1
|
Chen D, Wang J, Cao J, Zhu G. cAMP-PKA signaling pathway and anxiety: Where do we go next? Cell Signal 2024; 122:111311. [PMID: 39059755 DOI: 10.1016/j.cellsig.2024.111311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Cyclic adenosine monophosphate (cAMP) is an intracellular second messenger that is derived from the conversion of adenosine triphosphate catalysed by adenylyl cyclase (AC). Protein kinase A (PKA), the main effector of cAMP, is a dimeric protein kinase consisting of two catalytic subunits and two regulatory subunits. When cAMP binds to the regulatory subunits of PKA, it leads to the dissociation and activation of PKA, which allows the catalytic subunit of PKA to phosphorylate target proteins, thereby regulating various physiological functions and metabolic processes in cellular function. Recent researches also implicate the involvement of cAMP-PKA signaling in the pathologenesis of anxiety disorder. However, there are still debates on the prevention and treatment of anxiety disorders from this signaling pathway. To review the function of cAMP-PKA signaling in anxiety disorder, we searched the publications with the keywords including "cAMP", "PKA" and "Anxiety" from Pubmed, Embase, Web of Science and CNKI databases. The results showed that the number of publications on cAMP-PKA pathway in anxiety disorder tended to increase. Bioinformatics results displayed a close association between the cAMP-PKA pathway and the occurrence of anxiety. Mechanistically, cAMP-PKA signaling could influence brain-derived neurotrophic factor and neuropeptide Y and participate in the regulation of anxiety. cAMP-PKA signaling could also oppose the dysfunctions of gamma-aminobutyric acid (GABA), intestinal flora, hypothalamic-pituitary-adrenal axis, neuroinflammation, and signaling proteins (MAPK and AMPK) in anxiety. In addition, chemical agents with the ability to activate cAMP-PKA signaling demonstrated therapy potential against anxiety disorders. This review emphasizes the central roles of cAMP-PKA signaling in anxiety and the targets of the cAMP-PKA pathway would be potential candidates for treatment of anxiety. Nevertheless, more laboratory investigations to improve the therapeutic effect and reduce the adverse effect, and continuous clinical research will warrant the drug development.
Collapse
Affiliation(s)
- Daokang Chen
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jingji Wang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei 230061, China.
| | - Jian Cao
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
2
|
Bhandari A, Seguin A, Rothenfluh A. Synaptic Mechanisms of Ethanol Tolerance and Neuroplasticity: Insights from Invertebrate Models. Int J Mol Sci 2024; 25:6838. [PMID: 38999947 PMCID: PMC11241699 DOI: 10.3390/ijms25136838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
Alcohol tolerance is a neuroadaptive response that leads to a reduction in the effects of alcohol caused by previous exposure. Tolerance plays a critical role in the development of alcohol use disorder (AUD) because it leads to the escalation of drinking and dependence. Understanding the molecular mechanisms underlying alcohol tolerance is therefore important for the development of effective therapeutics and for understanding addiction in general. This review explores the molecular basis of alcohol tolerance in invertebrate models, Drosophila and C. elegans, focusing on synaptic transmission. Both organisms exhibit biphasic responses to ethanol and develop tolerance similar to that of mammals. Furthermore, the availability of several genetic tools makes them a great candidate to study the molecular basis of ethanol response. Studies in invertebrate models show that tolerance involves conserved changes in the neurotransmitter systems, ion channels, and synaptic proteins. These neuroadaptive changes lead to a change in neuronal excitability, most likely to compensate for the enhanced inhibition by ethanol.
Collapse
Affiliation(s)
- Aakriti Bhandari
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandra Seguin
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Adrian Rothenfluh
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
3
|
Fu Y, Li W, Mai Y, Guan J, Ding R, Hou J, Chen B, Cao G, Sun S, Tang Y, Fu R. Association between RMTg Neuropeptide Genes and Negative Effect during Alcohol Withdrawal in Mice. Int J Mol Sci 2024; 25:2933. [PMID: 38474180 DOI: 10.3390/ijms25052933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Alcohol use disorders (AUDs) frequently co-occur with negative mood disorders, such as anxiety and depression, exacerbating relapse through dopaminergic dysfunction. Stress-related neuropeptides play a crucial role in AUD pathophysiology by modulating dopamine (DA) function. The rostromedial tegmental nucleus (RMTg), which inhibits midbrain dopamine neurons and signals aversion, has been shown to increase ethanol consumption and negative emotional states during abstinence. Despite some stress-related neuropeptides acting through the RMTg to affect addiction behaviors, their specific roles in alcohol-induced contexts remain underexplored. This study utilized an intermittent voluntary drinking model in mice to induce negative effect behavior 24 h into ethanol (EtOH) abstinence (post-EtOH). It examined changes in pro-stress (Pnoc, Oxt, Npy) and anti-stress (Crf, Pomc, Avp, Orx, Pdyn) neuropeptide-coding genes and analyzed their correlations with aversive behaviors. We observed that adult male C57BL/6J mice displayed evident anxiety, anhedonia, and depression-like symptoms at 24 h post-EtOH. The laser-capture microdissection technique, coupled with or without retrograde tracing, was used to harvest total ventral tegmental area (VTA)-projecting neurons or the intact RMTg area. The findings revealed that post-EtOH consistently reduced Pnoc and Orx levels while elevating Crf levels in these neuronal populations. Notably, RMTg Pnoc and Npy levels counteracted ethanol consumption and depression severity, while Crf levels were indicative of the mice's anxiety levels. Together, these results underscore the potential role of stress-related neuropeptides in the RMTg in regulating the negative emotions related to AUDs, offering novel insights for future research.
Collapse
Affiliation(s)
- Yixin Fu
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Wenfu Li
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Yunlin Mai
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Junhao Guan
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Ruxuan Ding
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Jiawei Hou
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Bingqing Chen
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Guoxin Cao
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Shizhu Sun
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Ying Tang
- Clinical Skills Training Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Rao Fu
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| |
Collapse
|
4
|
Robinson SL, Bendrath SC, Yates EM, Thiele TE. Basolateral amygdala neuropeptide Y system modulates binge ethanol consumption. Neuropsychopharmacology 2024; 49:690-698. [PMID: 37758802 PMCID: PMC10876546 DOI: 10.1038/s41386-023-01742-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/22/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023]
Abstract
Neuropeptide Y (NPY) signaling regulation of corticolimbic communication is known to modulate binge-like ethanol consumption in rodents. In this work we sought to assess the impact of intra-BLA NPY system modulation on binge-like ethanol intake and to assess the role of the NPY1R+ projection from the BLA to the mPFC in this behavior. We used "drinking-in-the-dark" (DID) procedures in C57BL6J mice to address these questions. First, the impact of intra-BLA administration of NPY on binge-like ethanol intake was assessed. Next, the impact of repeated cycles of DID intake on NPY1R expression in the BLA was assessed with use of immunohistochemistry (IHC). Finally, chemogenetic inhibition of BLA→mPFC NPY1R+ projections was assessed to determine if limbic communication with the mPFC was specifically involved in binge-like ethanol intake. Importantly, as both the BLA and NPY system are sexually dimorphic, both sexes were assessed in these studies. Intra-BLA NPY dose-dependently decreased binge-like ethanol intake in males only. Repeated DID reduced NPY1R expression in the BLA of both sexes. Silencing of BLA→mPFC NPY1R+ neurons significantly reduced binge-like ethanol intake in both sexes in a dose-dependent manner. We provide novel evidence that (1) intra-BLA NPY reduces binge-like ethanol intake in males; (2) binge-like ethanol intake reduces NPY1R levels in the BLA; and (3) chemogenetic inhibition of BLA→mPFC NPY1R+ neurons blunts binge-like drinking in male and female mice. These observations provide the first direct evidence that NPY signaling in the BLA, and specifically BLA communication with the mPFC, modulates binge-like ethanol consumption.
Collapse
Affiliation(s)
- Stacey L Robinson
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC, 27599-3270, USA
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC, 27599-7178, USA
| | - Sophie C Bendrath
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC, 27599-3270, USA
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC, 27599-7178, USA
| | - Elizabeth M Yates
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC, 27599-3270, USA
| | - Todd E Thiele
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC, 27599-3270, USA.
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC, 27599-7178, USA.
| |
Collapse
|
5
|
Bale R, Doshi G. Cross talk about the role of Neuropeptide Y in CNS disorders and diseases. Neuropeptides 2023; 102:102388. [PMID: 37918268 DOI: 10.1016/j.npep.2023.102388] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
A peptide composed of a 36 amino acid called Neuropeptide Y (NPY) is employed in a variety of physiological processes to manage and treat conditions affecting the endocrine, circulatory, respiratory, digestive, and neurological systems. NPY naturally binds to G-protein coupled receptors, activating the Y-receptors (Y1-Y5 and y6). The findings on numerous therapeutic applications of NPY for CNS disease are presented in this review by the authors. New targets for treating diseases will be revealed by medication combinations that target NPY and its receptors. This review is mainly focused on disorders such as anxiety, Alzheimer's disease, Parkinson's disease, Huntington's disease, Machado Joseph disease, multiple sclerosis, schizophrenia, depression, migraine, alcohol use disorder, and substance use disorder. The findings from the preclinical studies and clinical studies covered in this article may help create efficient therapeutic plans to treat neurological conditions on the one hand and psychiatric disorders on the other. They may also open the door to the creation of novel NPY receptor ligands as medications to treat these conditions.
Collapse
Affiliation(s)
- Rajeshwari Bale
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai 400056, India.
| |
Collapse
|
6
|
Brigande AM, Darwich JG, Currie PJ. Mesolimbic exendin-4 attenuates reward salience evoked by neuropeptide Y and ghrelin. Behav Brain Res 2023; 440:114249. [PMID: 36496077 DOI: 10.1016/j.bbr.2022.114249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
In the present study, we investigated the effects of the glucagon-like peptide-1 (GLP-1) agonist exendin-4 (Ex-4) on the stimulatory action of neuropeptide Y (NPY) and ghrelin. These effects were examined in relation to operant responding for palatable food or voluntary ethanol intake in a two-bottle limited access paradigm. Male Sprague Dawley rats, each with ventral tegmental area (VTA) unilateral guide cannulae, were used. Ex-4 was paired with either NPY, ghrelin, or combined NPY and ghrelin treatment. Our results indicated that while NPY and ghrelin reliably stimulated operant responding for sucrose pellets and increased ethanol intake, Ex-4 suppressed intake and, most importantly, significantly reduced the effects of NPY and ghrelin. Overall, this work provides compelling evidence that VTA GLP-1, NPY, and ghrelin systems interact within the brain to modulate reward salience.
Collapse
Affiliation(s)
- Alev M Brigande
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Janet Guss Darwich
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA
| | - Paul J Currie
- Department of Psychology, Reed College, 3203 SE Woodstock Blvd., Portland, OR 97202, USA.
| |
Collapse
|
7
|
Companion MA, Gonzalez DA, Robinson SL, Herman MA, Thiele TE. Lateral habenula-projecting central amygdala circuits expressing GABA and NPY Y1 receptor modulate binge-like ethanol intake in mice. ADDICTION NEUROSCIENCE 2022; 3:100019. [PMID: 36059430 PMCID: PMC9435303 DOI: 10.1016/j.addicn.2022.100019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The central nucleus of the amygdala (CeA) is a critical brain region in the integration of emotional behaviors and is one of the major output areas of the amygdaloid complex. The CeA is composed of GABAergic interneurons and projection neurons which co-express a range of peptides including neuropeptide Y (NPY). Importantly, GABA and NPY signaling, via the NPY Y1 receptor (Y1R), in the CeA modulate binge-like ethanol intake in rodents and these systems undergo neuroplastic alterations following a history of ethanol consumption. Here we assessed the roles of GABAergic and Y1R+ circuits arising from the CeA and innervating the lateral habenula (LHb), a brain region that modulates the aversive properties of ethanol, in modulating binge-like ethanol intake in mice using "drinking in the dark" (DID) procedures. Using an anterograde cre-inducible reporter virus we established the CeA → LHb circuit in male and female vgat-ires-cre and NPY1r-cre mice. Next, we found that chemogenetic silencing of both the GABAergic or Y1R+ CeA → LHb circuit significantly blunted binge-like intake of a 20% ethanol solution but this same procedure failed to alter the consumption of a 3% sucrose solution. Finally, one, 4-day cycle of DID failed to alter basal or effects of ethanol or NPY on inhibitory transmission in Y1R+ CeA → LHb neurons. The present results suggest that blunting GABAergic tone in LHb-projecting CeA neurons may represent a new approach to preventing the development of AUDs. Drugs that target NPY Y1Rs are potential attractive targets.
Collapse
Affiliation(s)
- Michel A Companion
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Davie Hall, CB#3270, Chapel Hill, NC 27599-3270, United States.,The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Davie Hall, CB#7178, Chapel Hill, NC 27599-7178, United States
| | - David A Gonzalez
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Davie Hall, CB#3270, Chapel Hill, NC 27599-3270, United States
| | - Stacey L Robinson
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Davie Hall, CB#3270, Chapel Hill, NC 27599-3270, United States.,The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Davie Hall, CB#7178, Chapel Hill, NC 27599-7178, United States
| | - Melissa A Herman
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Davie Hall, CB#3270, Chapel Hill, NC 27599-3270, United States.,The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Davie Hall, CB#7178, Chapel Hill, NC 27599-7178, United States.,Department of Pharmacology, University of North Carolina at Chapel Hill, Davie Hall, CB#7365, Chapel Hill, NC 27599-7365, United States
| | - Todd E Thiele
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Davie Hall, CB#3270, Chapel Hill, NC 27599-3270, United States.,The Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Davie Hall, CB#7178, Chapel Hill, NC 27599-7178, United States
| |
Collapse
|
8
|
Grinevich VP, Krupitsky EM, Gainetdinov RR, Budygin EA. Linking Ethanol-Addictive Behaviors With Brain Catecholamines: Release Pattern Matters. Front Behav Neurosci 2022; 15:795030. [PMID: 34975429 PMCID: PMC8716449 DOI: 10.3389/fnbeh.2021.795030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/26/2021] [Indexed: 12/30/2022] Open
Abstract
Using a variety of animal models that simulate key features of the alcohol use disorder (AUD), remarkable progress has been made in identifying neurochemical targets that may contribute to the development of alcohol addiction. In this search, the dopamine (DA) and norepinephrine (NE) systems have been long thought to play a leading role in comparison with other brain systems. However, just recent development and application of optogenetic approaches into the alcohol research field provided opportunity to identify neuronal circuits and specific patterns of neurotransmission that govern the key components of ethanol-addictive behaviors. This critical review summarizes earlier findings, which initially disclosed catecholamine substrates of ethanol actions in the brain and shows how the latest methodologies help us to reveal the significance of DA and NE release changes. Specifically, we focused on recent optogenetic investigations aimed to reveal cause-effect relationships between ethanol-drinking (seeking and taking) behaviors and catecholamine dynamics in distinct brain pathways. These studies gain the knowledge that is needed for the better understanding addiction mechanisms and, therefore, for development of more effective AUD treatments. Based on the reviewed findings, new messages for researches were indicated, which may have broad applications beyond the field of alcohol addiction.
Collapse
Affiliation(s)
- Vladimir P Grinevich
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia
| | - Evgeny M Krupitsky
- V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, St. Petersburg, Russia.,Laboratory of Clinical Psychopharmacology of Addictions, St.-Petersburg First Pavlov State Medical University, St. Petersburg, Russia
| | - Raul R Gainetdinov
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia.,Institute of Translational Biomedicine and St. Petersburg State University Hospital, St. Petersburg State University, St. Petersburg, Russia
| | - Evgeny A Budygin
- Department of Neurobiology, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
9
|
Denny RR, Connelly KL, Ghilotti MG, Meissler JJ, Yu D, Eisenstein TK, Unterwald EM. Artificial Intelligence Identified Resilient and Vulnerable Female Rats After Traumatic Stress and Ethanol Exposure: Investigation of Neuropeptide Y Pathway Regulation. Front Neurosci 2021; 15:772946. [PMID: 34975380 PMCID: PMC8716605 DOI: 10.3389/fnins.2021.772946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is initiated by traumatic-stress exposure and manifests into a collection of symptoms including increased anxiety, sleep disturbances, enhanced response to triggers, and increased sympathetic nervous system arousal. PTSD is highly co-occurring with alcohol use disorder. Only some individuals experiencing traumatic stress develop PTSD and a subset of individuals with PTSD develop co-occurring alcohol use disorder. To investigate the basis of these individual responses to traumatic stress, single prolonged stress (SPS) a rodent model of traumatic stress was applied to young adult female rats. Individual responses to SPS were characterized by measuring anxiety-like behaviors with open field and elevated plus maze tests. Rats were then allowed to drink ethanol under an intermittent two bottle choice procedure for 8 weeks, and ethanol consumption was measured. An artificial intelligence algorithm was built to predict resilient and vulnerable individuals based on data from anxiety testing and ethanol consumption. This model was implemented in a second cohort of rats that underwent SPS without ethanol drinking to identify resilient and vulnerable individuals for further study. Analysis of neuropeptide Y (NPY) levels and expression of its receptors Y1R and Y2R mRNA in the central nucleus of the amygdala (CeA), basolateral amygdala (BLA), and bed nucleus stria terminalis (BNST) were performed. Results demonstrate that resilient rats had higher expression of Y2R mRNA in the CeA compared with vulnerable and control rats and had higher levels of NPY protein in the BNST compared to controls. The results of the study show that an artificial intelligence algorithm can identify individual differences in response to traumatic stress which can be used to predict subsequent ethanol drinking, and the NPY pathway is differentially altered following traumatic stress exposure in resilient and vulnerable populations. Understanding neurochemical alterations following traumatic-stress exposure is critical in developing prevention strategies for the vulnerable phenotype and will help further development of novel therapeutic approaches for individuals suffering from PTSD and at risk for alcohol use disorder.
Collapse
Affiliation(s)
- Ray R. Denny
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Krista L. Connelly
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Marco G. Ghilotti
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Joseph J. Meissler
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Daohai Yu
- Center for Biostatistics and Epidemiology, Department of Biomedical Education and Data Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Toby K. Eisenstein
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Ellen M. Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,*Correspondence: Ellen M. Unterwald,
| |
Collapse
|
10
|
Bhat US, Shahi N, Surendran S, Babu K. Neuropeptides and Behaviors: How Small Peptides Regulate Nervous System Function and Behavioral Outputs. Front Mol Neurosci 2021; 14:786471. [PMID: 34924955 PMCID: PMC8674661 DOI: 10.3389/fnmol.2021.786471] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
One of the reasons that most multicellular animals survive and thrive is because of the adaptable and plastic nature of their nervous systems. For an organism to survive, it is essential for the animal to respond and adapt to environmental changes. This is achieved by sensing external cues and translating them into behaviors through changes in synaptic activity. The nervous system plays a crucial role in constantly evaluating environmental cues and allowing for behavioral plasticity in the organism. Multiple neurotransmitters and neuropeptides have been implicated as key players for integrating sensory information to produce the desired output. Because of its simple nervous system and well-established neuronal connectome, C. elegans acts as an excellent model to understand the mechanisms underlying behavioral plasticity. Here, we critically review how neuropeptides modulate a wide range of behaviors by allowing for changes in neuronal and synaptic signaling. This review will have a specific focus on feeding, mating, sleep, addiction, learning and locomotory behaviors in C. elegans. With a view to understand evolutionary relationships, we explore the functions and associated pathophysiology of C. elegans neuropeptides that are conserved across different phyla. Further, we discuss the mechanisms of neuropeptidergic signaling and how these signals are regulated in different behaviors. Finally, we attempt to provide insight into developing potential therapeutics for neuropeptide-related disorders.
Collapse
Affiliation(s)
- Umer Saleem Bhat
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Navneet Shahi
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Siju Surendran
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Kavita Babu
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
11
|
Chen G, Ghazal M, Rahman S, Lutfy K. The impact of adolescent nicotine exposure on alcohol use during adulthood: The role of neuropeptides. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:53-93. [PMID: 34801174 DOI: 10.1016/bs.irn.2021.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Nicotine and alcohol abuse and co-dependence represent major public health crises. Indeed, previous research has shown that the prevalence of alcoholism is higher in smokers than in non-smokers. Adolescence is a susceptible period of life for the initiation of nicotine and alcohol use and the development of nicotine-alcohol codependence. However, there is a limited number of pharmacotherapeutic agents to treat addiction to nicotine or alcohol alone. Notably, there is no effective medication to treat this comorbid disorder. This chapter aims to review the early nicotine use and its impact on subsequent alcohol abuse during adolescence and adulthood as well as the role of neuropeptides in this comorbid disorder. The preclinical and clinical findings discussed in this chapter will advance our understanding of this comorbid disorder's neurobiology and lay a foundation for developing novel pharmacotherapies to treat nicotine and alcohol codependence.
Collapse
Affiliation(s)
- G Chen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States; Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - M Ghazal
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States
| | - K Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States.
| |
Collapse
|
12
|
Tanaka M, Yamada S, Watanabe Y. The Role of Neuropeptide Y in the Nucleus Accumbens. Int J Mol Sci 2021; 22:ijms22147287. [PMID: 34298907 PMCID: PMC8307209 DOI: 10.3390/ijms22147287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/22/2022] Open
Abstract
Neuropeptide Y (NPY), an abundant peptide in the central nervous system, is expressed in neurons of various regions throughout the brain. The physiological and behavioral effects of NPY are mainly mediated through Y1, Y2, and Y5 receptor subtypes, which are expressed in regions regulating food intake, fear and anxiety, learning and memory, depression, and posttraumatic stress. In particular, the nucleus accumbens (NAc) has one of the highest NPY concentrations in the brain. In this review, we summarize the role of NPY in the NAc. NPY is expressed principally in medium-sized aspiny neurons, and numerous NPY immunoreactive fibers are observed in the NAc. Alterations in NPY expression under certain conditions through intra-NAc injections of NPY or receptor agonists/antagonists revealed NPY to be involved in the characteristic functions of the NAc, such as alcohol intake and drug addiction. In addition, control of mesolimbic dopaminergic release via NPY receptors may take part in these functions. NPY in the NAc also participates in fat intake and emotional behavior. Accumbal NPY neurons and fibers may exert physiological and pathophysiological actions partly through neuroendocrine mechanisms and the autonomic nervous system.
Collapse
Affiliation(s)
- Masaki Tanaka
- Department of Anatomy, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan;
- Correspondence: ; Tel.: +81-75-251-5300
| | - Shunji Yamada
- Department of Anatomy, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan;
| | - Yoshihisa Watanabe
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan;
| |
Collapse
|
13
|
Drosophila Corazonin Neurons as a Hub for Regulating Growth, Stress Responses, Ethanol-Related Behaviors, Copulation Persistence and Sexually Dimorphic Reward Pathways. J Dev Biol 2021; 9:jdb9030026. [PMID: 34287347 PMCID: PMC8293205 DOI: 10.3390/jdb9030026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
The neuronal mechanisms by which complex behaviors are coordinated and timed often involve neuropeptidergic regulation of stress and reward pathways. Recent studies of the neuropeptide Corazonin (Crz), a homolog of the mammalian Gonadotrophin Releasing Hormone (GnRH), have suggested its crucial role in the regulation of growth, internal states and behavioral decision making. We focus this review on Crz neurons with the goal to (1) highlight the diverse roles of Crz neuron function, including mechanisms that may be independent of the Crz peptide, (2) emphasize current gaps in knowledge about Crz neuron functions, and (3) propose exciting ideas of novel research directions involving the use of Crz neurons. We describe the different developmental fates of distinct subsets of Crz neurons, including recent findings elucidating the molecular regulation of apoptosis. Crz regulates systemic growth, food intake, stress responses and homeostasis by interacting with the short Neuropeptide F (sNPF) and the steroid hormone ecdysone. Additionally, activation of Crz neurons is shown to be pleasurable by interacting with the Neuropeptide F (NPF) and regulates reward processes such as ejaculation and ethanol-related behaviors in a sexually dimorphic manner. Crz neurons are proposed to be a motivational switch regulating copulation duration using a CaMKII-dependent mechanism described as the first neuronal interval timer lasting longer than a few seconds. Lastly, we propose ideas to use Crz neuron-induced ejaculation to study the effects of fictive mating and sex addiction in flies, as well as to elucidate dimorphic molecular mechanisms underlying reward behaviors and feeding disorders.
Collapse
|
14
|
Dulman RS, Zhang H, Banerjee R, Krishnan HR, Dong B, Hungund BL, Vinod KY, Pandey SC. CB1 receptor neutral antagonist treatment epigenetically increases neuropeptide Y expression and decreases alcohol drinking. Neuropharmacology 2021; 195:108623. [PMID: 34048869 DOI: 10.1016/j.neuropharm.2021.108623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/22/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Alcohol consumption is mediated by several important neuromodulatory systems, including the endocannabinoid and neuropeptide Y (NPY) systems in the limbic brain circuitry. However, molecular mechanisms through which cannabinoid-1 (CB1) receptors regulate alcohol consumption are still unclear. Here, we investigated the role of the CB1 receptor-mediated downstream regulation of NPY via epigenetic mechanisms in the amygdala. Alcohol drinking behavior was measured in adult male C57BL/6J mice treated with a CB1 receptor neutral antagonist AM4113 using a two-bottle choice paradigm while anxiety-like behavior was assessed in the light-dark box (LDB) test. The CB1 receptor-mediated changes in the protein levels of phosphorylated cAMP-responsive element binding protein (pCREB), CREB binding protein (CBP), H3K9ac, H3K14ac and NPY, and the mRNA levels of Creb1, Cbp, and Npy were measured in amygdaloid brain structures. Npy-specific changes in the levels of acetylated histone (H3K9/14ac) and CBP in the amygdala were also measured. We found that the pharmacological blockade of CB1 receptors with AM4113 reduced alcohol consumption and, in an ethanol-naïve cohort, reduced anxiety-like behavior in the LDB test. Treatment with AM4113 also increased the mRNA levels of Creb1 and Cbp in the amygdala as well as the protein levels of pCREB, CBP, H3K9ac and H3K14ac in the central and medial nucleus of amygdala, but not in the basolateral amygdala. Additionally, AM4113 treatment increased occupancy of CBP and H3K9/14ac at the Npy gene promoter, leading to an increase in both mRNA and protein levels of NPY in the amygdala. These novel findings suggest that CB1 receptor-mediated CREB signaling plays an important role in the modulation of NPY function through an epigenetic mechanism and further support the potential use of CB1 receptor neutral antagonists for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Russell S Dulman
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA
| | - Ritabrata Banerjee
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Harish R Krishnan
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA
| | - Bin Dong
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Basalingappa L Hungund
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA; New York State Psychiatric Institute, New York, NY, 10032, USA
| | - K Yaragudri Vinod
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA; Emotional Brain Institute, Orangeburg, NY, 10962, USA; Department of Child and Adolescent Psychiatry, New York School of Medicine, New York, NY, 10016, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
15
|
Jensen BE, Townsley KG, Grigsby KB, Metten P, Chand M, Uzoekwe M, Tran A, Firsick E, LeBlanc K, Crabbe JC, Ozburn AR. Ethanol-Related Behaviors in Mouse Lines Selectively Bred for Drinking to Intoxication. Brain Sci 2021; 11:189. [PMID: 33557285 PMCID: PMC7915226 DOI: 10.3390/brainsci11020189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 11/18/2022] Open
Abstract
Alcohol use disorder (AUD) is a devastating psychiatric disorder that has significant wide-reaching effects on individuals and society. Selectively bred mouse lines are an effective means of exploring the genetic and neuronal mechanisms underlying AUD and such studies are translationally important for identifying treatment options. Here, we report on behavioral characterization of two replicate lines of mice that drink to intoxication, the High Drinking in the Dark (HDID)-1 and -2 mice, which have been selectively bred (20+ generations) for the primary phenotype of reaching high blood alcohol levels (BALs) during the drinking in the dark (DID) task, a binge-like drinking assay. Along with their genetically heterogenous progenitor line, Hs/Npt, we tested these mice on: DID and drinking in the light (DIL); temporal drinking patterns; ethanol sensitivity, through loss of righting reflex (LORR); and operant self-administration, including fixed ratio (FR1), fixed ratio 3:1 (FR3), extinction/reinstatement, and progressive ratio (PR). All mice consumed more ethanol during the dark than the light and both HDID lines consumed more ethanol than Hs/Npt during DIL and DID. In the dark, we found that the HDID lines achieved high blood alcohol levels early into a drinking session, suggesting that they exhibit front loading like drinking behavior in the absence of the chronicity usually required for such behavior. Surprisingly, HDID-1 (female and male) and HDID-2 (male) mice were more sensitive to the intoxicating effects of ethanol during the dark (as determined by LORR), while Hs/Npt (female and male) and HDID-2 (female) mice appeared less sensitive. We observed lower HDID-1 ethanol intake compared to either HDID-2 or Hs/Npt during operant ethanol self-administration. There were no genotype differences for either progressive ratio responding, or cue-induced ethanol reinstatement, though the latter is complicated by a lack of extinguished responding behavior. Taken together, these findings suggest that genes affecting one AUD-related behavior do not necessarily affect other AUD-related behaviors. Moreover, these findings highlight that alcohol-related behaviors can also differ between lines selectively bred for the same phenotype, and even between sexes within those same line.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Angela R. Ozburn
- Department of Behavioral Neuroscience, Oregon Health & Science University, and VA Portland Health Care System, Portland, OR 97239, USA; (B.E.J.); (K.G.T.); (K.B.G.); (P.M.); (M.C.); (M.U.); (A.T.); (E.F.); (K.L.); (J.C.C.)
| |
Collapse
|
16
|
Chen B, Yadav M, Mulkalwar M, Saikrishna L, Verma H, Ye W, Bhaskar LVKS. Meta-Analysis on the Association of Neuropeptide Y rs16139 Variant With the Risk of Alcoholism. Front Psychiatry 2021; 12:737440. [PMID: 34777047 PMCID: PMC8583313 DOI: 10.3389/fpsyt.2021.737440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: The neuropeptide-Y (NPY) is involved in the development of alcoholism through NPY receptors. A T>C mutation causes substitution of leucine to proline at codon 7 (L7P; rs16139) in the signal peptide of neuropeptide Y is known to cause a 42% increase in plasma NPY levels. Studies that analyzed the association between NPY rs16139 and alcoholism risk did not demonstrate conclusive evidence for this relationship. The present study aims to evaluate the association between NPY gene rs16139 variant and alcohol dependence. Method: An electronic search of databases including PubMed and Google Scholar was performed to retrieve studies investigating the association between NPY rs16139 and alcoholism. The pooled odds ratio (OR) with 95% confidence interval (CI) was calculated in allelic and dominant genetic models. Sensitivity analyses and publication bias were assessed in our meta-analysis. The meta-analysis was conducted using the MetaGenyo web tool. Result: Significant heterogeneity was observed across studies (p < 0.001). Our results have shown that there is no significant association between NPY rs16139 variant and the risk of alcoholism in allelic (OR = 0.98, 95% CI 0.70-1.38, p = 0.921) and dominant models (OR = 0.98, 95% CI 0.69-1.40, p = 0.919). Begg's funnel plot and Egger's test have not shown publication bias (p = 0.332). Conclusion: To the best of our knowledge, this is the first meta-analysis that evaluates the relationship between the NPY rs16139 polymorphism and the risk of alcoholism. Our large-scale meta-analysis suggests that NPY rs16139 polymorphism is not associated with alcoholism. However, further studies are needed to increase our understanding of the relationship between NPY variants in alcoholism.
Collapse
Affiliation(s)
- Biqing Chen
- Department of Sports Operation and Management, Jinhua Polytechnic, Jinhua, China
| | - Manish Yadav
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Madhubala Mulkalwar
- Department of Pathology, Shri Shankaracharya Institute of Medical Sciences (SSIMS), Bhilai, India
| | | | - Henu Verma
- Department of Immunopathology, Institute of Lungs Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum, Munich, Germany
| | - Weibing Ye
- Exercise and Metabolism Research Center, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - L V K S Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
17
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
18
|
Brockway DF, Crowley NA. Turning the 'Tides on Neuropsychiatric Diseases: The Role of Peptides in the Prefrontal Cortex. Front Behav Neurosci 2020; 14:588400. [PMID: 33192369 PMCID: PMC7606924 DOI: 10.3389/fnbeh.2020.588400] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
Recent advancements in technology have enabled researchers to probe the brain with the greater region, cell, and receptor specificity. These developments have allowed for a more thorough understanding of how regulation of the neurophysiology within a region is essential for maintaining healthy brain function. Stress has been shown to alter the prefrontal cortex (PFC) functioning, and evidence links functional impairments in PFC brain activity with neuropsychiatric disorders. Moreover, a growing body of literature highlights the importance of neuropeptides in the PFC to modulate neural signaling and to influence behavior. The converging evidence outlined in this review indicates that neuropeptides in the PFC are specifically impacted by stress, and are found to be dysregulated in numerous stress-related neuropsychiatric disorders including substance use disorder, major depressive disorder (MDD), posttraumatic stress disorder, and schizophrenia. This review explores how neuropeptides in the PFC function to regulate the neural activity, and how genetic and environmental factors, such as stress, lead to dysregulation in neuropeptide systems, which may ultimately contribute to the pathology of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Dakota F Brockway
- Neuroscience Curriculum, Pennsylvania State University, University Park, PA, United States
| | - Nicole A Crowley
- Neuroscience Curriculum, Pennsylvania State University, University Park, PA, United States.,The Department of Biology, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
19
|
Sex differences in behavioral and metabolic effects of gene inactivation: The neuropeptide Y and Y receptors in the brain. Neurosci Biobehav Rev 2020; 119:333-347. [PMID: 33045245 DOI: 10.1016/j.neubiorev.2020.09.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Brain and gonadal hormones interplay controls metabolic and behavioral functions in a sex-related manner. However, most translational neuroscience research related to animal models of endocrine and psychiatric disorders are often carried out in male animals only. The Neuropeptide Y (NPY) system shows sex-dependent differences and is sensitive to gonadal steroids. Based on published data from our and other laboratories, in this review we will discuss the sex related differences of NPY action on energy balance, bone homeostasis and behavior in rodents with the genetic manipulation of genes encoding NPY and its Y1, Y2 and Y5 cognate receptors. Comparative analyses of the phenotype of transgenic and knockout NPY and Y receptor rodents unravels sex dependent differences in the functions of this neurotransmission system, potentially helping to develop therapeutics for a variety of sex-related disorders including metabolic syndrome, osteoporosis and ethanol addiction.
Collapse
|
20
|
Lathen DR, Merrill CB, Rothenfluh A. Flying Together: Drosophila as a Tool to Understand the Genetics of Human Alcoholism. Int J Mol Sci 2020; 21:E6649. [PMID: 32932795 PMCID: PMC7555299 DOI: 10.3390/ijms21186649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Alcohol use disorder (AUD) exacts an immense toll on individuals, families, and society. Genetic factors determine up to 60% of an individual's risk of developing problematic alcohol habits. Effective AUD prevention and treatment requires knowledge of the genes that predispose people to alcoholism, play a role in alcohol responses, and/or contribute to the development of addiction. As a highly tractable and translatable genetic and behavioral model organism, Drosophila melanogaster has proven valuable to uncover important genes and mechanistic pathways that have obvious orthologs in humans and that help explain the complexities of addiction. Vinegar flies exhibit remarkably strong face and mechanistic validity as a model for AUDs, permitting many advancements in the quest to understand human genetic involvement in this disease. These advancements occur via approaches that essentially fall into one of two categories: (1) discovering candidate genes via human genome-wide association studies (GWAS), transcriptomics on post-mortem tissue from AUD patients, or relevant physiological connections, then using reverse genetics in flies to validate candidate genes' roles and investigate their molecular function in the context of alcohol. (2) Utilizing flies to discover candidate genes through unbiased screens, GWAS, quantitative trait locus analyses, transcriptomics, or single-gene studies, then validating their translational role in human genetic surveys. In this review, we highlight the utility of Drosophila as a model for alcoholism by surveying recent advances in our understanding of human AUDs that resulted from these various approaches. We summarize the genes that are conserved in alcohol-related function between humans and flies. We also provide insight into some advantages and limitations of these approaches. Overall, this review demonstrates how Drosophila have and can be used to answer important genetic questions about alcohol addiction.
Collapse
Affiliation(s)
- Daniel R. Lathen
- Department of Psychiatry and Neuroscience Ph.D. Program, University of Utah, Salt Lake City, UT 84108, USA;
| | - Collin B. Merrill
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA;
| | - Adrian Rothenfluh
- Department of Psychiatry and Neuroscience Ph.D. Program, University of Utah, Salt Lake City, UT 84108, USA;
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
21
|
Vena AA, Zandy SL, Cofresí RU, Gonzales RA. Behavioral, neurobiological, and neurochemical mechanisms of ethanol self-administration: A translational review. Pharmacol Ther 2020; 212:107573. [PMID: 32437827 PMCID: PMC7580704 DOI: 10.1016/j.pharmthera.2020.107573] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2020] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder has multiple characteristics including excessive ethanol consumption, impaired control over drinking behaviors, craving and withdrawal symptoms, compulsive seeking behaviors, and is considered a chronic condition. Relapse is common. Determining the neurobiological targets of ethanol and the adaptations induced by chronic ethanol exposure is critical to understanding the clinical manifestation of alcohol use disorders, the mechanisms underlying the various features of the disorder, and for informing medication development. In the present review, we discuss ethanol's interactions with a variety of neurotransmitter systems, summarizing findings from preclinical and translational studies to highlight recent progress in the field. We then describe animal models of ethanol self-administration, emphasizing the value, limitations, and validity of commonly used models. Lastly, we summarize the behavioral changes induced by chronic ethanol self-administration, with an emphasis on cue-elicited behavior, the role of ethanol-related memories, and the emergence of habitual ethanol seeking behavior.
Collapse
Affiliation(s)
- Ashley A Vena
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, United States of America
| | | | - Roberto U Cofresí
- Psychological Sciences, University of Missouri, United States of America
| | - Rueben A Gonzales
- Division of Pharmacology and Toxicology, College of Pharmacy and Institute for Neuroscience, The University of Texas at Austin, United States of America.
| |
Collapse
|
22
|
Corder KM, Li Q, Cortes MA, Bartley AF, Davis TR, Dobrunz LE. Overexpression of neuropeptide Y decreases responsiveness to neuropeptide Y. Neuropeptides 2020; 79:101979. [PMID: 31708112 PMCID: PMC6960342 DOI: 10.1016/j.npep.2019.101979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Abstract
Neuropeptide Y (NPY) is an endogenous neuropeptide that is abundantly expressed in the central nervous system. NPY is involved in various neurological processes and neuropsychiatric disorders, including fear learning and anxiety disorders. Reduced levels of NPY are reported in Post-Traumatic Stress Disorder (PTSD) patients, and NPY has been proposed as a potential therapeutic target for PTSD. It is therefore important to understand the effects of chronic enhancement of NPY on anxiety and fear learning. Previous studies have shown that acute elevation of NPY reduces anxiety, fear learning and locomotor activity. Models of chronic NPY overexpression have produced mixed results, possibly caused by ectopic NPY expression. NPY is expressed primarily by a subset of GABAergic interneurons, providing specific spatiotemporal release patterns. Administration of exogenous NPY throughout the brain, or overexpression in cells that do not normally release NPY, can have detrimental side effects, including memory impairment. In order to determine the effects of boosting NPY only in the cells that normally release it, we utilized a transgenic mouse line that overexpresses NPY only in NPY+ cells. We tested for effects on anxiety related behaviors in adolescent mice, an age with high incidence of anxiety disorders in humans. Surprisingly, we did not observe the expected reduction in anxiety-like behavior in NPY overexpression mice. There was no change in fear learning behavior, although there was a deficit in nest building. The effect of exogenous NPY on synaptic transmission in acute hippocampal slices was also diminished, indicating that the function of NPY receptors is impaired. Reduced NPY receptor function could contribute to the unexpected behavioral outcomes. We conclude that overexpression of NPY, even in cells that normally express it, can lead to reduced responsiveness of NPY receptors, potentially affecting the ability of NPY to function as a long-term therapeutic.
Collapse
Affiliation(s)
- Katelynn M Corder
- University of Alabama at Birmingham, Department of Neurobiology, 1825 University Blvd, SHEL 971, Birmingham, AL 35294, United States of America; University of Alabama at Birmingham, Department of Biology, 1670 University Blvd., VH G133B, Birmingham, AL 35233, United States of America
| | - Qin Li
- University of Alabama at Birmingham, Department of Neurobiology, 1825 University Blvd, SHEL 971, Birmingham, AL 35294, United States of America
| | - Mariana A Cortes
- University of Alabama at Birmingham, Department of Neurobiology, 1825 University Blvd, SHEL 971, Birmingham, AL 35294, United States of America
| | - Aundrea F Bartley
- University of Alabama at Birmingham, Department of Neurobiology, 1825 University Blvd, SHEL 971, Birmingham, AL 35294, United States of America
| | - Taylor R Davis
- University of Alabama at Birmingham, Department of Neurobiology, 1825 University Blvd, SHEL 971, Birmingham, AL 35294, United States of America
| | - Lynn E Dobrunz
- University of Alabama at Birmingham, Department of Neurobiology, 1825 University Blvd, SHEL 971, Birmingham, AL 35294, United States of America.
| |
Collapse
|
23
|
Bozler J, Kacsoh BZ, Bosco G. Transgeneratonal inheritance of ethanol preference is caused by maternal NPF repression. eLife 2019; 8:45391. [PMID: 31287057 PMCID: PMC6615861 DOI: 10.7554/elife.45391] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022] Open
Abstract
Rapid or even anticipatory adaptation to environmental conditions can provide a decisive fitness advantage to an organism. The memory of recurring conditions could also benefit future generations; however, neuronally-encoded behavior isn’t thought to be inherited across generations. We tested the possibility that environmentally triggered modifications could allow ‘memory’ of parental experiences to be inherited. In Drosophila melanogaster, exposure to predatory wasps leads to inheritance of a predisposition for ethanol-rich food for five generations. Inhibition of Neuropeptide-F (NPF) activates germline caspases required for transgenerational ethanol preference. Further, inheritance of low NPF expression in specific regions of F1 brains is required for the transmission of this food preference: a maternally derived NPF locus is necessary for this phenomenon, implicating a maternal epigenetic mechanism of NPF-repression. Given the conserved signaling functions of NPF and its mammalian NPY homolog in drug and alcohol disorders, these observations raise the intriguing possibility of NPY-related transgenerational effects in humans.
Collapse
Affiliation(s)
- Julianna Bozler
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, United States
| | - Balint Z Kacsoh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, United States
| | - Giovanni Bosco
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, United States
| |
Collapse
|
24
|
Franke M, Conzelmann A, Grünblatt E, Werling AM, Spieles H, Wewetzer C, Warnke A, Romanos M, Walitza S, Renner TJ. No Association of Variants of the NPY-System With Obsessive-Compulsive Disorder in Children and Adolescents. Front Mol Neurosci 2019; 12:112. [PMID: 31133798 PMCID: PMC6511743 DOI: 10.3389/fnmol.2019.00112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 04/17/2019] [Indexed: 01/08/2023] Open
Abstract
Obsessive-compulsive disorder (OCD) causes severe distress and is therefore counted by the World Health Organisation (WHO) as one of the 10 most impairing illnesses. There is evidence for a strong genetic underpinning especially in early onset OCD (eoOCD). Though several genes involved in neurotransmission have been reported as candidates, there is still a need to identify new pathways. In this study, we focussed on genetic variants of the Neuropeptide Y (NPY) system. NPY is one of the most abundant neuropeptides in the human brain with emerging evidence of capacity to modulate stress response, which is of high relevance in OCD. We focussed on tag-SNPs of NPY and its receptor gene NPY1R in a family-based approach. The sample comprised 86 patients (children and adolescents) with eoOCD with both their biological parents. However, this first study on genetic variants of the NPY-system could not confirm the association between the investigated SNPs and eoOCD. Based on the small sample size results have to be interpreted as preliminary and should be replicated in larger samples. However, also in an additional GWAS analysis in a large sample, we could not observe an associations between NPY and OCD. Overall, these preliminary results point to a minor role of NPY on the stress response of OCD.
Collapse
Affiliation(s)
- Maximilian Franke
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Annette Conzelmann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Edna Grünblatt
- University Hospital of Child and Adolescent Psychiatry, University of Zurich, Zurich, Switzerland
| | - Anna M. Werling
- University Hospital of Child and Adolescent Psychiatry, University of Zurich, Zurich, Switzerland
| | - Helen Spieles
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Christoph Wewetzer
- Department of Child and Adolescent Psychiatry and Psychotherapy, Kliniken der Stadt Köln, Cologne, Germany
| | - Andreas Warnke
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Marcel Romanos
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Susanne Walitza
- University Hospital of Child and Adolescent Psychiatry, University of Zurich, Zurich, Switzerland
| | - Tobias J. Renner
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Tübingen, Tübingen, Germany
| |
Collapse
|
25
|
Nonphosphorylatable Src Ser75 Mutation Increases Ethanol Preference and Consumption in Mice. eNeuro 2019; 6:eN-NWR-0418-18. [PMID: 30963106 PMCID: PMC6451160 DOI: 10.1523/eneuro.0418-18.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 11/21/2022] Open
Abstract
Src is highly expressed in CNS neurons and contributes not only to developmental proliferation and differentiation but also to high-order brain functions, such as those contributing to alcohol consumption. Src knock-out mice exhibit no CNS abnormalities, presumably due to compensation by other Src family kinases (SFKs), but have a shortened lifespan and osteopetrosis-associated defects, impeding investigations of the role of Src on behavior in adult mice. However, the Unique domain of Src differs from those in other SFKs and is phosphorylated by cyclin-dependent kinase 1 (Cdk1) and Cdk5 at Ser75, which influences its postmitotic function in neurons. Therefore, ethanol consumption in mice harboring nonphosphorylatable (Ser75Ala) or phosphomimetic (Ser75Asp) Src mutants was investigated. Mice harboring the Ser75Ala Src mutant, but not the Ser75Asp mutant, had a higher preference for and consumption of solutions containing 5% and 10% ethanol than wild-type mice. However, plasma ethanol concentrations and sensitivities to the sedative effects of ethanol were not different among the groups. In mice harboring the Ser75Ala Src mutant, the activity of Rho-associated kinase (ROCK) in the striatum was significantly lower and Akt Ser473 phosphorylation was significantly higher than in wild-type mice. These results suggest that Src regulates voluntary ethanol drinking in a manner that depends on Ser75 phosphorylation.
Collapse
|
26
|
Tangthavewattana S, Leelawatwattana L, Prapunpoj P. The hydrophobic C-terminal sequence of transthyretin affects its catalytic kinetics towards amidated neuropeptide Y. FEBS Open Bio 2019; 9:594-604. [PMID: 30984535 PMCID: PMC6443995 DOI: 10.1002/2211-5463.12604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/20/2018] [Indexed: 12/03/2022] Open
Abstract
Transthyretin (TTR) is a transporter for thyroid hormone and retinol binding protein that has recently been reported to have proteolytic activity against certain substrates, including amidated neuropeptide Y (NPY). However, the proteolytic activity of TTR towards NPY is not fully understood. Here, we used fluorescence-based assays to determine the catalytic kinetics of human TTR towards human amidated NPY. The Michaelis constant (KM) and catalytic efficiency (kcat/KM) of TTR proteolysis were 15.88 ± 0.44 μm and 687 081 ± 35 692 m -1·s-1, respectively. In addition, we demonstrated an effect of the C-terminal sequence of TTR. When the C-terminal sequence of TTR was made more hydrophobic, the KM and kcat/KM changed to 12.87 ± 0.22 μm and 983 755 ± 18 704 m -1·s-1, respectively. Our results may be useful for the development of TTR as a therapeutic agent with low risk of the undesirable symptoms that develop from amidated NPY, and for further improvement of the kcat/KM of TTR.
Collapse
Affiliation(s)
| | - Ladda Leelawatwattana
- Department of BiochemistryFaculty of SciencePrince of Songkla UniversityHat YaiThailand
| | - Porntip Prapunpoj
- Department of BiochemistryFaculty of SciencePrince of Songkla UniversityHat YaiThailand
| |
Collapse
|
27
|
Montesano A, Baumgart M, Avallone L, Castaldo L, Lucini C, Tozzini ET, Cellerino A, D'Angelo L, de Girolamo P. Age-related central regulation of orexin and NPY in the short-lived African killifish Nothobranchius furzeri. J Comp Neurol 2019; 527:1508-1526. [PMID: 30666646 DOI: 10.1002/cne.24638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 12/28/2018] [Accepted: 01/02/2019] [Indexed: 12/15/2022]
Abstract
Orexin A (OXA) and neuropeptide Y (NPY) are two hypothalamic neuropeptides involved in the regulation of feeding behavior and food intake in all vertebrates. Accumulating evidences document that they undergo age-related modifications, with consequences on metabolism, sleep/wake disorders and progression of neurodegenerations. The present study addressed the age related changes in expression and distribution of orexin A (its precursor is also known as hypocretin-HCRT) and NPY, and their regulation by food intake in the short-lived vertebrate model Nothobranchius furzeri. Our experiments, conducted on male specimens, show that: (a) HCRT and OXA and NPY mRNA and protein are localized in neurons of diencephalon and optic tectum, as well as in numerous fibers projecting through the entire neuroaxis, and are colocalized in specific nuclei; (b) in course of aging, HCRT and NPY expressing neurons are localized also in telencephalon and rhombencephalon; (c) HCRT expressing neurons increased slightly in the diencephalic area of old animals and in fasted animals, whereas NPY increased sharply; (d) central HCRT levels are not regulated neither in course of aging nor by food intake; and (e) central NPY levels are augmented in course of aging, and regulated by food intake only in young. These findings represent a great novelty in the study of central orexinergic and NPY-ergic systems in vertebrates', demonstrating an uncommon and unprecedented described regulation of these two orexigenic neuropeptides.
Collapse
Affiliation(s)
- Alessia Montesano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy.,Leibniz-Institute on Aging - Fritz Lipmann Institute (FLI), Lab. Biology of Aging, Jena, Germany
| | - Mario Baumgart
- Leibniz-Institute on Aging - Fritz Lipmann Institute (FLI), Lab. Biology of Aging, Jena, Germany
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Luciana Castaldo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Carla Lucini
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | | | - Alessandro Cellerino
- Leibniz-Institute on Aging - Fritz Lipmann Institute (FLI), Lab. Biology of Aging, Jena, Germany.,Scuola Normale Superiore, Bio@SNS, c/o Istituto di Biofisica del CNR, Pisa, Italy
| | - Livia D'Angelo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy.,Stazione Zoologica Anton Dohrn, Biology and Evolution of Marine Organisms, Naples, Italy
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| |
Collapse
|
28
|
Berkel TDM, Zhang H, Teppen T, Sakharkar AJ, Pandey SC. Essential Role of Histone Methyltransferase G9a in Rapid Tolerance to the Anxiolytic Effects of Ethanol. Int J Neuropsychopharmacol 2018; 22:292-302. [PMID: 30590608 PMCID: PMC6441132 DOI: 10.1093/ijnp/pyy102] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/08/2018] [Accepted: 12/23/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Tolerance to ethanol-induced anxiolysis promotes alcohol intake, thus contributing to alcohol use disorder development. Recent studies implicate histone deacetylase-mediated histone H3K9 deacetylation in regulating neuropeptide Y expression during rapid ethanol tolerance to the anxiolytic effects of ethanol. Furthermore, the histone methyltransferase, G9a, and G9a-mediated H3K9 dimethylation (H3K9me2) have recently emerged as regulators of addiction and anxiety; however, their role in rapid ethanol tolerance is unknown. Therefore, we investigated the role of G9a-mediated H3K9me2 in neuropeptide Y expression during rapid ethanol tolerance. METHODS Adult male rats were administered one injection of n-saline followed by single acute ethanol injection (1 g/kg) 24 hours later (ethanol group) or 2 injections (24 hours apart) of either n-saline (saline group) or ethanol (tolerance group). Anxiety-like behaviors and global and Npy-specific G9a and H3K9me2 levels in the amygdala were measured. Effects of G9a inhibitor (UNC0642) treatment on behavioral and epigenetic measures were also examined. RESULTS Acute ethanol produced anxiolysis and decreased global H3K9me2 and G9a protein levels in the central and medial nucleus of the amygdala as well as decreased occupancy levels of H3K9me2 and G9a near a putative binding site for cAMP-response element binding protein on the Npy gene. Two identical doses of ethanol produced no behavioral or epigenetic changes relative to controls, indicating development of rapid ethanol tolerance. Interestingly, treatment with UNC0642, before the second ethanol dose reversed rapid ethanol tolerance, decreased global H3K9me2 and increased neuropeptide Y levels in the central and medial nucleus of the amygdala. CONCLUSIONS These results implicate amygdaloid G9a-mediated H3K9me2 mechanisms in regulating rapid tolerance to the anxiolytic effects of ethanol via neuropeptide Y expression regulation.
Collapse
Affiliation(s)
- Tiffani D M Berkel
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Tara Teppen
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Amul J Sakharkar
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois,Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois,Correspondence: Subhash C. Pandey, PhD, Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago; and Jesse Brown Veterans Affairs Medical Center, 1601 West Taylor Street (m/c 912), Chicago, IL 60612 ()
| |
Collapse
|
29
|
van der Vaart A, Meng X, Bowers MS, Batman AM, Aliev F, Farris SP, Hill JS, Green TA, Dick D, Wolstenholme JT, Miles MF. Glycogen synthase kinase 3 beta regulates ethanol consumption and is a risk factor for alcohol dependence. Neuropsychopharmacology 2018; 43:2521-2531. [PMID: 30188517 PMCID: PMC6224501 DOI: 10.1038/s41386-018-0202-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 01/12/2023]
Abstract
Understanding how ethanol actions on brain signal transduction and gene expression lead to excessive consumption and addiction could identify new treatments for alcohol dependence. We previously identified glycogen synthase kinase 3-beta (Gsk3b) as a member of a highly ethanol-responsive gene network in mouse medial prefrontal cortex (mPFC). Gsk3b has been implicated in dendritic function, synaptic plasticity and behavioral responses to other drugs of abuse. Here, we investigate Gsk3b in rodent models of ethanol consumption and as a risk factor for human alcohol dependence. Stereotactic viral vector gene delivery overexpression of Gsk3b in mouse mPFC increased 2-bottle choice ethanol consumption, which was blocked by lithium, a known GSK3B inhibitor. Further, Gsk3b overexpression increased anxiety-like behavior following abstinence from ethanol. Protein or mRNA expression studies following Gsk3b over-expression identified synaptojanin 2, brain-derived neurotrophic factor and the neuropeptide Y Y5 receptor as potential downstream factors altering ethanol behaviors. Rat operant studies showed that selective pharmacologic inhibition of GSK3B with TDZD-8 dose-dependently decreased motivation to self-administer ethanol and sucrose and selectively blocked ethanol relapse-like behavior. In set-based and gene-wise genetic association analysis, a GSK3b-centric gene expression network had significant genetic associations, at a gene and network level, with risk for alcohol dependence in humans. These mutually reinforcing cross-species findings implicate GSK3B in neurobiological mechanisms controlling ethanol consumption, and as both a potential risk factor and therapeutic target for alcohol dependence.
Collapse
Affiliation(s)
- Andrew van der Vaart
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Xianfang Meng
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - M Scott Bowers
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Departments of Psychiatry, Virginia Commonwealth University, Richmond, VA, 23298, USA
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Angela M Batman
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Fazil Aliev
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Departments of Psychology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- College Behavioral and Emotional Health Institute, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Sean P Farris
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Jennifer S Hill
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Thomas A Green
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | | | - Jennifer T Wolstenholme
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Michael F Miles
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
30
|
Guevara A, Gates H, Urbina B, French R. Developmental Ethanol Exposure Causes Reduced Feeding and Reveals a Critical Role for Neuropeptide F in Survival. Front Physiol 2018; 9:237. [PMID: 29623043 PMCID: PMC5875382 DOI: 10.3389/fphys.2018.00237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 03/02/2018] [Indexed: 12/25/2022] Open
Abstract
Food intake is necessary for survival, and natural reward circuitry has evolved to help ensure that animals ingest sufficient food to maintain development, growth, and survival. Drugs of abuse, including alcohol, co-opt the natural reward circuitry in the brain, and this is a major factor in the reinforcement of drug behaviors leading to addiction. At the junction of these two aspects of reward are alterations in feeding behavior due to alcohol consumption. In particular, developmental alcohol exposure (DAE) results in a collection of physical and neurobehavioral disorders collectively referred to as Fetal Alcohol Spectrum Disorder (FASD). The deleterious effects of DAE include intellectual disabilities and other neurobehavioral changes, including altered feeding behaviors. Here we use Drosophila melanogaster as a genetic model organism to study the effects of DAE on feeding behavior and the expression and function of Neuropeptide F. We show that addition of a defined concentration of ethanol to food leads to reduced feeding at all stages of development. Further, genetic conditions that reduce or eliminate NPF signaling combine with ethanol exposure to further reduce feeding, and the distribution of NPF is altered in the brains of ethanol-supplemented larvae. Most strikingly, we find that the vast majority of flies with a null mutation in the NPF receptor die early in larval development when reared in ethanol, and provide evidence that this lethality is due to voluntary starvation. Collectively, we find a critical role for NPF signaling in protecting against altered feeding behavior induced by developmental ethanol exposure.
Collapse
Affiliation(s)
- Amanda Guevara
- Biological Sciences, San Jose State University, San Jose, CA, United States
| | - Hillary Gates
- Biological Sciences, San Jose State University, San Jose, CA, United States
| | - Brianna Urbina
- Biological Sciences, San Jose State University, San Jose, CA, United States
| | - Rachael French
- Biological Sciences, San Jose State University, San Jose, CA, United States
| |
Collapse
|
31
|
Robinson SL, Thiele TE. The Role of Neuropeptide Y (NPY) in Alcohol and Drug Abuse Disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:177-197. [PMID: 29056151 DOI: 10.1016/bs.irn.2017.06.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropeptide Y (NPY) is a neuromodulator that is widely expressed throughout the central nervous system (CNS) and which is cosecreted with classic neurotransmitters including GABA and glutamate. There is a long history of research implicating a role for NPY in modulating neurobiological responses to alcohol (ethanol) as well as other drugs of abuse. Both ethanol exposure and withdrawal from chronic ethanol have been shown to produce changes in NPY and NPY receptor protein levels and mRNA expression in the CNS. Importantly, manipulations of NPY Y1 and Y2 receptor signaling have been shown to alter ethanol consumption and self-administration in a brain region-specific manner, with Y1 receptor activation and Y2 receptor blockade in regions of the extended amygdala promoting robust reductions of ethanol intake. Similar observations have been made in studies examining neurobiological responses to nicotine, psychostimulants, and opioids. When taken together with observations of potential genetic linkage between the NPY system and the human alcohol abuse disorders, NPY represents a promising target for treating problematic alcohol and drug use, and in protecting individuals from relapse during abstinence.
Collapse
Affiliation(s)
- Stacey L Robinson
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Todd E Thiele
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
32
|
Marcus DJ, Henderson-Redmond AN, Gonek M, Zee ML, Farnsworth JC, Amin RA, Andrews MJ, Davis BJ, Mackie K, Morgan DJ. Mice expressing a "hyper-sensitive" form of the CB1 cannabinoid receptor (CB1) show modestly enhanced alcohol preference and consumption. PLoS One 2017; 12:e0174826. [PMID: 28426670 PMCID: PMC5398885 DOI: 10.1371/journal.pone.0174826] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/15/2017] [Indexed: 11/17/2022] Open
Abstract
We recently characterized S426A/S430A mutant mice expressing a desensitization-resistant form of the CB1 receptor. These mice display an enhanced response to endocannabinoids and ∆9-THC. In this study, S426A/S430A mutants were used as a novel model to test whether ethanol consumption, morphine dependence, and reward for these drugs are potentiated in mice with a "hyper-sensitive" form of CB1. Using an unlimited-access, two-bottle choice, voluntary drinking paradigm, S426A/S430A mutants exhibit modestly increased intake and preference for low (6%) but not higher concentrations of ethanol. S426A/S430A mutants and wild-type mice show similar taste preference for sucrose and quinine, exhibit normal sensitivity to the hypothermic and ataxic effects of ethanol, and have normal blood ethanol concentrations following administration of ethanol. S426A/S430A mutants develop robust conditioned place preference for ethanol (2 g/kg), morphine (10 mg/kg), and cocaine (10 mg/kg), demonstrating that drug reward is not changed in S426A/S430A mutants. Precipitated morphine withdrawal is also unchanged in opioid-dependent S426A/S430A mutant mice. Although ethanol consumption is modestly changed by enhanced CB1 signaling, reward, tolerance, and acute sensitivity to ethanol and morphine are normal in this model.
Collapse
Affiliation(s)
- David J. Marcus
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA, United States of America
| | - Angela N. Henderson-Redmond
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA, United States of America
| | - Maciej Gonek
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Michael L. Zee
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA, United States of America
| | - Jill C. Farnsworth
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Randa A. Amin
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Mary-Jeanette Andrews
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Brian J. Davis
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Ken Mackie
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
| | - Daniel J. Morgan
- Department of Psychological and Brain Sciences and The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States of America
- Department of Anesthesiology, Penn State University College of Medicine, Hershey, PA, United States of America
| |
Collapse
|
33
|
Brancato A, Lavanco G, Cavallaro A, Plescia F, Cannizzaro C. Acetaldehyde, Motivation and Stress: Behavioral Evidence of an Addictive ménage à trois. Front Behav Neurosci 2017; 11:23. [PMID: 28232795 PMCID: PMC5299001 DOI: 10.3389/fnbeh.2017.00023] [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] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/27/2017] [Indexed: 11/13/2022] Open
Abstract
Acetaldehyde (ACD) contributes to alcohol's psychoactive effects through its own rewarding properties. Recent studies shed light on the behavioral correlates of ACD administration and the possible interactions with key neurotransmitters for motivation, reward and stress-related response, such as dopamine and endocannabinoids. This mini review article critically examines ACD psychoactive properties, focusing on behavioral investigations able to unveil ACD motivational effects and their pharmacological modulation in vivo. Similarly to alcohol, rats spontaneously drink ACD, whose presence is detected in the brain following chronic self-administration paradigm. ACD motivational properties are demonstrated by operant paradigms tailored to model several drug-related behaviors, such as induction and maintenance of operant self-administration, extinction, relapse and punishment resistance. ACD-related addictive-like behaviors are sensitive to pharmacological manipulations of dopamine and endocannabinoid signaling. Interestingly, the ACD-dopamine-endocannabinoids relationship also contributes to neuroplastic alterations of the NPYergic system, a stress-related peptide critically involved in alcohol abuse. The understanding of the ménage-a-trois among ACD, reward- and stress-related circuits holds promising potential for the development of novel pharmacological approaches aimed at reducing alcohol abuse.
Collapse
Affiliation(s)
- Anna Brancato
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo Palermo, Italy
| | - Gianluca Lavanco
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo Palermo, Italy
| | - Angela Cavallaro
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo Palermo, Italy
| | - Fulvio Plescia
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo Palermo, Italy
| | - Carla Cannizzaro
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo Palermo, Italy
| |
Collapse
|
34
|
Park A, Ghezzi A, Wijesekera TP, Atkinson NS. Genetics and genomics of alcohol responses in Drosophila. Neuropharmacology 2017; 122:22-35. [PMID: 28161376 DOI: 10.1016/j.neuropharm.2017.01.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/24/2017] [Accepted: 01/29/2017] [Indexed: 02/07/2023]
Abstract
Drosophila melanogaster has become a significant model organism for alcohol research. In flies, a rich variety of behaviors can be leveraged for identifying genes affecting alcohol responses and adaptations. Furthermore, almost all genes can be easily genetically manipulated. Despite the great evolutionary distance between flies and mammals, many of the same genes have been implicated in strikingly similar alcohol-induced behaviors. A major problem in medical research today is that it is difficult to extrapolate from any single model system to humans. Strong evolutionary conservation of a mechanistic response between distantly related organisms, such as flies and mammals, is a powerful predictor that conservation will continue all the way to humans. This review describes the state of the Drosophila alcohol research field. It describes common alcohol behavioral assays, the independent origins of resistance and tolerance, the results of classical genetic screens and candidate gene analysis, and the outcomes of recent genomics studies employing GWAS, transcriptome, miRNA, and genome-wide histone acetylation surveys. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Annie Park
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Alfredo Ghezzi
- Department of Biology, University of Puerto Rico, Rio Piedras. San Juan, PR, United States
| | - Thilini P Wijesekera
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Nigel S Atkinson
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
35
|
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: 45] [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
|
36
|
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
|
37
|
Su Y, Foppen E, Fliers E, Kalsbeek A. Effects of Intracerebroventricular Administration of Neuropeptide Y on Metabolic Gene Expression and Energy Metabolism in Male Rats. Endocrinology 2016; 157:3070-85. [PMID: 27267712 DOI: 10.1210/en.2016-1083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neuropeptide Y (NPY) is an important neurotransmitter in the control of energy metabolism. Several studies have shown that obesity is associated with increased levels of NPY in the hypothalamus. We hypothesized that the central release of NPY has coordinated and integrated effects on energy metabolism in different tissues, resulting in increased energy storage and decreased energy expenditure (EE). We first investigated the acute effects of an intracerebroventricular (ICV) infusion of NPY on gene expression in liver, brown adipose tissue, soleus muscle, and sc and epididymal white adipose tissue (WAT). We found increased expression of genes involved in gluconeogenesis and triglyceride secretion in the liver already 2-hour after the start of the NPY administration. In brown adipose tissue, the expression of thermogenic genes was decreased. In sc WAT, the expression of genes involved in lipogenesis was increased, whereas in soleus muscle, the expression of lipolytic genes was decreased after ICV NPY. These findings indicate that the ICV infusion of NPY acutely and simultaneously increases lipogenesis and decreases lipolysis in different tissues. Subsequently, we investigated the acute effects of ICV NPY on locomotor activity, respiratory exchange ratio, EE, and body temperature. The ICV infusion of NPY increased locomotor activity, body temperature, and EE as well as respiratory exchange ratio. Together, these results show that an acutely increased central availability of NPY results in a shift of metabolism towards lipid storage and an increased use of carbohydrates, while at the same time increasing activity, EE, and body temperature.
Collapse
Affiliation(s)
- Yan Su
- Hypothalamic Integration Mechanisms (Y.S., A.K.), Netherlands Institute for Neuroscience, 1105 BA Amsterdam, The Netherlands; and Department of Endocrinology and Metabolism (E.Fo.,E.Fl., A.K.), Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Ewout Foppen
- Hypothalamic Integration Mechanisms (Y.S., A.K.), Netherlands Institute for Neuroscience, 1105 BA Amsterdam, The Netherlands; and Department of Endocrinology and Metabolism (E.Fo.,E.Fl., A.K.), Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Eric Fliers
- Hypothalamic Integration Mechanisms (Y.S., A.K.), Netherlands Institute for Neuroscience, 1105 BA Amsterdam, The Netherlands; and Department of Endocrinology and Metabolism (E.Fo.,E.Fl., A.K.), Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms (Y.S., A.K.), Netherlands Institute for Neuroscience, 1105 BA Amsterdam, The Netherlands; and Department of Endocrinology and Metabolism (E.Fo.,E.Fl., A.K.), Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
38
|
Qiu B, Bell RL, Cao Y, Zhang L, Stewart RB, Graves T, Lumeng L, Yong W, Liang T. Npy deletion in an alcohol non-preferring rat model elicits differential effects on alcohol consumption and body weight. J Genet Genomics 2016; 43:421-30. [PMID: 27461754 PMCID: PMC5055068 DOI: 10.1016/j.jgg.2016.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 04/21/2016] [Accepted: 04/27/2016] [Indexed: 11/21/2022]
Abstract
Neuropeptide Y (NPY) is widely expressed in the central nervous system and influences many physiological processes. It is located within the rat quantitative trait locus (QTL) for alcohol preference on chromosome 4. Alcohol-nonpreferring (NP) rats consume very little alcohol, but have significantly higher NPY expression in the brain than alcohol-preferring (P) rats. We capitalized on this phenotypic difference by creating an Npy knockout (KO) rat using the inbred NP background to evaluate NPY effects on alcohol consumption. Zinc finger nuclease (ZNF) technology was applied, resulting in a 26-bp deletion in the Npy gene. RT-PCR, Western blotting and immunohistochemistry confirmed the absence of Npy mRNA and protein in KO rats. Alcohol consumption was increased in Npy(+/-) but not Npy(-/-) rats, while Npy(-/-) rats displayed significantly lower body weight when compared to Npy(+/+) rats. In whole brain tissue, expression levels of Npy-related and other alcohol-associated genes, Npy1r, Npy2r, Npy5r, Agrp, Mc3r, Mc4r, Crh and Crh1r, were significantly greater in Npy(-/-) rats, whereas Pomc and Crhr2 expressions were highest in Npy(+/-) rats. These findings suggest that the NPY-system works in close coordination with the melanocortin (MC) and corticotropin-releasing hormone (CRH) systems to modulate alcohol intake and body weight.
Collapse
Affiliation(s)
- Bin Qiu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yong Cao
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China; Experimental Medicine Center, The First Affiliated Hospital of Sichuan Medical University, Luzhou 646000, China
| | - Lingling Zhang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Robert B Stewart
- Department of Psychology, Purdue School of Science, Indiana University-Purdue University of Indianapolis, Indianapolis, IN 46202, USA
| | - Tamara Graves
- Department of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lawrence Lumeng
- Department of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Weidong Yong
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China.
| | - Tiebing Liang
- Department of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
39
|
|
40
|
Gonçalves J, Martins J, Baptista S, Ambrósio AF, Silva AP. Effects of drugs of abuse on the central neuropeptide Y system. Addict Biol 2016; 21:755-65. [PMID: 25904345 DOI: 10.1111/adb.12250] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Neuropeptide Y (NPY), which is widely expressed in the central nervous system is involved in several neuropathologies including addiction. Here we comprehensively and systematically review alterations on the central NPY system induced by several drugs. We report on the effects of psychostimulants [cocaine, amphetamine, methamphetamine, 3,4-methylenedioxymethamphetamine (MDMA) and nicotine], ethanol, and opioids on NPY protein levels and expression of different NPY receptors. Overall, expression and function of NPY and its receptors are changed under conditions of drug exposure, thus affecting several physiologic behaviors, such as feeding, stress and anxiety. Drugs of abuse differentially affect the components of the NPY system. For example methamphetamine and nicotine lead to a consistent increase in NPY mRNA and protein levels in different brain sites whereas ethanol and opioids decrease NPY mRNA and protein expression. Drug-induced alterations on the different NPY receptors show more complex regulation pattern. Manipulation of the NPY system can have opposing effects on reinforcing and addictive properties of drugs of abuse. NPY can produce pro-addictive effects (nicotine and heroin), but can also exert inhibitory effects on addictive behavior (AMPH, ethanol). Furthermore, NPY can act as a neuroprotective agent in chronically methamphetamine and MDMA-treated rodents. In conclusion, manipulation of the NPY system seems to be a potential target to counteract neural alterations, addiction-related behaviors and cognitive deficits induced by these drugs.
Collapse
Affiliation(s)
- Joana Gonçalves
- Institute of Nuclear Sciences Applied to Health (ICNAS); University of Coimbra; Portugal
- Institute for Biomedical Imaging and Life Sciences (IBILI); University of Coimbra; Portugal
- Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit; University of Coimbra; Portugal
| | - João Martins
- Institute for Biomedical Imaging and Life Sciences (IBILI); University of Coimbra; Portugal
- Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit; University of Coimbra; Portugal
- Centre of Ophthalmology and Vision Sciences; Faculty of Medicine; University of Coimbra; Portugal
| | - Sofia Baptista
- Institute for Biomedical Imaging and Life Sciences (IBILI); University of Coimbra; Portugal
- Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit; University of Coimbra; Portugal
- Laboratory of Pharmacology and Experimental Therapeutics; Faculty of Medicine; University of Coimbra; Portugal
| | - António Francisco Ambrósio
- Institute for Biomedical Imaging and Life Sciences (IBILI); University of Coimbra; Portugal
- Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit; University of Coimbra; Portugal
- Centre of Ophthalmology and Vision Sciences; Faculty of Medicine; University of Coimbra; Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI); Portugal
| | - Ana Paula Silva
- Institute for Biomedical Imaging and Life Sciences (IBILI); University of Coimbra; Portugal
- Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit; University of Coimbra; Portugal
- Laboratory of Pharmacology and Experimental Therapeutics; Faculty of Medicine; University of Coimbra; Portugal
| |
Collapse
|
41
|
Borkar CD, Upadhya MA, Shelkar GP, Subhedar NK, Kokare DM. Neuropeptide Y system in accumbens shell mediates ethanol self-administration in posterior ventral tegmental area. Addict Biol 2016; 21:766-75. [PMID: 25929272 DOI: 10.1111/adb.12254] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although modulatory effects of neuropeptide Y (NPY) on ethanol consumption are well established, its role in ethanol reward, in the framework of mesolimbic dopaminergic system, has not been studied. We investigated the influence of nucleus accumbens shell (AcbSh) NPYergic system on ethanol self-administration in posterior ventral tegmental area (p-VTA) using intracranial self-administration paradigm. Rats were stereotaxically implanted with cannulae targeted unilaterally at the right p-VTA and trained to self-administer ethanol (200 mg%) in standard two-lever (active/inactive) operant chamber, an animal model with high predictive validity to test the rewarding mechanisms. Over a period of 7 days, these rats showed a significant increase in the number of lever presses for ethanol self-administration suggesting reinforcement. While intra-AcbSh NPY (1 or 2 ng/rat) or [Leu(31) , Pro(34) ]-NPY (0.5 or 1 ng/rat) dose-dependently increased ethanol self-administration, BIBP3226 (0.4 or 0.8 ng/rat) produced opposite effect. The rats conditioned to self-administer ethanol showed significant increase in the population of NPY-immunoreactive cells and fibres in the AcbSh, central nucleus of amygdala (CeA), hypothalamic arcuate nucleus (ARC) and lateral part of bed nucleus of stria terminalis as compared with that in the naïve rats. Neuronal tracing studies showed that NPY innervations in the AcbSh may derive from the neurons of ARC and CeA. As NPY and dopamine systems in reward areas are known to interact, we suggest that NPY inputs from ARC and CeA may play an important role in modulation of the dopaminergic system in the AcbSh and consequently influence the ethanol induced reward and addiction.
Collapse
Affiliation(s)
- Chandrashekhar D. Borkar
- Department of Pharmaceutical Sciences; Rashtrasant Tukadoji Maharaj Nagpur University; Nagpur Maharashtra India
| | - Manoj A. Upadhya
- Pharmacology Department; Shrimati Kishoritai Bhoyar College of Pharmacy; New Kamptee, Nagpur Maharashtra India
| | - Gajanan P. Shelkar
- Department of Pharmaceutical Sciences; Rashtrasant Tukadoji Maharaj Nagpur University; Nagpur Maharashtra India
| | - Nishikant K. Subhedar
- Biology Department; Indian Institute of Science Education and Research (IISER); Pune Maharashtra India
| | - Dadasaheb M. Kokare
- Department of Pharmaceutical Sciences; Rashtrasant Tukadoji Maharaj Nagpur University; Nagpur Maharashtra India
| |
Collapse
|
42
|
Castilla-Ortega E, Pavón FJ, Sánchez-Marín L, Estivill-Torrús G, Pedraza C, Blanco E, Suárez J, Santín L, Rodríguez de Fonseca F, Serrano A. Both genetic deletion and pharmacological blockade of lysophosphatidic acid LPA1 receptor results in increased alcohol consumption. Neuropharmacology 2016; 103:92-103. [DOI: 10.1016/j.neuropharm.2015.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/25/2015] [Accepted: 12/11/2015] [Indexed: 12/21/2022]
|
43
|
Fritz BM, Boehm SL. Rodent models and mechanisms of voluntary binge-like ethanol consumption: Examples, opportunities, and strategies for preclinical research. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:297-308. [PMID: 26021391 PMCID: PMC4668238 DOI: 10.1016/j.pnpbp.2015.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 05/02/2015] [Accepted: 05/21/2015] [Indexed: 02/03/2023]
Abstract
Binge ethanol consumption has widespread negative consequences for global public health. Rodent models offer exceptional power to explore the neurobiology underlying and affected by binge-like drinking as well as target potential prevention, intervention, and treatment strategies. An important characteristic of these models is their ability to consistently produce pharmacologically-relevant blood ethanol concentration. This review examines the current available rodent models of voluntary, pre-dependent binge-like ethanol consumption and their utility in various research strategies. Studies have demonstrated that a diverse array of neurotransmitters regulate binge-like drinking, resembling some findings from other drinking models. Furthermore, repeated binge-like drinking recruits neuroadaptive mechanisms in mesolimbocortical reward circuitry. New opportunities that these models offer in the current context of mechanistic research are also discussed.
Collapse
Affiliation(s)
| | - Stephen L Boehm
- Indiana Alcohol Research Center, Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States.
| |
Collapse
|
44
|
Van Skike CE, Maggio SE, Reynolds AR, Casey EM, Bardo MT, Dwoskin LP, Prendergast MA, Nixon K. Critical needs in drug discovery for cessation of alcohol and nicotine polysubstance abuse. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:269-87. [PMID: 26582145 PMCID: PMC4679525 DOI: 10.1016/j.pnpbp.2015.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 11/10/2015] [Accepted: 11/11/2015] [Indexed: 01/29/2023]
Abstract
Polysubstance abuse of alcohol and nicotine has been overlooked in our understanding of the neurobiology of addiction and especially in the development of novel therapeutics for its treatment. Estimates show that as many as 92% of people with alcohol use disorders also smoke tobacco. The health risks associated with both excessive alcohol consumption and tobacco smoking create an urgent biomedical need for the discovery of effective cessation treatments, as opposed to current approaches that attempt to independently treat each abused agent. The lack of treatment approaches for alcohol and nicotine abuse/dependence mirrors a similar lack of research in the neurobiology of polysubstance abuse. This review discusses three critical needs in medications development for alcohol and nicotine co-abuse: (1) the need for a better understanding of the clinical condition (i.e. alcohol and nicotine polysubstance abuse), (2) the need to better understand how these drugs interact in order to identify new targets for therapeutic development and (3) the need for animal models that better mimic this human condition. Current and emerging treatments available for the cessation of each drug and their mechanisms of action are discussed within this context followed by what is known about the pharmacological interactions of alcohol and nicotine. Much has been and will continue to be gained from studying comorbid alcohol and nicotine exposure.
Collapse
Affiliation(s)
- C E Van Skike
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - S E Maggio
- Department of Psychology, University of Kentucky, Lexington, KY 40536, United States
| | - A R Reynolds
- Department of Psychology, University of Kentucky, Lexington, KY 40536, United States
| | - E M Casey
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - M T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY 40536, United States; Center for Drug Abuse and Research Translation, University of Kentucky, Lexington, KY 40536, United States; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, United States
| | - L P Dwoskin
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States; Center for Drug Abuse and Research Translation, University of Kentucky, Lexington, KY 40536, United States
| | - M A Prendergast
- Department of Psychology, University of Kentucky, Lexington, KY 40536, United States; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, United States
| | - K Nixon
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, United States.
| |
Collapse
|
45
|
Barkley-Levenson AM, Ryabinin AE, Crabbe JC. Neuropeptide Y response to alcohol is altered in nucleus accumbens of mice selectively bred for drinking to intoxication. Behav Brain Res 2016; 302:160-70. [PMID: 26779672 DOI: 10.1016/j.bbr.2016.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/06/2015] [Accepted: 01/05/2016] [Indexed: 11/17/2022]
Abstract
The High Drinking in the Dark (HDID) mice have been selectively bred for drinking to intoxicating blood alcohol levels and represent a genetic model of risk for binge-like drinking. Presently, little is known about the specific genetic factors that promote excessive intake in these mice. Previous studies have identified neuropeptide Y (NPY) as a potential target for modulating alcohol intake. NPY expression differs in some rodent lines that have been selected for high and low alcohol drinking phenotypes, as well as inbred mouse strains that differ in alcohol preference. Alcohol drinking and alcohol withdrawal also produce differential effects on NPY expression in the brain. Here, we assessed brain NPY protein levels in HDID mice of two replicates of selection and control heterogeneous stock (HS) mice at baseline (water drinking) and after binge-like alcohol drinking to determine whether selection is associated with differences in NPY expression and its sensitivity to alcohol. NPY levels did not differ between HDID and HS mice in any brain region in the water-drinking animals. HS mice showed a reduction in NPY levels in the nucleus accumbens (NAc) - especially in the shell - in ethanol-drinking animals vs. water-drinking controls. However, HDID mice showed a blunted NPY response to alcohol in the NAc core and shell compared to HS mice. These findings suggest that the NPY response to alcohol has been altered by selection for drinking to intoxication in a region-specific manner. Thus, the NPY system may represent a potential target for altering binge-like alcohol drinking in these mice.
Collapse
Affiliation(s)
- Amanda M Barkley-Levenson
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States; Portland Alcohol Research Center, Portland, OR 97239, United States; VA Portland Health Care System, Portland, OR 97239, United States.
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States; Portland Alcohol Research Center, Portland, OR 97239, United States
| | - John C Crabbe
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States; Portland Alcohol Research Center, Portland, OR 97239, United States; VA Portland Health Care System, Portland, OR 97239, United States
| |
Collapse
|
46
|
Mayfield J, Arends MA, Harris RA, Blednov YA. Genes and Alcohol Consumption: Studies with Mutant Mice. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:293-355. [PMID: 27055617 PMCID: PMC5302130 DOI: 10.1016/bs.irn.2016.02.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this chapter, we review the effects of global null mutant and overexpressing transgenic mouse lines on voluntary self-administration of alcohol. We examine approximately 200 publications pertaining to the effects of 155 mouse genes on alcohol consumption in different drinking models. The targeted genes vary in function and include neurotransmitter, ion channel, neuroimmune, and neuropeptide signaling systems. The alcohol self-administration models include operant conditioning, two- and four-bottle choice continuous and intermittent access, drinking in the dark limited access, chronic intermittent ethanol, and scheduled high alcohol consumption tests. Comparisons of different drinking models using the same mutant mice are potentially the most informative, and we will highlight those examples. More mutants have been tested for continuous two-bottle choice consumption than any other test; of the 137 mouse genes examined using this model, 97 (72%) altered drinking in at least one sex. Overall, the effects of genetic manipulations on alcohol drinking often depend on the sex of the mice, alcohol concentration and time of access, genetic background, as well as the drinking test.
Collapse
Affiliation(s)
- J Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - M A Arends
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, United States
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States.
| | - Y A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
47
|
Keil MF, Briassoulis G, Stratakis CA. The Role of Protein Kinase A in Anxiety Behaviors. Neuroendocrinology 2016; 103:625-39. [PMID: 26939049 DOI: 10.1159/000444880] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/19/2016] [Indexed: 11/19/2022]
Abstract
This review focuses on the genetic and other evidence supporting the notion that the cyclic AMP (cAMP) signaling pathway and its mediator, the protein kinase A (PKA) enzyme, which respond to environmental stressors and regulate stress responses, are central to the pathogenesis of disorders related to anxiety. We describe the PKA pathway and review in vitro animal studies (mouse) and other evidence that support the importance of PKA in regulating behaviors that lead to anxiety. Since cAMP signaling and PKA have been pharmacologically exploited since the 1940s (even before the identification of cAMP as a second messenger with PKA as its mediator) for a number of disorders from asthma to cardiovascular diseases, there is ample opportunity to develop therapies using this new knowledge about cAMP, PKA, and anxiety disorders.
Collapse
Affiliation(s)
- Margaret F Keil
- Section on Endocrinology and Genetics, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Md., USA
| | | | | |
Collapse
|
48
|
Shirahase T, Aoki M, Watanabe R, Watanabe Y, Tanaka M. Increased alcohol consumption in relaxin-3 deficient male mice. Neurosci Lett 2015; 612:155-160. [PMID: 26687275 DOI: 10.1016/j.neulet.2015.12.014] [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: 11/10/2015] [Revised: 12/03/2015] [Accepted: 12/09/2015] [Indexed: 11/29/2022]
Abstract
Relaxin-3 is a neuropeptide expressed in the brainstem, and predominantly localized in the gray matter of the midline dorsal pons termed the nucleus incertus. Relaxin-3-expressing neurons densely project axons rostrally to various forebrain regions including the septum, hippocampus, and lateral hypothalamus. Several relaxin-3 functions have been reported including food intake, stress responses, neuroendocrine function, emotion, and spatial memory. In addition, recently relaxin-3 and its receptor, RXFP3, were shown to regulate alcohol intake using an RXFP3 antagonist and RXFP3 gene knockout mice. In the present study, we investigated alcohol consumption in relaxin-3 knockout mice, and found that male but not female mice significantly drank more alcohol than wild-type mice in the two-bottle choice test. However, after chronic alcohol vapor exposure, wild-type and mutant mice did not show this difference in alcohol intake, although both genotypes exhibited increased alcohol consumption compared with non-alcohol-exposed control mice. There was no genotype difference in sucrose or quinine preference. These results suggest that the relaxin-3 neuronal system modestly affects alcohol preference and consumption.
Collapse
Affiliation(s)
- Takahira Shirahase
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan; Department of Dental Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Miku Aoki
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan; Department of Dental Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Ryuji Watanabe
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Yoshihisa Watanabe
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan
| | - Masaki Tanaka
- Department of Basic Geriatrics, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-ku, Kyoto 602-8566, Japan.
| |
Collapse
|
49
|
Korpi ER, den Hollander B, Farooq U, Vashchinkina E, Rajkumar R, Nutt DJ, Hyytiä P, Dawe GS. Mechanisms of Action and Persistent Neuroplasticity by Drugs of Abuse. Pharmacol Rev 2015; 67:872-1004. [DOI: 10.1124/pr.115.010967] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
50
|
Munn-Chernoff MA, Grant JD, Bucholz KK, Agrawal A, Lynskey MT, Madden PAF, Heath AC, Duncan AE. Bulimic Behaviors and Early Substance Use: Findings from a Cotwin-Control Study. Alcohol Clin Exp Res 2015; 39:1740-8. [PMID: 26248308 DOI: 10.1111/acer.12829] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/26/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Bulimic behaviors (i.e., binge eating and compensatory behaviors) and substance use frequently co-occur. However, the etiology underlying this association is poorly understood. This study evaluated the association between bulimic behaviors and early substance use, controlling for genetic and shared environmental factors. METHODS Participants were 3,540 young adult women from the Missouri Adolescent Female Twin Study. A telephone adaptation of the Semi-Structured Assessment for the Genetics of Alcoholism interview assessed DSM-IV bulimic behaviors, substance use, and other psychological characteristics. Lifetime bulimic behaviors were examined in twin pairs concordant and discordant for early substance use. Logistic regressions were adjusted for the nonindependence of twin data, zygosity, age, body mass index, early menarche (onset before age 12), and early sex (first consensual sexual intercourse before age 15). RESULTS In the entire study population, women who reported early use of alcohol or nicotine were more likely to engage in bulimic behaviors after adjusting for covariates. In 53 pairs of monozygotic twins discordant for alcohol experimentation before age 15, the twin who reported early alcohol experimentation had 3.21 (95% confidence interval = 1.54 to 6.67) times higher odds of reporting bulimic behaviors than the cotwin who did not report early alcohol experimentation, even after adjustment for covariates. CONCLUSIONS Findings suggest that early alcohol experimentation may contribute to the development of bulimic behaviors via mechanisms extending beyond shared vulnerability, including individual-specific environmental experiences or causal pathways.
Collapse
Affiliation(s)
- Melissa A Munn-Chernoff
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri.,Midwest Alcoholism Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Julia D Grant
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri.,Midwest Alcoholism Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Kathleen K Bucholz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri.,Midwest Alcoholism Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri.,Midwest Alcoholism Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Michael T Lynskey
- Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, United Kingdom
| | - Pamela A F Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri.,Midwest Alcoholism Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew C Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri.,Midwest Alcoholism Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Alexis E Duncan
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri.,Midwest Alcoholism Research Center, Washington University School of Medicine, St. Louis, Missouri.,George Warren Brown School of Social Work, Washington University, St. Louis, Missouri
| |
Collapse
|