/-) mice compared with wild-type mice, although basal DA levels did not differ between the two groups. Microinjections of either exogenous tPA (100 ng) or plasmin (100 ng) into the NAc of tPA-/-mice restored 60 mM KCl-induced DA release, as observed in wild-type mice. In contrast, there was no difference in either basal or 60 mM KCl-induced ACh release in the hippocampus between wild-type and tPA-/-mice. Our findings suggest that the tPA-plasmin system is involved in the regulation of depolarization-evoked DA release in the NAc.
Collapse
Affiliation(s)
- Mina Ito
- Laboratory of Neuropsychopharmacology, Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Gu PF, Wu CF, Yang JY, Shang Y, Hou Y, Bi XL, Dai F. Differential effects of drug-induced ascorbic acid release in the striatum and nucleus accumbens of freely moving rats. Neurosci Lett 2006; 399:79-84. [PMID: 16480820 DOI: 10.1016/j.neulet.2006.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2005] [Revised: 01/04/2006] [Accepted: 01/13/2006] [Indexed: 11/16/2022]
Abstract
Previous studies have shown that striatum and nucleus accumbens (NAc) are two different structures in mediating addictive drug-induced ascorbic acid (AA) release. In order to further characterize the different effects of drugs-induced AA release in the striatum and NAc, in the present study, we investigated the effect of ethanol, morphine, methamphetamine, nicotine-induced AA release in these two nuclei using microdialysis coupled to high performance liquid chromatography with electrochemical detection (HPLC-ECD). All drugs were continuously perfused directly into the striatum or NAc. This study showed that local intrastriatal or intra-accumbensal perfusion of ethanol (500 microM) could increase AA release to 280, 260% in the striatum and NAc, respectively. Intra-striatal infusion of morphine (1 mM), methamphetamine (250 microM) or nicotine (500 microM), reduce striatal AA release to 48, 50, 45%, respectively. While given intra-accumbensally, morphine (1 mM), methamphetamine (250 microM) or nicotine (500 microM) increase AA release to 165, 160, 160%, respectively. These results suggested that different presynaptic or postsynaptic mechanisms might be involved in addictive drug-induced AA release in the striatum and NAc.
Collapse
Affiliation(s)
- Pei Fei Gu
- Department of Pharmacology, Shenyang Pharmaceutical University, Wenhua Road 103, 110016 Shenyang, PR China
| | | | | | | | | | | | | |
Collapse
|
48
|
Funk D, Li Z, Lê AD. Effects of environmental and pharmacological stressors on c-fos and corticotropin-releasing factor mRNA in rat brain: Relationship to the reinstatement of alcohol seeking. Neuroscience 2005; 138:235-43. [PMID: 16359808 DOI: 10.1016/j.neuroscience.2005.10.062] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Revised: 09/12/2005] [Accepted: 10/31/2005] [Indexed: 11/15/2022]
Abstract
We have observed marked heterogeneity among different stressors in their ability to reinstate alcohol seeking in rats. Of the stressors we have tested, only the environmental stressor footshock and the pharmacological stressor yohimbine induce reinstatement. The reasons for such differences among stressors are not known. The purpose of the experiments presented here is to determine the neuroanatomical substrates that underlie these behavioral differences. To this end, we assessed whether stressors effective in inducing reinstatement of alcohol seeking activate a different set of neuronal pathways than do those that are ineffective, using the technique of in situ hybridization of the mRNAs for c-fos, a marker of neuronal activation, and corticotropin-releasing factor (CRF), a stress-related peptide we have shown to be critical to footshock-induced reinstatement of alcohol seeking. Exposure of rats to the environmental stressors footshock, restraint or social defeat, or the pharmacological stressors yohimbine or FG-7142 increased levels of the mRNAs for c-fos and CRF in the brain in a number of areas previously shown to be responsive to stressors. We found regionally specific effects of the stressors on c-fos and CRF mRNA in brain regions associated with the rewarding effects of alcohol and other abused drugs. The two stressors we have previously shown to be effective in inducing reinstatement of alcohol seeking, footshock and yohimbine, induced c-fos mRNA in the shell of the nucleus accumbens, and the basolateral and central amygdalar nuclei. These two stressors also induced CRF mRNA in the dorsal region of the bed nucleus of the stria terminalis. Taken together, these results provide evidence that activity in these regions may be involved in the reinstatement of alcohol seeking induced by these stressors. These results are also in keeping with the previously demonstrated role of CRF neurons in the dorsal bed nucleus of the stria terminalis in the reinstatement of alcohol seeking induced by stress.
Collapse
Affiliation(s)
- D Funk
- Department of Neuroscience, Center for Addiction and Mental Health, 33 Russell Street, Toronto, Ontario, Canada M5S 2S1.
| | | | | |
Collapse
|
49
|
Hasebe K, Kawai K, Suzuki T, Kawamura K, Tanaka T, Narita M, Nagase H, Suzuki T. Possible pharmacotherapy of the opioid kappa receptor agonist for drug dependence. Ann N Y Acad Sci 2005; 1025:404-13. [PMID: 15542743 DOI: 10.1196/annals.1316.050] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Because there are few efficacious medications for drug dependence, many clinical trials are being conducted in earnest to find such medications. Considerable evidence has shown that opioid kappa receptor agonists attenuate several behavioral responses induced by drugs of abuse. Although this raises the possibility that opioid kappa receptor agonists may be useful for the treatment of drug dependence on drugs of abuse, it has been previously reported that treatment with selective opioid kappa receptor agonists causes a psychotomimetic effect and dysphoria both in clinical studies and experimental animal models. As a result, we found the novel opioid kappa receptor agonist TRK-820, another chemical class of opioid kappa receptor agonist that has a morphinan scaffold unlike prototypical opioid kappa receptor agonists, by application of a modified message-address concept. TRK-820 showed high selectivity for an opioid kappa receptor, and strong agonistic activity in both in vitro and in vivo experiments. Like other opioid kappa receptor agonists, TRK-820 could markedly suppress the rewarding effects induced by morphine and cocaine and the discriminative stimulus effect of cocaine. Furthermore, TRK-820 attenuated the mecamylamine-precipitated nicotine-withdrawal aversion in a conditioned place preference paradigm. It is worthwhile to note that unlike prototypical opioid kappa receptor agonists, TRK-820 failed to produce a significant place aversion in rodents at doses that were sufficient to produce significant antinociception. Taken together, these findings indicate that TRK-820 may be useful for the treatment of drug dependence without any aversive effects.
Collapse
Affiliation(s)
- Ko Hasebe
- Pharmaceutical Research Laboratories, Toray Industries Inc., Kanagawa 248-8555, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Andersen SL, Navalta CP. Altering the course of neurodevelopment: a framework for understanding the enduring effects of psychotropic drugs. Int J Dev Neurosci 2004; 22:423-40. [PMID: 15380841 DOI: 10.1016/j.ijdevneu.2004.06.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Revised: 06/07/2004] [Accepted: 06/07/2004] [Indexed: 12/13/2022] Open
Abstract
Childhood is a time filled with wondrous changes, as brain plasticity permits experiences to shape the immature brain to meet the demands of the environment. Change occurs at various levels--from neuroanatomy, including within a given region and its connectivity to other regions, to the function of neurotransmitter systems and their reactivity to pharmacological agents in the short- and long-term. The nature and degree to which drug exposure influences the final adult topography is influenced greatly by the maturational phase of these critical factors. Moreover, evidence is slowly emerging that suggests that the long-term effects of drug exposure are delayed and expressed once the vulnerable system reaches maturation (i.e., typically during adulthood). This phenomenon is known as neuronal imprinting and occurs when the effects of drug exposure outlast the drug itself. Thus, understanding the persistent effects critically depends on the window of observation. Embracing this concept should influence how we conduct preclinical assessments of developmental drug exposure, and ultimately how we conduct clinical assessments of drug efficacy, effectiveness, and safety for the treatment of childhood psychiatric disorders. In this article, we present a model to provide a heuristic framework for making predictions about imprinted effects of childhood drug exposure. We then review epidemiological data on attention deficit hyperactivity disorder (ADHD) and childhood depression, prescription practices, and what is known regarding the long-term consequences of drug exposure in these populations. We conclude with a discussion of the current status of preclinical studies on juvenile stimulant exposure.
Collapse
Affiliation(s)
- Susan L Andersen
- Department of Psychiatry, McLean Hospital and Harvard Medical School, 115 Mill Street, Belmont, MA 02478, USA.
| | | |
Collapse
|