101
|
Logrip ML, Walker JR, Ayanwuyi LO, Sabino V, Ciccocioppo R, Koob GF, Zorrilla EP. Evaluation of Alcohol Preference and Drinking in msP Rats Bearing a Crhr1 Promoter Polymorphism. Front Psychiatry 2018; 9:28. [PMID: 29497387 PMCID: PMC5818434 DOI: 10.3389/fpsyt.2018.00028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 01/23/2018] [Indexed: 12/22/2022] Open
Abstract
Alcoholism is a pervasive societal problem, yet available pharmacotherapies fail to treat most sufferers. The type 1 corticotropin-releasing factor (CRF1) receptor has received much attention for its putative role in the progression to alcohol dependence, although at present its success in clinical trials has been limited. Two single-nucleotide polymorphisms in the rat Crhr1 promoter have been identified in the Marchigian substrain of Sardinian alcohol-preferring (msP) rats. Unlike other Wistar-derived alcohol-preferring lines, nondependent msP rats reduce their alcohol self-administration in response to CRF1 antagonists and show increased brain CRF1 expression. The current study tested the hypotheses that the A alleles in the Crhr1 promoter polymorphisms are: (1) unique to msP (vs. CRF1 antagonist-insensitive) alcohol-preferring lines and (2) associate with greater alcohol preference or intake. Two related polymorphisms were observed in which both loci on a given chromosome were either mutant variant (A) or wild-type (G) alleles within the distal Crhr1 promoter of 17/25 msP rats (68%), as compared to 0/23 Indiana P rats, 0/20 Sardinian alcohol-preferring rats bred at Scripps (Scr:sP) and 0/21 outbred Wistar rats. Alcohol consumption in msP rats did not differ according to the presence of Crhr1 A alleles, but greater alcohol preference (98%) was observed in A allele homozygous msP rats (AA) compared to msP rats with wild-type (GG, 91%) or heterozygous (GA, 91%) genotypes. The greater alcohol preference reflected decreased water intake, accompanied by reduced total calories consumed by AA rats. The data show that msP rats differentially possess mutant A variant alleles in the polymorphic promoter region of the Crhr1 gene that may differentially regulate consumption.
Collapse
Affiliation(s)
- Marian L Logrip
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, United States.,Department of Psychology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States
| | - John R Walker
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Lydia O Ayanwuyi
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Valentina Sabino
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, United States.,Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - George F Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Eric P Zorrilla
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
102
|
Abstract
Animal models provide rapid, inexpensive assessments of an investigational drug's therapeutic potential. Ideally, they support the plausibility of therapeutic efficacy and provide a rationale for further investigation. Here, I discuss how the absence of clear effective-ineffective categories for alcohol use disorder (AUD) medications and biases in the clinical and preclinical literature affect the development of predictive preclinical alcohol dependence (AD) models. Invoking the analogical argument concept from the philosophy of science field, I discuss how models of excessive alcohol drinking support the plausibility of clinical pharmacotherapy effects. Even though these models are not likely be completely discriminative, they are sensitive to clinically effective medications and have revealed dozens of novel medication targets. In that context, I discuss recent preclinical work on GLP-1 receptor agonists, phosphodiesterase inhibitors, glucocorticoid receptor antagonists, nociception agonists and antagonists, and CRF1 antagonists. Clinically approved medications are available for each of these drug classes. I conclude by advocating a translational approach in which drugs are evaluated highly congruent preclinical models and human laboratory studies. Once translation is established, I suggest the burden is to develop hypothesis-based therapeutic interventions maximizing the impact of the confirmed pharmacotherapeutic effects in the context of additional variables falling outside the model.
Collapse
Affiliation(s)
- Mark Egli
- Division of Neuroscience and Behavior, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
103
|
Klenowski PM, Tapper AR. Molecular, Neuronal, and Behavioral Effects of Ethanol and Nicotine Interactions. Handb Exp Pharmacol 2018; 248:187-212. [PMID: 29423839 DOI: 10.1007/164_2017_89] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
104
|
Abreu-Villaça Y, Manhães AC, Krahe TE, Filgueiras CC, Ribeiro-Carvalho A. Tobacco and alcohol use during adolescence: Interactive mechanisms in animal models. Biochem Pharmacol 2017; 144:1-17. [DOI: 10.1016/j.bcp.2017.06.113] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/05/2017] [Indexed: 12/13/2022]
|
105
|
Čupić Ž, Stanojević A, Marković VM, Kolar-Anić L, Terenius L, Vukojević V. The HPA axis and ethanol: a synthesis of mathematical modelling and experimental observations. Addict Biol 2017; 22:1486-1500. [PMID: 27189379 DOI: 10.1111/adb.12409] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 03/24/2016] [Accepted: 04/11/2016] [Indexed: 12/21/2022]
Abstract
Stress and alcohol use are interrelated-stress contributes to the initiation and upholding of alcohol use and alcohol use alters the way we perceive and respond to stress. Intricate mechanisms through which ethanol alters the organism's response to stress remain elusive. We have developed a stoichiometric network model to succinctly describe neurochemical transformations underlying the stress response axis and use numerical simulations to model ethanol effects on complex daily changes of blood levels of cholesterol, 6 peptide and 8 steroid hormones. Modelling suggests that ethanol alters the dynamical regulation of hypothalamic-pituitary-adrenal (HPA) axis activity by affecting the amplitude of ultradian oscillations of HPA axis hormones, which defines the threshold with respect to which the response to stress is being set. These effects are complex-low/moderate acute ethanol challenge (<8 mM) may reduce, leave unaltered or increase the amplitude of ultradian cortisol (CORT) oscillations, giving rise to an intricate response at the organism level, offering also a potential explanation as to why apparently discordant results were observed in experimental studies. In contrast, high-dose acute ethanol challenge (>8 mM) increases instantaneous CORT levels and the amplitude of ultradian CORT oscillations in a dose-dependent manner, affecting the HPA axis activity also during the following day(s). Chronic exposure to ethanol qualitatively changes the HPA axis dynamics, whereas ethanol at intoxicating levels shuts down this dynamic regulation mechanism. Mathematical modelling gives a quantitative biology-based framework that can be used for predicting how the integral HPA axis response is perturbed by alcohol.
Collapse
Affiliation(s)
- Željko Čupić
- Institute of Chemistry, Technology and Metallurgy, Department of Catalysis and Chemical Engineering; University of Belgrade; Belgrade Serbia
| | - Ana Stanojević
- Faculty of Physical Chemistry; University of Belgrade; Belgrade Serbia
| | | | - Ljiljana Kolar-Anić
- Institute of Chemistry, Technology and Metallurgy, Department of Catalysis and Chemical Engineering; University of Belgrade; Belgrade Serbia
- Faculty of Physical Chemistry; University of Belgrade; Belgrade Serbia
| | - Lars Terenius
- Department of Clinical Neuroscience, Center for Molecular Medicine; Karolinska Institutet; Stockholm Sweden
| | - Vladana Vukojević
- Department of Clinical Neuroscience, Center for Molecular Medicine; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
106
|
Somkuwar SS, Vendruscolo LF, Fannon MJ, Schmeichel B, Nguyen TB, Guevara J, Sidhu H, Contet C, Zorrilla EP, Mandyam CD. Abstinence from prolonged ethanol exposure affects plasma corticosterone, glucocorticoid receptor signaling and stress-related behaviors. Psychoneuroendocrinology 2017; 84. [PMID: 28647675 PMCID: PMC5557646 DOI: 10.1016/j.psyneuen.2017.06.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Alcohol dependence is linked to dysregulation of the hypothalamic-pituitary-adrenal axis. Here, we investigated effects of repeated ethanol intoxication-withdrawal cycles (using chronic intermittent ethanol vapor inhalation; CIE) and abstinence from CIE on peak and nadir plasma corticosterone (CORT) levels. Irritability- and anxiety-like behaviors as well as glucocorticoid receptors (GR) in the medial prefrontal cortex (mPFC) were assessed at various intervals (2h-28d) after cessation of CIE. Results show that peak CORT increased during CIE, transiently decreased during early abstinence (1-11d), and returned to pre-abstinence levels during protracted abstinence (17-27d). Acute withdrawal from CIE enhanced aggression- and anxiety-like behaviors. Early abstinence from CIE reduced anxiety-like behavior. mPFC-GR signaling (indexed by relative phosphorylation of GR at Ser211) was transiently decreased when measured at time points during early and protracted abstinence. Further, voluntary ethanol drinking in CIE (CIE-ED) and CIE-naïve (ED) rats, and effects of CIE-ED and ED on peak CORT levels and mPFC-GR were investigated during acute withdrawal (8h) and protracted abstinence (28d). CIE-ED and ED increased peak CORT during drinking. CIE-ED and ED decreased expression and signaling of mPFC-GR during acute withdrawal, an effect that was reversed by systemic mifepristone treatment. CIE-ED and ED demonstrate robust reinstatement of ethanol seeking during protracted abstinence and show increases in mPFC-GR expression. Collectively, the data demonstrate that acute withdrawal from CIE produces robust alterations in GR signaling, CORT and negative affect symptoms which could facilitate excessive drinking. The findings also show that CIE-ED and ED demonstrate enhanced relapse vulnerability triggered by ethanol cues and these changes are partially mediated by altered GR expression in the mPFC. Taken together, transition to alcohol dependence could be accompanied by alterations in mPFC stress-related pathways that may increase negative emotional symptoms and increase vulnerability to relapse.
Collapse
Affiliation(s)
| | | | | | - Brooke Schmeichel
- National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Tran Bao Nguyen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA, USA
| | | | - Harpreet Sidhu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA USA
| | - Candice Contet
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA USA
| | - Eric P. Zorrilla
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA USA
| | - Chitra D. Mandyam
- VA San Diego Healthcare System, San Diego, CA, USA,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA, USA,Department of Neuroscience, The Scripps Research Institute, La Jolla, CA USA,Department of Anesthesiology, University of California San Diego, CA, USA
| |
Collapse
|
107
|
Leonard MZ, DeBold JF, Miczek KA. Escalated cocaine "binges" in rats: enduring effects of social defeat stress or intra-VTA CRF. Psychopharmacology (Berl) 2017; 234:2823-2836. [PMID: 28725939 PMCID: PMC5709163 DOI: 10.1007/s00213-017-4677-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/16/2017] [Indexed: 12/16/2022]
Abstract
RATIONALE Exposure to intermittent social defeat stress elicits corticotropin releasing factor (CRF) release into the VTA and induces long-term modulation of mesocorticolimbic dopamine activity in rats. These adaptations are associated with an intense cocaine-taking phenotype, which is prevented by CRF receptor antagonists. OBJECTIVE The present studies examine whether infusion of CRF into the VTA is sufficient to escalate cocaine-taking behavior, in the absence of social defeat experience. Additionally, we aimed to characterize changes in cocaine valuation that may promote binge-like cocaine intake. METHODS Male Long-Evans rats were microinjected into the VTA with CRF (50 or 500 ng/side), vehicle, or subjected to social defeat stress, intermittently over 10 days. Animals were then trained to self-administer IV cocaine (FR5). Economic demand for cocaine was evaluated using a within-session behavioral-economics threshold procedure, which was followed by a 24-h extended access "binge." RESULTS Rats that experienced social defeat or received intra-VTA CRF microinfusions (50 ng) both took significantly more cocaine than controls over the 24-h binge but showed distinct patterns of intake. Behavioral economic analysis revealed that individual demand for cocaine strongly predicts binge-like consumption, and demand elasticity (i.e. α) is augmented by intra-VTA CRF, but not by social defeat. The effects of CRF on cocaine-taking were also prevented by intra-VTA pretreatment with CP376395, but not Astressin-2B. CONCLUSIONS Repeated infusion of CRF into the VTA persistently alters cocaine valuation and intensifies binge-like drug intake in a CRF-R1-dependent manner. Conversely, the persistent pattern of cocaine bingeing induced by social defeat stress may suggest impaired inhibitory control, independent of reward valuation.
Collapse
Affiliation(s)
| | - Joseph F DeBold
- Department of Psychology, Tufts University, Medford, MA, USA
| | - Klaus A Miczek
- Department of Psychology, Tufts University, Medford, MA, USA.
- Department of Neuroscience, Tufts University, Boston, MA, USA.
| |
Collapse
|
108
|
Shimamoto A, Rappeneau V. Sex-dependent mental illnesses and mitochondria. Schizophr Res 2017; 187:38-46. [PMID: 28279571 PMCID: PMC5581986 DOI: 10.1016/j.schres.2017.02.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 12/11/2022]
Abstract
The prevalence of some mental illnesses, including major depression, anxiety-, trauma-, and stress-related disorders, some substance use disorders, and later onset of schizophrenia, is higher in women than men. While the higher prevalence in women could simply be explained by socioeconomic determinants, such as income, social status, or cultural background, extensive studies show sex differences in biological, pharmacokinetic, and pharmacological factors contribute to females' vulnerability to these mental illnesses. In this review, we focus on estrogens, chronic stress, and neurotoxicity from behavioral, pharmacological, biological, and molecular perspectives to delineate the sex differences in these mental illnesses. Particularly, we investigate a possible role of mitochondrial function, including biosynthesis, bioenergetics, and signaling, on mediating the sex differences in psychiatric disorders.
Collapse
Affiliation(s)
- Akiko Shimamoto
- Department of Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, TN 37028-3599, United States.
| | - Virginie Rappeneau
- Department of Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, TN 37028-3599, United States
| |
Collapse
|
109
|
Meye FJ, Trusel M, Soiza-Reilly M, Mameli M. Neural circuit adaptations during drug withdrawal - Spotlight on the lateral habenula. Pharmacol Biochem Behav 2017; 162:87-93. [PMID: 28843423 DOI: 10.1016/j.pbb.2017.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 07/13/2017] [Accepted: 08/21/2017] [Indexed: 02/03/2023]
Abstract
Withdrawal after drug intake triggers a wealth of affective states including negative feelings reminiscent of depressive symptoms. This negative state can ultimately be crucial for relapse, a hallmark of addiction. Adaptations in a wide number of neuronal circuits underlie aspects of drug withdrawal, however causality between cellular modifications within these systems and precise behavioral phenotypes remains poorly described. Recent advances point to an instrumental role of the lateral habenula in driving depressive-like states during drug withdrawal. In this review we will discuss the general behavioral features of drug withdrawal, the importance of plasticity mechanisms in the mesolimbic systems, and the latest discoveries highlighting the implications of lateral habenula in drug addiction. We will further stress how specific interventions in the lateral habenula efficiently ameliorate depressive symptoms. Altogether, this work aims to provide a general knowledge on the cellular and circuit basis underlying drug withdrawal, ultimately speculating on potential treatment for precise aspects of addiction.
Collapse
Affiliation(s)
- Frank J Meye
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Massimo Trusel
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland; Inserm UMR-S 839, Institut du Fer à Moulin, Paris, France
| | | | - Manuel Mameli
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland; Inserm UMR-S 839, Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
110
|
Bell RL, Hauser SR, Liang T, Sari Y, Maldonado-Devincci A, Rodd ZA. Rat animal models for screening medications to treat alcohol use disorders. Neuropharmacology 2017; 122:201-243. [PMID: 28215999 PMCID: PMC5659204 DOI: 10.1016/j.neuropharm.2017.02.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 01/21/2023]
Abstract
The purpose of this review is to present animal research models that can be used to screen and/or repurpose medications for the treatment of alcohol abuse and dependence. The focus will be on rats and in particular selectively bred rats. Brief introductions discuss various aspects of the clinical picture, which provide characteristics of individuals with alcohol use disorders (AUDs) to model in animals. Following this, multiple selectively bred rat lines will be described and evaluated in the context of animal models used to screen medications to treat AUDs. Next, common behavioral tests for drug efficacy will be discussed particularly as they relate to stages in the addiction cycle. Tables highlighting studies that have tested the effects of compounds using the respective techniques are included. Wherever possible the Tables are organized chronologically in ascending order to describe changes in the focus of research on AUDs over time. In general, high ethanol-consuming selectively bred rats have been used to test a wide range of compounds. Older studies usually followed neurobiological findings in the selected lines that supported an association with a propensity for high ethanol intake. Most of these tests evaluated the compound's effects on the maintenance of ethanol drinking. Very few compounds have been tested during ethanol-seeking and/or relapse and fewer still have assessed their effects during the acquisition of AUDs. Overall, while a substantial number of neurotransmitter and neuromodulatory system targets have been assessed; the roles of sex- and age-of-animal, as well as the acquisition of AUDs, ethanol-seeking and relapse continue to be factors and behaviors needing further study. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Richard L Bell
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA.
| | - Sheketha R Hauser
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA
| | - Tiebing Liang
- Indiana University School of Medicine, Department of Gastroenterology, Indianapolis, IN 46202, USA
| | - Youssef Sari
- University of Toledo, Department of Pharmacology, Toledo, OH 43614, USA
| | | | - Zachary A Rodd
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA
| |
Collapse
|
111
|
Inda C, Armando NG, Dos Santos Claro PA, Silberstein S. Endocrinology and the brain: corticotropin-releasing hormone signaling. Endocr Connect 2017; 6:R99-R120. [PMID: 28710078 PMCID: PMC5551434 DOI: 10.1530/ec-17-0111] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 01/01/2023]
Abstract
Corticotropin-releasing hormone (CRH) is a key player of basal and stress-activated responses in the hypothalamic-pituitary-adrenal axis (HPA) and in extrahypothalamic circuits, where it functions as a neuromodulator to orchestrate humoral and behavioral adaptive responses to stress. This review describes molecular components and cellular mechanisms involved in CRH signaling downstream of its G protein-coupled receptors (GPCRs) CRHR1 and CRHR2 and summarizes recent findings that challenge the classical view of GPCR signaling and impact on our understanding of CRHRs function. Special emphasis is placed on recent studies of CRH signaling that revealed new mechanistic aspects of cAMP generation and ERK1/2 activation in physiologically relevant contexts of the neurohormone action. In addition, we present an overview of the pathophysiological role of the CRH system, which highlights the need for a precise definition of CRHRs signaling at molecular level to identify novel targets for pharmacological intervention in neuroendocrine tissues and specific brain areas involved in CRH-related disorders.
Collapse
Affiliation(s)
- Carolina Inda
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
- DFBMCFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia G Armando
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - Paula A Dos Santos Claro
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - Susana Silberstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
- DFBMCFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
112
|
Becker HC. Influence of stress associated with chronic alcohol exposure on drinking. Neuropharmacology 2017; 122:115-126. [PMID: 28431971 PMCID: PMC5497303 DOI: 10.1016/j.neuropharm.2017.04.028] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/12/2017] [Accepted: 04/17/2017] [Indexed: 12/24/2022]
Abstract
Stress is commonly regarded as an important trigger for relapse and a significant factor that promotes increased motivation to drink in some individuals. However, the relationship between stress and alcohol is complex, likely changing in form during the transition from early moderated alcohol use to more heavy uncontrolled alcohol intake. A growing body of evidence indicates that prolonged excessive alcohol consumption serves as a potent stressor, producing persistent dysregulation of brain reward and stress systems beyond normal homeostatic limits. This progressive dysfunctional (allostatic) state is characterized by changes in neuroendocrine and brain stress pathways that underlie expression of withdrawal symptoms that reflect a negative affective state (dysphoria, anxiety), as well as increased motivation to self-administer alcohol. This review highlights literature supportive of this theoretical framework for alcohol addiction. In particular, evidence for stress-related neural, physiological, and behavioral changes associated with chronic alcohol exposure and withdrawal experience is presented. Additionally, this review focuses on the effects of chronic alcohol-induced changes in several pro-stress neuropeptides (corticotropin-releasing factor, dynorphin) and anti-stress neuropeptide systems (nocicepton, neuropeptide Y, oxytocin) in contributing to the stress, negative emotional, and motivational consequences of chronic alcohol exposure. Studies involving use of animal models have significantly increased our understanding of the dynamic stress-related physiological mechanisms and psychological underpinnings of alcohol addiction. This, in turn, is crucial for developing new and more effective therapeutics for treating excessive, harmful drinking, particularly stress-enhanced alcohol consumption. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Department of Neuroscience, Medical University of South Carolina, RHJ Department of Veterans Affairs, Charleston, SC 29464, USA.
| |
Collapse
|
113
|
Kwako LE, Momenan R, Grodin EN, Litten RZ, Koob GF, Goldman D. Addictions Neuroclinical Assessment: A reverse translational approach. Neuropharmacology 2017; 122:254-264. [PMID: 28283392 PMCID: PMC5569299 DOI: 10.1016/j.neuropharm.2017.03.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/02/2017] [Accepted: 03/04/2017] [Indexed: 12/21/2022]
Abstract
Incentive salience, negative emotionality, and executive function are functional domains that are etiologic in the initiation and progression of addictive disorders, having been implicated in humans with addictive disorders and in animal models of addictions. Measures of these three neuroscience-based functional domains can capture much of the effects of inheritance and early exposures that lead to trait vulnerability shared across different addictive disorders. For specific addictive disorders, these measures can be supplemented by agent specific measures such as those that access pharmacodynamic and pharmacokinetic variation attributable to agent-specific gatekeeper molecules including receptors and drug-metabolizing enzymes. Herein, we focus on the translation and reverse translation of knowledge derived from animal models of addiction to the human condition via measures of neurobiological processes that are orthologous in animals and humans, and that are shared in addictions to different agents. Based on preclinical data and human studies, measures of these domains in a general framework of an Addictions Neuroclinical Assessment (ANA) can transform the assessment and nosology of addictive disorders, and can be informative for staging disease progression. We consider next steps and challenges for implementation of ANA in clinical care and research. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Laura E Kwako
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Reza Momenan
- Clinical Neuroimaging Research Core, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Erica N Grodin
- Clinical Neuroimaging Research Core, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Raye Z Litten
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - George F Koob
- Office of the Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David Goldman
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA; Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
114
|
Peng J, Long B, Yuan J, Peng X, Ni H, Li X, Gong H, Luo Q, Li A. A Quantitative Analysis of the Distribution of CRH Neurons in Whole Mouse Brain. Front Neuroanat 2017; 11:63. [PMID: 28790896 PMCID: PMC5524767 DOI: 10.3389/fnana.2017.00063] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/11/2017] [Indexed: 11/13/2022] Open
Abstract
Corticotropin-releasing hormone (CRH), with widespread expression in the brain, plays a key role in modulating a series of behaviors, including anxiety, arousal, motor function, learning and memory. Previous studies have focused on some brain regions with densely distributed CRH neurons such as paraventricular hypothalamic nucleus (PVH) and bed nuclei of the stria terminalis (BST) and revealed some basic structural and functional knowledge of CRH neurons. However, there is no systematic analysis of brain-wide distribution of CRH neurons. Here, we performed a comprehensive study of CRH neurons in CRH-IRES-Cre;Ai3 mice via automatic imaging and stereoscopic cell counting in a whole mouse brain. We acquired four datasets of the CRH distributions with co-localized cytoarchitecture at a voxel resolution of 0.32 μm × 0.32 μm × 2 μm using brain-wide positioning system (BPS). Next, we precisely located and counted the EYFP-labeled neurons in different regions according to propidium iodide counterstained anatomical reference using Neuronal Global Position System. In particular, dense EYFP expression was found in piriform area, BST, central amygdalar nucleus, PVH, Barrington's nucleus, and inferior olivary complex. Considerable CRH neurons were also found in main olfactory bulb, medial preoptic nucleus, pontine gray, tegmental reticular nucleus, external cuneate nucleus, and midline thalamus. We reconstructed and compared the soma morphology of CRH neurons in 11 brain regions. The results demonstrated that CRH neurons had regional diversities of both cell distribution and soma morphology. This anatomical knowledge enhances the current understanding of the functions of CRH neurons. These results also demonstrated the ability of our platform to accurately orient, reconstruct and count neuronal somas in three-dimension for type-specific neurons in the whole brain, making it feasible to answer the fundamental neuroscience question of exact numbers of various neurons in the whole brain.
Collapse
Affiliation(s)
- Jie Peng
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| | - Ben Long
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| | - Jing Yuan
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| | - Xue Peng
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| | - Hong Ni
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| | - Xiangning Li
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| | - Hui Gong
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| | - Qingming Luo
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| | - Anan Li
- Collaborative Innovation Center for Biomedical Engineering, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and TechnologyWuhan, China.,Britton Chance Center, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China.,MOE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and TechnologyWuhan, China
| |
Collapse
|
115
|
Li J, Baird MA, Davis MA, Tai W, Zweifel LS, Adams Waldorf KM, Gale M, Rajagopal L, Pierce RH, Gao X. Dramatic enhancement of the detection limits of bioassays via ultrafast deposition of polydopamine. Nat Biomed Eng 2017; 1. [PMID: 29082104 PMCID: PMC5654575 DOI: 10.1038/s41551-017-0082] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The ability to detect biomarkers with ultrahigh sensitivity radically transformed biology and disease diagnosis. However, owing to incompatibilities with infrastructure in current biological and medical laboratories, recent innovations in analytical technology have not received broad adoption. Here, we report a simple, universal ‘add-on’ technology (dubbed EASE) that can be directly plugged into the routine practices of current research and clinical laboratories and that converts the ordinary sensitivities of common bioassays to extraordinary ones. The assay relies on the bioconjugation capabilities and ultrafast and localized deposition of polydopamine at the target site, which permit a large number of reporter molecules to be captured and lead to detection-sensitivity enhancements exceeding 3 orders of magnitude. The application of EASE in the enzyme-linked-immunosorbent-assay-based detection of the HIV antigen in blood from patients leads to a sensitivity lower than 3 fg ml−1. We also show that EASE allows for the direct visualization, in tissues, of the Zika virus and of low-abundance biomarkers related to neurological diseases and cancer immunotherapy.
Collapse
Affiliation(s)
- Junwei Li
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Madison A Baird
- Department of Pharmacology, University of Washington, Seattle, WA 98105, USA
| | - Michael A Davis
- Department of Immunology, University of Washington, Seattle, WA 98105, USA.,Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98105, USA
| | - Wanyi Tai
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98105, USA.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98105, USA
| | - Kristina M Adams Waldorf
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98105, USA.,Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98105, USA.,Department of Global Health, University of Washington, Seattle, WA 98105, USA.,Sahlgrenska Academy, Gothenburg University, Sweden
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA 98105, USA.,Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98105, USA.,Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Lakshmi Rajagopal
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98105, USA.,Department of Pediatrics, University of Washington, Seattle, WA 98105, USA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Robert H Pierce
- Fred Hutchinson Cancer Research Center, Program in Immunology, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Xiaohu Gao
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
116
|
Corticosteroid modulation and testosterone changes during alcohol intoxication affects voluntary alcohol drinking. Pharmacol Biochem Behav 2017; 157:9-15. [DOI: 10.1016/j.pbb.2017.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/19/2017] [Accepted: 04/21/2017] [Indexed: 11/20/2022]
|
117
|
Jokinen J, Boström AE, Chatzittofis A, Ciuculete DM, Öberg KG, Flanagan JN, Arver S, Schiöth HB. Methylation of HPA axis related genes in men with hypersexual disorder. Psychoneuroendocrinology 2017; 80:67-73. [PMID: 28319850 DOI: 10.1016/j.psyneuen.2017.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/26/2017] [Accepted: 03/03/2017] [Indexed: 12/21/2022]
Abstract
Hypersexual Disorder (HD) defined as non-paraphilic sexual desire disorder with components of compulsivity, impulsivity and behavioral addiction, and proposed as a diagnosis in the DSM 5, shares some overlapping features with substance use disorder including common neurotransmitter systems and dysregulated hypothalamic-pituitary-adrenal (HPA) axis function. In this study, comprising 67 HD male patients and 39 male healthy volunteers, we aimed to identify HPA-axis coupled CpG-sites, in which modifications of the epigenetic profile are associated with hypersexuality. The genome-wide methylation pattern was measured in whole blood using the Illumina Infinium Methylation EPIC BeadChip, measuring the methylation state of over 850K CpG sites. Prior to analysis, the global DNA methylation pattern was pre-processed according to standard protocols and adjusted for white blood cell type heterogeneity. We included CpG sites located within 2000bp of the transcriptional start site of the following HPA-axis coupled genes: Corticotropin releasing hormone (CRH), corticotropin releasing hormone binding protein (CRHBP), corticotropin releasing hormone receptor 1 (CRHR1), corticotropin releasing hormone receptor 2 (CRHR2), FKBP5 and the glucocorticoid receptor (NR3C1). We performed multiple linear regression models of methylation M-values to a categorical variable of hypersexuality, adjusting for depression, dexamethasone non-suppression status, Childhood Trauma Questionnaire total score and plasma levels of TNF-alpha and IL-6. Of 76 tested individual CpG sites, four were nominally significant (p<0.05), associated with the genes CRH, CRHR2 and NR3C1. Cg23409074-located 48bp upstream of the transcription start site of the CRH gene - was significantly hypomethylated in hypersexual patients after corrections for multiple testing using the FDR-method. Methylation levels of cg23409074 were positively correlated with gene expression of the CRH gene in an independent cohort of 11 healthy male subjects. The methylation levels at the identified CRH site, cg23409074, were significantly correlated between blood and four different brain regions. CRH is an important integrator of neuroendocrine stress responses in the brain, with a key role in the addiction processes. Our results show epigenetic changes in the CRH gene related to hypersexual disorder in men.
Collapse
Affiliation(s)
- Jussi Jokinen
- Department of Clinical Neuroscience/Psychiatry, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Sciences/Psychiatry, Umeå University, Umeå, Sweden.
| | | | - Andreas Chatzittofis
- Department of Clinical Neuroscience/Psychiatry, Karolinska Institutet, Stockholm, Sweden; Medical School, University of Cyprus, Nicosia, Cyprus
| | | | | | - John N Flanagan
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Arver
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Helgi B Schiöth
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
118
|
Abstract
W. Horsley Gantt and Joseph V. Brady laid a rich foundation for understanding the concept of emotion, derived from 2 prominent traditions of physiology and psychology: classic conditioning and operant conditioning, respectively. This framework guided my fierce interest in motivation in general and the interaction between reward and stress, which began at John Hopkins with my thesis work under the guidance of Drs. Zoltan Annau, Solomon Synder, and Joseph Brady, among many others. Using the study of the neurobiology of addiction as a framework, I argue that drug addiction not only involves positive reinforcement associated with the rewarding effects of drugs of abuse but also involves another major source of reinforcement, specifically negative reinforcement driven by negative emotional states (termed the "dark side" of addiction). Excessive activation of the brain reward systems leads to antireward or a decrease in the function of normal reward-related neurocircuitry and persistent recruitment of the brain stress systems, both of which may be neurobiologically linked. Understanding the neuroplasticity of the neurocircuitry that comprises the negative reinforcement associated with addiction is a key to understanding negative emotional states in general and their pathophysiology.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
119
|
Thorsell A, Mathé AA. Neuropeptide Y in Alcohol Addiction and Affective Disorders. Front Endocrinol (Lausanne) 2017; 8:178. [PMID: 28824541 PMCID: PMC5534438 DOI: 10.3389/fendo.2017.00178] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 07/07/2017] [Indexed: 12/31/2022] Open
Abstract
Neuropeptide Y (NPY), a neuropeptide highly conserved throughout evolution, is present at high levels in the central nervous system (CNS), as well as in peripheral tissues such as the gut and cardiovascular system. The peptide exerts its effects via multiple receptor subtypes, all belonging to the G-protein-coupled receptor superfamily. Of these subtypes, the Y1 and the Y2 are the most thoroughly characterized, followed by the Y5 subtype. NPY and its receptors have been shown to be of importance in central regulation of events underlying, for example, affective disorders, drug/alcohol use disorders, and energy homeostasis. Furthermore, within the CNS, NPY also affects sleep regulation and circadian rhythm, memory function, tissue growth, and plasticity. The potential roles of NPY in the etiology and pathophysiology of mood and anxiety disorders, as well as alcohol use disorders, have been extensively studied. This focus was prompted by early indications for an involvement of NPY in acute responses to stress, and, later, also data pointing to a role in alterations within the CNS during chronic, or repeated, exposure to adverse events. These functions of NPY, in addition to the peptide's regulation of disease states, suggest that modulation of the activity of the NPY system via receptor agonists/antagonists may be a putative treatment mechanism in affective disorders as well as alcohol use disorders. In this review, we present an overview of findings with regard to the NPY system in relation to anxiety and stress, acute as well as chronic; furthermore we discuss post-traumatic stress disorder and, in part depression. In addition, we summarize findings on alcohol use disorders and related behaviors. Finally, we briefly touch upon genetic as well as epigenetic mechanisms that may be of importance for NPY function and regulation. In conclusion, we suggest that modulation of NPY-ergic activity within the CNS, via ligands aimed at different receptor subtypes, may be attractive targets for treatment development for affective disorders, as well as for alcohol use disorders.
Collapse
Affiliation(s)
- Annika Thorsell
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- *Correspondence: Annika Thorsell,
| | - Aleksander A. Mathé
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
120
|
Dragan WŁ, Czerski PM, Dragan M. PAC1 receptor ( ADCYAP1R1) genotype and problematic alcohol use in a sample of young women. Neuropsychiatr Dis Treat 2017; 13:1483-1489. [PMID: 28652748 PMCID: PMC5473483 DOI: 10.2147/ndt.s137331] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Recent studies revealed the role of the PAC1 (ADCYAP1R1) gene variability in vulnerability to posttraumatic stress disorder in women. Due to the relatively high comorbidity of posttraumatic stress disorder and substance use disorder, we hypothesized about possible associations between PAC1 gene and problematic alcohol use. METHOD The sample studied consisted of 491 women aged 18-28 years (mean age =21.76 years; SD =1.83) and the Alcohol Use Disorders Identification Test was used to assess drinking problems. We successfully genotyped 17 single-nucleotide polymorphisms in the PAC1 gene. RESULTS Single locus analysis revealed a significant (after correction for multiple testing) association between intronic polymorphism rs2302475 and problematic alcohol use (P=0.00048; recessive model). This result was strengthened by the haplotype analysis (P=0.00379). CONCLUSION Our results suggest that the PACAP/PAC1 signaling system is implicated in the development of problematic alcohol use in women.
Collapse
Affiliation(s)
- Wojciech Łukasz Dragan
- The Interdisciplinary Center for Behavior Genetic Research, Faculty of Psychology, University of Warsaw, Warsaw
| | - Piotr M Czerski
- Laboratory of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan
| | | |
Collapse
|
121
|
Litvin Y, Turner CA, Rios MB, Maras PM, Chaudhury S, Baker MR, Blandino P, Watson SJ, Akil H, McEwen B. Fibroblast growth factor 2 alters the oxytocin receptor in a developmental model of anxiety-like behavior in male rat pups. Horm Behav 2016; 86:64-70. [PMID: 27693608 PMCID: PMC5789801 DOI: 10.1016/j.yhbeh.2016.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 09/21/2016] [Accepted: 09/26/2016] [Indexed: 12/21/2022]
Abstract
We aimed to determine the short-term effects of early-life stress in the form of maternal separation (MS) on anxiety-like behavior in male rat pups. In order to assess anxiety, we measured 40kHz separation-induced ultrasonic vocalizations (USV) on postnatal day (PND) 11. We further aimed to evaluate the potential involvement of two neurochemical systems known to regulate social and anxiety-like behaviors throughout life: oxytocin (OT) and fibroblast growth factor 2 (FGF2). For these purposes, we tested the effects of neonatal administration (on PND1) of an acute dose of FGF2 on USV and its potential interaction with MS. In addition, we validated the anxiolytic effects of OT and measured oxytocin receptor (OTR) gene expression, binding and epigenetic regulation via histone acetylation. Our results show that MS potentiated USV while acute administration of OT and FGF2 attenuated them. Further, we found that both FGF2 and MS increased OTR gene expression and the association of acH3K14 with the OTR promoter in the bed nucleus of the stria terminalis (BNST). Comparable changes, though not as pronounced, were also found for the central amygdala (CeA). Our findings suggest that FGF2 may exert its anxiolytic effects in male MS rats by a compensatory increase in the acetylation of the OTR promoter to overcome reduced OT levels in the BNST.
Collapse
Affiliation(s)
- Yoav Litvin
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States
| | - Cortney A Turner
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Mariel B Rios
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States
| | - Pamela M Maras
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Sraboni Chaudhury
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Miriam R Baker
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States
| | - Peter Blandino
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Stanley J Watson
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Huda Akil
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Bruce McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States.
| |
Collapse
|
122
|
Slater PG, Yarur HE, Gysling K. Corticotropin-Releasing Factor Receptors and Their Interacting Proteins: Functional Consequences. Mol Pharmacol 2016; 90:627-632. [PMID: 27612874 DOI: 10.1124/mol.116.104927] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/08/2016] [Indexed: 02/14/2025] Open
Abstract
The corticotropin-releasing factor (CRF) system, which is involved in stress, addiction, and anxiety disorders such as depression, acts through G-protein-coupled receptors (GPCRs) known as type-1 and type-2 CRF receptors. The purpose of this review is to highlight recent advances in the interactions of CRF receptors with other GPCRs and non-GPCR proteins and their associated functional consequences. A better understanding of these interactions may generate new pharmacological alternatives for the treatment of addiction and stress-related disorders.
Collapse
Affiliation(s)
- Paula G Slater
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hector E Yarur
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katia Gysling
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
123
|
Quadros IMH, Macedo GC, Domingues LP, Favoretto CA. An Update on CRF Mechanisms Underlying Alcohol Use Disorders and Dependence. Front Endocrinol (Lausanne) 2016; 7:134. [PMID: 27818644 PMCID: PMC5073134 DOI: 10.3389/fendo.2016.00134] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/09/2016] [Indexed: 12/31/2022] Open
Abstract
Alcohol is the most commonly used and abused substance worldwide. The emergence of alcohol use disorders, and alcohol dependence in particular, is accompanied by functional changes in brain reward and stress systems, which contribute to escalated alcohol drinking and seeking. Corticotropin-releasing factor (CRF) systems have been critically implied in the transition toward problematic alcohol drinking and alcohol dependence. This review will discuss how dysregulation of CRF function contributes to the vulnerability for escalated alcohol drinking and other consequences of alcohol consumption, based on preclinical evidence. CRF signaling, mostly via CRF1 receptors, seems to be particularly important in conditions of excessive alcohol taking and seeking, including during early and protracted withdrawal, relapse, as well as during withdrawal-induced anxiety and escalated aggression promoted by alcohol. Modulation of CRF1 function seems to exert a less prominent role over low to moderate alcohol intake, or to species-typical behaviors. While CRF mechanisms in the hypothalamic-pituitary-adrenal axis have some contribution to the neurobiology of alcohol abuse and dependence, a pivotal role for extra-hypothalamic CRF pathways, particularly in the extended amygdala, is well characterized. More recent studies further suggest a direct modulation of brain reward function by CRF signaling in the ventral tegmental area, nucleus accumbens, and the prefrontal cortex, among other structures. This review will further discuss a putative role for other components of the CRF system that contribute for the overall balance of CRF function in reward and stress pathways, including CRF2 receptors, CRF-binding protein, and urocortins, a family of CRF-related peptides.
Collapse
Affiliation(s)
- Isabel Marian Hartmann Quadros
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Giovana Camila Macedo
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Liz Paola Domingues
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Cristiane Aparecida Favoretto
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
124
|
Henckens MJAG, Deussing JM, Chen A. Region-specific roles of the corticotropin-releasing factor-urocortin system in stress. Nat Rev Neurosci 2016; 17:636-51. [PMID: 27586075 DOI: 10.1038/nrn.2016.94] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dysregulation of the corticotropin-releasing factor (CRF)-urocortin (UCN) system has been implicated in stress-related psychopathologies such as depression and anxiety. It has been proposed that CRF-CRF receptor type 1 (CRFR1) signalling promotes the stress response and anxiety-like behaviour, whereas UCNs and CRFR2 activation mediate stress recovery and the restoration of homeostasis. Recent findings, however, provide clear evidence that this view is overly simplistic. Instead, a more complex picture has emerged that suggests that there are brain region- and cell type-specific effects of CRFR signalling that are influenced by the individual's prior experience and that shape molecular, cellular and ultimately behavioural responses to stressful challenges.
Collapse
Affiliation(s)
- Marloes J A G Henckens
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany.,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
125
|
Abstract
The main characteristic of alcohol use disorder is the consumption of large quantities of alcohol despite the negative consequences. The transition from the moderate use of alcohol to excessive, uncontrolled alcohol consumption results from neuroadaptations that cause aberrant motivational learning and memory processes. Here, we examine studies that have combined molecular and behavioural approaches in rodents to elucidate the molecular mechanisms that keep the social intake of alcohol in check, which we term 'stop pathways', and the neuroadaptations that underlie the transition from moderate to uncontrolled, excessive alcohol intake, which we term 'go pathways'. We also discuss post-transcriptional, genetic and epigenetic alterations that underlie both types of pathways.
Collapse
Affiliation(s)
- Dorit Ron
- Corresponding author: Dorit Ron, 675 Nelson Rising Lane, BOX 0663, San Francisco, CA 94143-0663,
| | - Segev Barak
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
- School of Psychological Sciences and Sagol School of Neuroscience, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| |
Collapse
|
126
|
Koob GF. Corticotropin-Releasing Factor From Rodents to Primates: Translational Hope Expresses Itself, Pun Intended. Biol Psychiatry 2016; 80:340-2. [PMID: 27499011 PMCID: PMC6145175 DOI: 10.1016/j.biopsych.2016.06.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 10/21/2022]
|
127
|
Boyson CO, Holly EN, Burke AR, Montagud-Romero S, DeBold JF, Miczek KA. Maladaptive choices by defeated rats: link between rapid approach to social threat and escalated cocaine self-administration. Psychopharmacology (Berl) 2016; 233:3173-86. [PMID: 27376946 PMCID: PMC4990818 DOI: 10.1007/s00213-016-4363-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
Abstract
RATIONALE Intermittent social defeat stress engenders persistent neuroadaptations and can result in later increased cocaine taking and seeking. However, there are individual differences in stress-escalated cocaine self-administration behavior, which may be a direct result of individual differences in the manner in which rats experience social defeat stress. OBJECTIVE The present study dissected the discrete behavioral phases of social defeat and analyzed which behavioral characteristics may be predictive of subsequent cocaine self-administration. METHODS Male Long-Evans rats underwent nine intermittent social defeat episodes over 21 days in a three-compartment apparatus permitting approach to and escape from a confrontation with an aggressive resident rat. Rats then self-administered intravenous cocaine, which culminated in a 24-h unlimited access "binge." Behaviors during social defeat and cocaine self-administration were evaluated by principal component analysis (PCA). RESULTS PCA revealed that the latency to enter the threatening environment was highly predictive of later cocaine self-administration during the 24-h binge. This behavior was not associated with other cocaine-predictive traits, such as reactivity to novelty in an open field, saccharin preference, and motor impulsivity. Additionally, there was no effect of latency to enter a threatening environment on physiological measures of stress, including plasma corticosterone and corticotropin releasing factor (CRF) in the extended amygdala. However, latency to enter the threatening environment was negatively correlated with brain-derived neurotropic factor (BDNF) and its receptor, tyrosine kinase B (TrkB) in the hippocampus. CONCLUSION These data suggest that latency to enter a threatening environment is a novel behavioral characteristic predictive of later cocaine self-administration.
Collapse
Affiliation(s)
- Christopher O. Boyson
- Department of Psychology, Tufts University, Bacon Hall, 530 Boston Ave, Medford, MA 02155, USA
| | - Elizabeth N. Holly
- Department of Psychology, Tufts University, Bacon Hall, 530 Boston Ave, Medford, MA 02155, USA
,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew R. Burke
- Department of Psychology, Tufts University, Bacon Hall, 530 Boston Ave, Medford, MA 02155, USA
| | - Sandra Montagud-Romero
- Department of Psychology, Tufts University, Bacon Hall, 530 Boston Ave, Medford, MA 02155, USA
,Unidad de Investigación Psicobiología de las Drogodependencias, Departmento de Psicobiología, Universitat de València, València, Spain
| | - Joseph F. DeBold
- Department of Psychology, Tufts University, Bacon Hall, 530 Boston Ave, Medford, MA 02155, USA
| | - Klaus A. Miczek
- Department of Psychology, Tufts University, Bacon Hall, 530 Boston Ave, Medford, MA 02155, USA
,Department of Psychiatry, Tufts University School of Medicine, Boston, MA, USA
,Department of Pharmacology, Tufts University School of Medicine, Boston, MA, USA
,Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
128
|
Solanki RR, Scholl JL, Watt MJ, Renner KJ, Forster GL. Amphetamine Withdrawal Differentially Increases the Expression of Organic Cation Transporter 3 and Serotonin Transporter in Limbic Brain Regions. J Exp Neurosci 2016; 10:93-100. [PMID: 27478387 PMCID: PMC4957605 DOI: 10.4137/jen.s40231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/26/2022] Open
Abstract
Amphetamine withdrawal increases anxiety and stress sensitivity related to blunted ventral hippocampus (vHipp) and enhances the central nucleus of the amygdala (CeA) serotonin responses. Extracellular serotonin levels are regulated by the serotonin transporter (SERT) and organic cation transporter 3 (OCT3), and vHipp OCT3 expression is enhanced during 24 hours of amphetamine withdrawal, while SERT expression is unaltered. Here, we tested whether OCT3 and SERT expression in the CeA is also affected during acute withdrawal to explain opposing regional alterations in limbic serotonergic neurotransmission and if respective changes continued with two weeks of withdrawal. We also determined whether changes in transporter expression were confined to these regions. Male rats received amphetamine or saline for two weeks followed by 24 hours or two weeks of withdrawal, with transporter expression measured using Western immunoblot. OCT3 and SERT expression increased in the CeA at both withdrawal timepoints. In the vHipp, OCT3 expression increased only at 24 hours of withdrawal, with an equivalent pattern seen in the dorsomedial hypothalamus. No changes were evident in any other regions sampled. These regionally specific changes in limbic OCT3 and SERT expression may partially contribute to the serotonergic imbalance and negative affect during amphetamine withdrawal.
Collapse
Affiliation(s)
- Rajeshwari R. Solanki
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Jamie L. Scholl
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Michael J. Watt
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Kenneth J. Renner
- Biology Department, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Gina L. Forster
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
129
|
Barra de la Tremblaye P, Plamondon H. Alterations in the corticotropin-releasing hormone (CRH) neurocircuitry: Insights into post stroke functional impairments. Front Neuroendocrinol 2016; 42:53-75. [PMID: 27455847 DOI: 10.1016/j.yfrne.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/04/2016] [Accepted: 07/06/2016] [Indexed: 10/21/2022]
Abstract
Although it is well accepted that changes in the regulation of the hypothalamic-pituitary adrenal (HPA) axis may increase susceptibility to affective disorders in the general population, this link has been less examined in stroke patients. Yet, the bidirectional association between depression and cardiovascular disease is strong, and stress increases vulnerability to stroke. Corticotropin-releasing hormone (CRH) is the central stress hormone of the HPA axis pathway and acts by binding to CRH receptors (CRHR) 1 and 2, which are located in several stress-related brain regions. Evidence from clinical and animal studies suggests a role for CRH in the neurobiological basis of depression and ischemic brain injury. Given its importance in the regulation of the neuroendocrine, autonomic, and behavioral correlates of adaptation and maladaptation to stress, CRH is likely associated in the pathophysiology of post stroke emotional impairments. The goals of this review article are to examine the clinical and experimental data describing (1) that CRH regulates the molecular signaling brain circuit underlying anxiety- and depression-like behaviors, (2) the influence of CRH and other stress markers in the pathophysiology of post stroke emotional and cognitive impairments, and (3) context and site specific interactions of CRH and BDNF as a basis for the development of novel therapeutic targets. This review addresses how the production and release of the neuropeptide CRH within the various regions of the mesocorticolimbic system influences emotional and cognitive behaviors with a look into its role in psychiatric disorders post stroke.
Collapse
Affiliation(s)
- P Barra de la Tremblaye
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada
| | - H Plamondon
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
130
|
Belmer A, Patkar OL, Pitman KM, Bartlett SE. Serotonergic Neuroplasticity in Alcohol Addiction. Brain Plast 2016; 1:177-206. [PMID: 29765841 PMCID: PMC5928559 DOI: 10.3233/bpl-150022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Alcohol addiction is a debilitating disorder producing maladaptive changes in the brain, leading drinkers to become more sensitive to stress and anxiety. These changes are key factors contributing to alcohol craving and maintaining a persistent vulnerability to relapse. Serotonin (5-Hydroxytryptamine, 5-HT) is a monoamine neurotransmitter widely expressed in the central nervous system where it plays an important role in the regulation of mood. The serotonin system has been extensively implicated in the regulation of stress and anxiety, as well as the reinforcing properties of all of the major classes of drugs of abuse, including alcohol. Dysregulation within the 5-HT system has been postulated to underlie the negative mood states associated with alcohol use disorders. This review will describe the serotonergic (5-HTergic) neuroplastic changes observed in animal models throughout the alcohol addiction cycle, from prenatal to adulthood exposure. The first section will focus on alcohol-induced 5-HTergic neuroadaptations in offspring prenatally exposed to alcohol and the consequences on the regulation of stress/anxiety. The second section will compare alterations in 5-HT signalling induced by acute or chronic alcohol exposure during adulthood and following alcohol withdrawal, highlighting the impact on the regulation of stress/anxiety signalling pathways. The third section will outline 5-HTergic neuroadaptations observed in various genetically-selected ethanol preferring rat lines. Finally, we will discuss the pharmacological manipulation of the 5-HTergic system on ethanol- and anxiety/stress-related behaviours demonstrated by clinical trials, with an emphasis on current and potential treatments.
Collapse
Affiliation(s)
- Arnauld Belmer
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| | - Omkar L Patkar
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| | - Kim M Pitman
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| | - Selena E Bartlett
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.,Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
131
|
Etiological theories of addiction: A comprehensive update on neurobiological, genetic and behavioural vulnerability. Pharmacol Biochem Behav 2016; 148:59-68. [PMID: 27306332 DOI: 10.1016/j.pbb.2016.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 06/01/2016] [Accepted: 06/10/2016] [Indexed: 12/21/2022]
Abstract
Currently, about 246 million people around the world have used an illicit drug. The reasons for this use are multiple: e.g. to augment the sensation of pleasure or to reduce the withdrawal and other aversive effects of a given substance. This raises the problem of addiction, which remains a disease of modern society. This review offers a comprehensive update of the different theories about the etiology of addictive behaviors with emphasis on the neurobiological, environmental, psychopathological, behavioural and genetic aspects of addictions, discussed from an evolutionary perspective. The main conclusion of this review is that vulnerability to drug addiction suggests an interaction between many brain systems (including the reward, decision-making, serotonergic, oxytocin, interoceptive insula, CRF, norepinephrine, dynorphin/KOR, orexin and vasopressin systems), genetic predisposition, sociocultural context, impulsivity and drugs types. Further advances in biological and psychological science are needed to address the problems of addiction at its roots.
Collapse
|
132
|
Georgiou P, Zanos P, Garcia-Carmona JA, Hourani S, Kitchen I, Laorden ML, Bailey A. Methamphetamine abstinence induces changes in μ-opioid receptor, oxytocin and CRF systems: Association with an anxiogenic phenotype. Neuropharmacology 2016; 105:520-532. [DOI: 10.1016/j.neuropharm.2016.02.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/14/2016] [Accepted: 02/14/2016] [Indexed: 01/08/2023]
|
133
|
Fattore L, Diana M. Drug addiction: An affective-cognitive disorder in need of a cure. Neurosci Biobehav Rev 2016; 65:341-61. [DOI: 10.1016/j.neubiorev.2016.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/24/2016] [Accepted: 04/11/2016] [Indexed: 12/22/2022]
|
134
|
Liew OW, Yandle TG, Chong JPC, Ng YX, Frampton CM, Ng TP, Lam CSP, Richards AM. High-Sensitivity Sandwich ELISA for Plasma NT-proUcn2: Plasma Concentrations and Relationship to Mortality in Heart Failure. Clin Chem 2016; 62:856-65. [DOI: 10.1373/clinchem.2015.252932] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/21/2016] [Indexed: 01/01/2023]
Abstract
Abstract
BACKGROUND
Urocortin 2 (Ucn2) has powerful hemodynamic, renal, and neurohormonal actions and likely participates in normal circulatory homeostasis and the compensatory response to heart failure (HF). A validated assay for endogenous circulating Ucn2 would facilitate investigations into Ucn2 physiology and elucidate its derangement and potential as a biomarker in heart disease.
METHOD
We developed a chemiluminescence-based sandwich ELISA to measure plasma N-terminal (NT)-proUcn2 in non-HF patients (control; n = 160) and HF patients with reduced (HFREF; n = 134) and preserved (HFPEF; n = 121) left ventricular ejection fraction (LVEF).
RESULTS
The ELISA had a limit of detection of 8.47 ng/L (1.52 pmol/L) and working range of 23.8–572 ng/L. Intra- and interassay CV and total error were 4.8, 16.2, and 17.7%, respectively. The median (interquartile range) plasma NT-proUcn2 concentration in controls was 112 (86–132) ng/L. HFREF, HFPEF, and all HF plasma concentrations were significantly increased [117 (98–141) ng/L, P = 0.0007; 119 (93–136) ng/L, P = 0.0376, and 119 (97–140) ng/L, P = 0.001] compared with controls but did not differ significantly between HFREF and HFPEF. NT-proUcn2 was modestly related to age (r = 0.264, P = 0.001) and cardiac troponin T (r = 0.258, P = 0.001) but not N-terminal pro-B-type natriuretic peptide, body mass index, LVEF, or estimated glomerular filtration rate. On multivariate analysis, plasma NT-proUcn2 was independently and inversely related to 2-year mortality in HF.
CONCLUSIONS
The validated ELISA measured human NT-proUcn2 in plasma and showed modest but significant increases in HF patients compared with controls. In HF, the unusual inverse relationship between plasma NT-proUcn2 and 2-year mortality portends potential prognostic value but requires further corroboration.
Collapse
Affiliation(s)
- Oi Wah Liew
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Timothy G Yandle
- Christchurch Heart Institute, University of Otago, Otago, New Zealand
| | - Jenny P C Chong
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Yan Xia Ng
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | | | - Tze Pin Ng
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Carolyn S P Lam
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
- Christchurch Heart Institute, University of Otago, Otago, New Zealand
| |
Collapse
|
135
|
Vicario A, Mendoza E, Abellán A, Scharff C, Medina L. Genoarchitecture of the extended amygdala in zebra finch, and expression of FoxP2 in cell corridors of different genetic profile. Brain Struct Funct 2016; 222:481-514. [PMID: 27160258 PMCID: PMC5225162 DOI: 10.1007/s00429-016-1229-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 04/21/2016] [Indexed: 02/01/2023]
Abstract
We used a battery of genes encoding transcription factors (Pax6, Islet1, Nkx2.1, Lhx6, Lhx5, Lhx9, FoxP2) and neuropeptides to study the extended amygdala in developing zebra finches. We identified different components of the central extended amygdala comparable to those found in mice and chickens, including the intercalated amygdalar cells, the central amygdala, and the lateral bed nucleus of the stria terminalis. Many cells likely originate in the dorsal striatal domain, ventral striatal domain, or the pallidal domain, as is the case in mice and chickens. Moreover, a cell subpopulation of the central extended amygdala appears to originate in the prethalamic eminence. As a general principle, these different cells with specific genetic profiles and embryonic origin form separate or partially intermingled cell corridors along the extended amygdala, which may be involved in different functional pathways. In addition, we identified the medial amygdala of the zebra finch. Like in the chickens and mice, it is located in the subpallium and is rich in cells of pallido-preoptic origin, containing minor subpopulations of immigrant cells from the ventral pallium, alar hypothalamus and prethalamic eminence. We also proposed that the medial bed nucleus of the stria terminalis is composed of several parallel cell corridors with different genetic profile and embryonic origin: preoptic, pallidal, hypothalamic, and prethalamic. Several of these cell corridors with distinct origin express FoxP2, a transcription factor implicated in synaptic plasticity. Our results pave the way for studies using zebra finches to understand the neural basis of social behavior, in which the extended amygdala is involved.
Collapse
Affiliation(s)
- Alba Vicario
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain
| | | | - Antonio Abellán
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain
| | | | - Loreta Medina
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain.
| |
Collapse
|
136
|
Vallöf D, Vestlund J, Engel JA, Jerlhag E. The Anorexigenic Peptide Neuromedin U (NMU) Attenuates Amphetamine-Induced Locomotor Stimulation, Accumbal Dopamine Release and Expression of Conditioned Place Preference in Mice. PLoS One 2016; 11:e0154477. [PMID: 27139195 PMCID: PMC4854378 DOI: 10.1371/journal.pone.0154477] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/14/2016] [Indexed: 12/28/2022] Open
Abstract
Amphetamine dependence, besides its substantial economical consequence, is a serious cause of mortality and morbidity. By investigations of the neurochemical correlates through which addictive drugs, such as amphetamine, activate the mesoaccumbal dopamine system unique targets for treatment of drug addiction can be identified. This reward link consists of a dopamine projection from the ventral tegmental area to the nucleus accumbens (NAc) suggesting that these brain areas are important for reward. The physiological function of gut-brain peptides has expanded beyond food intake modulation and involves regulation of drug reinforcement. A novel candidate for reward regulation is the anorexigenic peptide neuromedin U (NMU). We therefore investigated the effects of intracerebroventricular (icv) administration of NMU on amphetamine’s well-documented effects on the mesoaccumbal dopamine system, i.e. locomotor stimulation and accumbal dopamine release in mice. In addition, the effect of accumbal NMU administration on locomotor activity was examined. The effect of NMU, icv or intra-NAc, on the expression of conditioned place preference (CPP) was elucidated. Firstly, we showed that icv administration of NMU attenuate the amphetamine-induced locomotor stimulation, accumbal dopamine release and expression of CPP in mice. Secondly, we found that a lower dose of NMU (icv) reduce the amphetamine-induced locomotor stimulation in mice. Thirdly, we demonstrated that NMU administration into the NAc block the ability of amphetamine to cause a locomotor stimulation in mice. However, accumbal NMU administration did not attenuate the amphetamine-induced expression of CPP in mice. Our novel data suggest that central NMU signalling is involved in development of amphetamine dependence.
Collapse
Affiliation(s)
- Daniel Vallöf
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jesper Vestlund
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jörgen A Engel
- 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
|
137
|
Castilla-Ortega E, Serrano A, Blanco E, Araos P, Suárez J, Pavón FJ, Rodríguez de Fonseca F, Santín LJ. A place for the hippocampus in the cocaine addiction circuit: Potential roles for adult hippocampal neurogenesis. Neurosci Biobehav Rev 2016; 66:15-32. [PMID: 27118134 DOI: 10.1016/j.neubiorev.2016.03.030] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 03/08/2016] [Accepted: 03/08/2016] [Indexed: 02/07/2023]
Abstract
Cocaine addiction is a chronic brain disease in which the drug seeking habits and profound cognitive, emotional and motivational alterations emerge from drug-induced neuroadaptations on a vulnerable brain. Therefore, a 'cocaine addiction brain circuit' has been described to explain this disorder. Studies in both cocaine patients and rodents reveal the hippocampus as a main node in the cocaine addiction circuit. The contribution of the hippocampus to cocaine craving and the associated memories is essential to understand the chronic relapsing nature of addiction, which is the main obstacle for the recovery. Interestingly, the hippocampus holds a particular form of plasticity that is rare in the adult brain: the ability to generate new functional neurons. There is an active scientific debate on the contributions of these new neurons to the addicted brain. This review focuses on the potential role(s) of adult hippocampal neurogenesis (AHN) in cocaine addiction. Although the current evidence primarily originates from animal research, these preclinical studies support AHN as a relevant component for the hippocampal effects of cocaine.
Collapse
Affiliation(s)
- Estela Castilla-Ortega
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain.
| | - Antonia Serrano
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Eduardo Blanco
- Departament de Pedagogia i Psicologia, Facultat d'Educació, Psicologia i Treball Social, Universitat de Lleida, Spain
| | - Pedro Araos
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Juan Suárez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Francisco J Pavón
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Spain
| | - Luis J Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Instituto de Investigación Biomédica de Málaga (IBIMA), Facultad de Psicología, Universidad de Málaga, Spain.
| |
Collapse
|
138
|
Poon K, Leibowitz SF. Consumption of Substances of Abuse during Pregnancy Increases Consumption in Offspring: Possible Underlying Mechanisms. Front Nutr 2016; 3:11. [PMID: 27148536 PMCID: PMC4837147 DOI: 10.3389/fnut.2016.00011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 04/04/2016] [Indexed: 12/16/2022] Open
Abstract
Correlative human observational studies on substances of abuse have been highly dependent on the use of rodent models to determine the neuronal and molecular mechanisms that control behavioral outcomes. This is particularly true for gestational exposure to non-illicit substances of abuse, such as excessive dietary fat, ethanol, and nicotine, which are commonly consumed in our society. Exposure to these substances during the prenatal period has been shown in offspring to increase their intake of these substances, induce other behavioral changes, and affect neurochemical systems in several brain areas that are known to control behavior. More importantly, emerging studies are linking the function of the immune system to these neurochemicals and ingestion of these abused substances. This review article will summarize the prenatal rodent models used to study developmental changes in offspring caused by prenatal exposure to dietary fat, ethanol, or nicotine. We will discuss the various techniques used for the administration of these substances into rodents and summarize the published outcomes induced by prenatal exposure to these substances. Finally, this review will cover some of the recent evidence for the role of immune factors in causing these behavioral and neuronal changes.
Collapse
Affiliation(s)
- Kinning Poon
- Laboratory of Behavioral Neurobiology, The Rockefeller University , New York, NY , USA
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University , New York, NY , USA
| |
Collapse
|
139
|
Curran HV, Freeman TP, Mokrysz C, Lewis DA, Morgan CJA, Parsons LH. Keep off the grass? Cannabis, cognition and addiction. Nat Rev Neurosci 2016; 17:293-306. [PMID: 27052382 DOI: 10.1038/nrn.2016.28] [Citation(s) in RCA: 252] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In an increasing number of states and countries, cannabis now stands poised to join alcohol and tobacco as a legal drug. Quantifying the relative adverse and beneficial effects of cannabis and its constituent cannabinoids should therefore be prioritized. Whereas newspaper headlines have focused on links between cannabis and psychosis, less attention has been paid to the much more common problem of cannabis addiction. Certain cognitive changes have also been attributed to cannabis use, although their causality and longevity are fiercely debated. Identifying why some individuals are more vulnerable than others to the adverse effects of cannabis is now of paramount importance to public health. Here, we review the current state of knowledge about such vulnerability factors, the variations in types of cannabis, and the relationship between these and cognition and addiction.
Collapse
Affiliation(s)
- H Valerie Curran
- Clinical Psychopharmacology Unit, University College London, Gower Street, London WC1E 6BT, UK
| | - Tom P Freeman
- Clinical Psychopharmacology Unit, University College London, Gower Street, London WC1E 6BT, UK
| | - Claire Mokrysz
- Clinical Psychopharmacology Unit, University College London, Gower Street, London WC1E 6BT, UK
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, Pennsylvania 15213, USA
| | - Celia J A Morgan
- Clinical Psychopharmacology Unit, University College London, Gower Street, London WC1E 6BT, UK.,Psychopharmacology and Addiction Research Centre, University of Exeter, Perry Road, Exeter EX4 4QG, UK
| | - Loren H Parsons
- The Scripps Research Institute, 10550 N. Torrey Pines Road, SP30-2001, La Jolla, California 92037, USA
| |
Collapse
|
140
|
Partridge JG, Forcelli PA, Luo R, Cashdan JM, Schulkin J, Valentino RJ, Vicini S. Stress increases GABAergic neurotransmission in CRF neurons of the central amygdala and bed nucleus stria terminalis. Neuropharmacology 2016; 107:239-250. [PMID: 27016019 DOI: 10.1016/j.neuropharm.2016.03.029] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/17/2016] [Accepted: 03/21/2016] [Indexed: 11/20/2022]
Abstract
Corticotrophin Releasing Factor (CRF) is a critical stress-related neuropeptide in major output pathways of the amygdala, including the central nucleus (CeA), and in a key projection target of the CeA, the bed nucleus of the stria terminalis (BnST). While progress has been made in understanding the contributions and characteristics of CRF as a neuropeptide in rodent behavior, little attention has been committed to determine the properties and synaptic physiology of specific populations of CRF-expressing (CRF(+)) and non-expressing (CRF(-)) neurons in the CeA and BnST. Here, we fill this gap by electrophysiologically characterizing distinct neuronal subtypes in CeA and BnST. Crossing tdTomato or channelrhodopsin-2 (ChR2-YFP) reporter mice to those expressing Cre-recombinase under the CRF promoter allowed us to identify and manipulate CRF(+) and CRF(-) neurons in CeA and BnST, the two largest areas with fluorescently labeled neurons in these mice. We optogenetically activated CRF(+) neurons to elicit action potentials or synaptic responses in CRF(+) and CRF(-) neurons. We found that GABA is the predominant co-transmitter in CRF(+) neurons within the CeA and BnST. CRF(+) neurons are highly interconnected with CRF(-) neurons and to a lesser extent with CRF(+) neurons. CRF(+) and CRF(-) neurons differentially express tonic GABA currents. Chronic, unpredictable stress increase the amplitude of evoked IPSCs and connectivity between CRF(+) neurons, but not between CRF(+) and CRF(-) neurons in both regions. We propose that reciprocal inhibition of interconnected neurons controls CRF(+) output in these nuclei.
Collapse
Affiliation(s)
- John G Partridge
- Department of Pharmacology & Physiology, Georgetown University School of Medicine, Washington, DC 20007, USA; Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC 20007, USA.
| | - Patrick A Forcelli
- Department of Pharmacology & Physiology, Georgetown University School of Medicine, Washington, DC 20007, USA; Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Ruixi Luo
- Department of Pharmacology & Physiology, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Jonah M Cashdan
- Department of Biology, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Jay Schulkin
- Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC 20007, USA; Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Rita J Valentino
- Abramson Pediatric Research Center, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stefano Vicini
- Department of Pharmacology & Physiology, Georgetown University School of Medicine, Washington, DC 20007, USA; Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC 20007, USA
| |
Collapse
|
141
|
Localized brain differences in Arc expression between mice showing low vs. high propensity to ethanol sensitization. Pharmacol Biochem Behav 2016; 142:15-22. [DOI: 10.1016/j.pbb.2015.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 12/11/2015] [Accepted: 12/13/2015] [Indexed: 02/06/2023]
|
142
|
Baclofen blocks yohimbine-induced increases in ethanol-reinforced responding in rats. Pharmacol Biochem Behav 2016; 144:20-5. [PMID: 26921456 DOI: 10.1016/j.pbb.2016.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/19/2016] [Accepted: 02/22/2016] [Indexed: 12/25/2022]
Abstract
Chronic or repeated stress increases alcohol consumption. The GABA-B agonist baclofen decreases alcohol consumption and may be most effective for individuals with comorbid anxiety/stress disorders. The present study sought to determine if baclofen blocks stress-induced increases in ethanol self-administration as modeled by repeated yohimbine injections in rats. Rats were trained to respond for 15% w/v ethanol in operant chambers using a method that applies neither water deprivation nor saccharin/sucrose fading. Following training, the rats received 6 injections of 1.25mg/kg yohimbine were given immediately prior to the operant sessions during a 2-week time period. Subsequently, some rats were pair-matched to receive either 1.25mg/kg yohimbine or saline in the presence of 0.3, 1, and 3mg/kg baclofen prior to sessions. Acquisition of ethanol self-administration was poor. Pretreatment with yohimbine consistently increased responding across repeated injections. Yohimbine's effect on ethanol intake unexpectedly diverged from the effect on responding as the rats failed to consume all reinforcers earned. Smaller doses of baclofen paired with saline injections had no effect on ethanol responding; only 3mg/kg baclofen reduced ethanol self-administration. The smallest baclofen dose of 0.3mg/kg failed to block the yohimbine-induced increase in self-administration. The large baclofen dose of 3mg/kg continued to suppress ethanol self-administration when given with yohimbine. Baclofen 1mg/kg blocked the effect of yohimbine even though it had no effect when given in the absence of yohimbine. Exposure to high ethanol concentrations may induce self-administration only in certain conditions. The dissociation between responding and intake suggests that repeated yohimbine injections may initiate other behavioral or physiological mechanisms that confound its effects as a pharmacological stressor. Furthermore, an optimal baclofen dose range may specifically protect against stress-induced alcohol self-administration, highlighting a specific contribution of GABA-B receptors and a potential therapeutic efficacy of GABA-B agonists at a non-sedating dose.
Collapse
|
143
|
[Theoretical reflection on the place of memory and temporal cognitive mechanisms in addictive disorders]. Encephale 2016; 42:264-9. [PMID: 26875869 DOI: 10.1016/j.encep.2014.10.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/27/2014] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Addictions can be regarded as cognitive disorders related to neurobiological impairments. On the one hand, some cognitive impairments occur as a result of substance intake and withdrawal upon stopping intake, while, on the other hand, cognitive mechanisms are responsible for initiating and maintaining addiction. In this review, we detail the memory and temporal mechanisms involved in this pathology. METHODOLOGY We reviewed the literature dedicated to the mechanisms of conditioning association between a substance and a context, and the memory and temporal mechanisms involved in the maintenance of addiction. Cognitive impairments in this context are accompanied by both short-term and long-term neurobiological disorders. RESULTS Drug-context conditioning is dependent on learning abilities in rats and humans, and it is the first step towards the development of an addiction. In fact, with the beginning of an addiction, it is the context associated with the substance intake, which determines the reinforcing factors (such as pleasure in the case of drug consumption) for the development of an addiction. Maintenance of addiction is related to the persistence of this association between context and substance. Furthermore, the impulsiveness of patients renders them unable to delay their gratification. Consequently, even if delayed gratifications are more valuable, patients prefer immediate gratification such as substance use. DISCUSSION The memory and temporal mechanisms of addiction are central to the initiation and maintenance of drug addiction. They also affect patients' ability to develop projects for the future. The salience of the memory association between drug and context is accompanied by a decline in autobiographical memories, which become poor and lacking in detail. It is probably these impairments which are responsible for the difficulty that the patients have while investigating their story during psychotherapy. On the other hand, given that even though delayed gratification is greater patients prefer immediate gratification, they have difficulty making plans for the future and constructing their own personality. These cognitive impairments are sustained by neurobiological correlates such as dopamine dysregulation in the short-term and changes in neural plasticity in the cortico-meso-limbic system in the long term. CONCLUSION We reviewed full arguments which highlight that addiction is mediated by cognitive mechanisms which are related on the one hand to clinical symptoms and, on the other hand, to neurobiological alterations. According to the literature, memory and time mechanisms seem to be central to the initiation and maintenance of addictive behaviours. More research is needed to improve our knowledge of the cognitive mechanisms of addiction and to develop new tools for treating patients.
Collapse
|
144
|
Cadoni C. Fischer 344 and Lewis Rat Strains as a Model of Genetic Vulnerability to Drug Addiction. Front Neurosci 2016; 10:13. [PMID: 26903787 PMCID: PMC4746315 DOI: 10.3389/fnins.2016.00013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/11/2016] [Indexed: 01/02/2023] Open
Abstract
Today it is well acknowledged that both nature and nurture play important roles in the genesis of psychopathologies, including drug addiction. Increasing evidence suggests that genetic factors contribute for at least 40–60% of the variation in liability to drug dependence. Human genetic studies suggest that multiple genes of small effect, rather than single genes, contribute to the genesis of behavioral psychopathologies. Therefore, the use of inbred rat strains might provide a valuable tool to identify differences, linked to genotype, important in liability to addiction and related disorders. In this regard, Lewis and Fischer 344 inbred rats have been proposed as a model of genetic vulnerability to drug addiction, given their innate differences in sensitivity to the reinforcing and rewarding effects of drugs of abuse, as well their different responsiveness to stressful stimuli. This review will provide evidence in support of this model for the study of the genetic influence on addiction vulnerability, with particular emphasis on differences in mesolimbic dopamine (DA) transmission, rewarding and emotional function. It will be highlighted that Lewis and Fischer 344 rats differ not only in several indices of DA transmission and adaptive changes following repeated drug exposure, but also in hypothalamic-pituitary-adrenal (HPA) axis responsiveness, influencing not only the ability of the individual to cope with stressful events, but also interfering with rewarding and motivational processes, given the influence of corticosteroids on dopamine neuron functionality. Further differences between the two strains, as impulsivity or anxiousness, might contribute to their different proneness to addiction, and likely these features might be linked to their different DA neurotransmission plasticity. Although differences in other neurotransmitter systems might deserve further investigation, results from the reviewed studies might open new vistas in understanding aberrant deviations in reward and motivational functions.
Collapse
Affiliation(s)
- Cristina Cadoni
- Institute of Neuroscience, Cagliari Section, Department of Biomedical Sciences, National Research Council of ItalyCagliari, Italy; Centre of Excellence "Neurobiology of Dependence", University of CagliariCagliari, Italy
| |
Collapse
|
145
|
Gorky J, Schwaber J. The role of the gut-brain axis in alcohol use disorders. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:234-41. [PMID: 26188287 PMCID: PMC4679635 DOI: 10.1016/j.pnpbp.2015.06.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 02/08/2023]
Abstract
Neuroimmune and inflammatory processes have been locally associated with the amygdala in alcohol exposure and withdrawal. We and others have suggested that this inflammation in the amygdala may cause disturbance of neural function observed as anxiety and autonomic distress in withdrawal. Despite the potential importance of the robust neuroinflammatory response, the mechanisms contributing to this response are not well understood. We review literature that suggests the effects of alcohol, and other substances of abuse, cause dysbiosis of the gut microbiome. This peripheral response may modulate neuroprotective vagal afferent signaling that permits and exacerbates a neuroinflammatory response in the amygdala. We will examine the mounting evidence that suggests that (1) gut dysbiosis contributes to neuroinflammation, especially in the context of alcohol exposure and withdrawal, (2) the neuroinflammation in the amygdala involves the microglia and astrocytes and their effect on neural cells, and (3) amygdala neuroinflammation itself contributes directly to withdrawal behavior and symptoms. The contribution of the gut to an anxiogenic response is a promising therapeutic target for patients suffering with withdrawal symptoms given the safe and well-established methods of modulating the gut microbiome.
Collapse
Affiliation(s)
- Jonathan Gorky
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - James Schwaber
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
146
|
Lalanne L, Lutz PE, Trojak B, Lang JP, Kieffer BL, Bacon E. Medications between psychiatric and addictive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:215-23. [PMID: 26514592 DOI: 10.1016/j.pnpbp.2015.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/02/2015] [Accepted: 10/25/2015] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Many epidemiological studies have revealed a frequent co-occurrence of psychiatric and substance use disorders. The term used in the literature to refer to this co-occurrence is dual diagnosis. The high prevalence of dual diagnosis has led physicians to observe the effects of medication prescribed to treat psychiatric disorders on the co-occurring substance use disorder and vice versa. The concept of medications between psychiatric and addictive disorders stems from these clinical observations, alongside which, however, it has developed from the observation that both psychiatric and substance use disorders share common neurobiological pathways and trigger common cognitive disorders. This has led researchers to develop medications on the basis of neurobiological and cognitive rationales. MATERIAL AND METHOD In our article, we review peculiar medications based on neurobiological and cognitive rationales and that have an impact in both psychiatric and addictive disorders. RESULTS We highlight how interesting these new prescriptions are for clinical observation and for the treatment of patients suffering from dual diagnosis. CONCLUSION We then go on to discuss the interest in them from the perspective of clinical practice and clinical research, in that the development of medications to treat dual diagnosis helps to further our knowledge of both psychiatric and substance use disorders.
Collapse
Affiliation(s)
- Laurence Lalanne
- Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Molé-culaire et Cellulaire, INSERM U-964, CNRS UMR-7104, Université de Strasbourg, France; CHRU de Strasbourg-INSERM1114, Department of Psychiatry, University of Strasbourg, France.
| | - Pierre-Eric Lutz
- McGill Group for Suicide Studies, Douglas Mental Health research Centre, McGill University, Montréal, Canada; Douglas Mental Health Research Centre, McGill University, Montréal, Canada.
| | - Benoit Trojak
- Department of Psychiatry and Addictology, University Hospital of Dijon, France, EA 4452, LPPM, University of Burgundy, France.
| | - Jean-Philippe Lang
- CHRU de Strasbourg-INSERM1114, Department of Psychiatry, University of Strasbourg, France.
| | - Brigitte L Kieffer
- Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Molé-culaire et Cellulaire, INSERM U-964, CNRS UMR-7104, Université de Strasbourg, France; Douglas Mental Health Research Centre, McGill University, Montréal, Canada.
| | - Elisabeth Bacon
- Inserm U-1114, Department of Psychiatry, University of Strasbourg, France.
| |
Collapse
|
147
|
Wscieklica T, de Barros Viana M, Le Sueur Maluf L, Pouza KCP, Spadari RC, Céspedes IC. Alcohol consumption increases locomotion in an open field and induces Fos-immunoreactivity in reward and approach/withdrawal-related neurocircuitries. Alcohol 2016; 50:73-82. [PMID: 26786746 DOI: 10.1016/j.alcohol.2015.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 12/16/2022]
Abstract
Drug addiction is a chronically relapsing disorder characterized by compulsion to seek and take the drug, loss of control in limiting intake and, eventually, the emergence of a negative emotional state when access to the drug is prevented. Both dopamine and corticotropin-releasing factor (CRF)-mediated systems seem to play important roles in the modulation of alcohol abuse and dependence. The present study investigated the effects of alcohol consumption on anxiety and locomotor parameters and on the activation of dopamine and CRF-innervated brain regions. Male Wistar rats were given a choice of two bottles for 31 days, one containing water and the other a solution of saccharin + alcohol. Control animals only received water and a solution of 0.2% saccharin. On the 31st day, animals were tested in the elevated plus-maze and open field, and euthanized immediately after the behavioral tests. An independent group of animals was treated with ethanol and used to measure blood ethanol concentration. Results showed that alcohol intake did not alter behavioral measurements in the plus-maze, but increased the number of crossings in the open field, an index of locomotor activity. Additionally, alcohol intake increased Fos-immunoreactivity (Fos-ir) in the prefrontal cortex, in the shell region of the nucleus accumbens, in the medial and central amygdala, in the bed nucleus of the stria terminalis, in the septal region, and in the paraventricular and dorsomedial hypothalamus, structures that have been linked to reward and to approach/withdrawal behavior. These observations might be relevant to a better understanding of the behavioral and physiological alterations that follow alcohol consumption.
Collapse
Affiliation(s)
- Tatiana Wscieklica
- Departamento de Biociências, Universidade Federal de São Paulo, 11060-001 Santos, Brazil
| | - Milena de Barros Viana
- Departamento de Biociências, Universidade Federal de São Paulo, 11060-001 Santos, Brazil
| | - Luciana Le Sueur Maluf
- Departamento de Biociências, Universidade Federal de São Paulo, 11060-001 Santos, Brazil
| | | | - Regina Célia Spadari
- Departamento de Biociências, Universidade Federal de São Paulo, 11060-001 Santos, Brazil
| | | |
Collapse
|
148
|
Hwa LS, Holly EN, DeBold JF, Miczek KA. Social stress-escalated intermittent alcohol drinking: modulation by CRF-R1 in the ventral tegmental area and accumbal dopamine in mice. Psychopharmacology (Berl) 2016; 233:681-90. [PMID: 26576941 PMCID: PMC4729595 DOI: 10.1007/s00213-015-4144-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 10/29/2015] [Indexed: 12/22/2022]
Abstract
RATIONALE Excessive alcohol (EtOH) drinking is difficult to model in animals despite the extensive human literature demonstrating that stress increases EtOH consumption. OBJECTIVE The current experiments show escalations in voluntary EtOH drinking caused by a history of social defeat stress and intermittent access to EtOH in C57BL/6J mice compared to non-stressed mice given intermittent EtOH or continuous EtOH. To explore a mechanistic link between stress and drinking, we studied the role of corticotropin-releasing factor type-1 receptors (CRF-R1) in the dopamine-rich ventral tegmental area (VTA). RESULTS Intra-VTA infusions of a CRF-R1 antagonist, CP376395, infused into the VTA dose-dependently and selectively reduced intermittent EtOH intake in stressed and non-stressed mice, but not in mice given continuous EtOH. In contrast, intra-VTA infusions of the CRF-R2 antagonist astressin2B non-specifically suppressed both EtOH and H2O drinking in the stressed group without effects in the non-stressed mice. Using in vivo microdialysis in the nucleus accumbens (NAc) shell, we observed that stressed mice drinking EtOH intermittently had elevated levels of tonic dopamine concentrations compared to non-stressed drinking mice. Also, VTA CP376395 potentiated dopamine output to the NAc only in the stressed group causing further elevations of dopamine post-infusion. CONCLUSIONS These findings illustrate a role for extrahypothalamic CRF-R1 as especially important for stress-escalated EtOH drinking beyond schedule-escalated EtOH drinking. CRF-R1 may be a mechanism for balancing the dysregulation of stress and reward in alcohol use disorders.
Collapse
Affiliation(s)
- Lara S Hwa
- Psychology Department, Tufts University, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Elizabeth N Holly
- Psychology Department, Tufts University, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Joseph F DeBold
- Psychology Department, Tufts University, 530 Boston Avenue, Medford, MA, 02155, USA
| | - Klaus A Miczek
- Psychology Department, Tufts University, 530 Boston Avenue, Medford, MA, 02155, USA.
- Neuroscience Department, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
149
|
Huang CH, Yu YJ, Chang CH, Gean PW. Involvement of metabotropic glutamate receptor 5 in the inhibition of methamphetamine-associated contextual memory after prolonged extinction training. J Neurochem 2016; 137:216-25. [PMID: 26748780 DOI: 10.1111/jnc.13525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/15/2015] [Accepted: 12/29/2015] [Indexed: 12/31/2022]
Abstract
Addiction is thought to be a memory process between perception and environmental cues and addicted patients often relapse when they come into contact with the drug-related context once again. Here, we used a conditioned place preference protocol to seek a more effective extinction methodology of methamphetamine (METH) memory and delineate its underlying mechanism. Conditioning METH for 3 days in mice markedly increased the time spent in the METH-paired compartment. Then the mice were conditioned with saline for 6 days, from day 6 to day 11, a procedure termed extinction training. However, METH memory returned after a priming injection of METH. We prolonged extinction duration from 6 to 10 days and found that this extensive extinction (EE) training prevented priming effect. At the molecular level, we discovered that prolonged extinction training reversed the METH-conditioned place preference-induced increase in surface expression of GluA2 and alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)/NMDA ratio in the basolateral amygdala. In addition, we found that extinction with metabotropic glutamate receptor 5 (mGluR5) activation had similar results to EE: reduced relapse after extinction, decreased synaptic AMPA receptors AMPARs and the AMPA/NMDA ratio. On the contrary, EE with mGluR5 inhibition suppressed the results of EE. These data indicate that EE training-elicited inhibition of METH-primed reinstatement is mediated by the mGluR5. Conditioning mice with methamphetamine place preference (METH CPP) increases surface expression of AMPA receptors (AMPARs) in the basolateral amygdala. We found prolongation of extinction duration from 6 to 10 days prevented priming effect. At the molecular level, we discovered that extensive extinction (EE) reversed the METH CPP-induced increase in surface expression of GluA2 and AMPA/NMDA ratio. In addition, we found that extinction with the metabotropic glutamate receptor 5 (mGluR5) activation had similar results to EE: reduced relapse after extinction, decreased synaptic AMPARs and the AMPA/NMDA ratio. On the contrary, EE with mGluR5 inhibition suppressed the results of EE. These data indicate that EE training-elicited inhibition of METH-primed reinstatement is mediated by mGluR5 (PAM: positive allosteric modulator).
Collapse
Affiliation(s)
- Chien-Hsuan Huang
- Department of Pharmacology, National Cheng-Kung University, Tainan, Taiwan
| | - Yang-Jung Yu
- Department of Pharmacology, National Cheng-Kung University, Tainan, Taiwan
| | - Chih-Hua Chang
- Department of Pharmacology, National Cheng-Kung University, Tainan, Taiwan
| | - Po-Wu Gean
- Department of Pharmacology, National Cheng-Kung University, Tainan, Taiwan
| |
Collapse
|
150
|
Lukkes JL, Norman KJ, Meda S, Andersen SL. Sex differences in the ontogeny of CRF receptors during adolescent development in the dorsal raphe nucleus and ventral tegmental area. Synapse 2016; 70:125-32. [DOI: 10.1002/syn.21882] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/29/2015] [Accepted: 12/15/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Jodi L. Lukkes
- Laboratory for Developmental Neuropharmacology; McLean Hospital; Belmont Massachusetts
- Department of Psychiatry, Harvard Medical School; Belmont Massachusetts
| | - Kevin J. Norman
- Laboratory for Developmental Neuropharmacology; McLean Hospital; Belmont Massachusetts
| | - Shirisha Meda
- Laboratory for Developmental Neuropharmacology; McLean Hospital; Belmont Massachusetts
| | - Susan L. Andersen
- Laboratory for Developmental Neuropharmacology; McLean Hospital; Belmont Massachusetts
- Department of Psychiatry, Harvard Medical School; Belmont Massachusetts
| |
Collapse
|