1
|
Samanta S, Bagchi D, Gold MS, Badgaiyan RD, Barh D, Blum K. A Complex Relationship Among the Circadian Rhythm, Reward Circuit and Substance Use Disorder (SUD). Psychol Res Behav Manag 2024; 17:3485-3501. [PMID: 39411118 PMCID: PMC11479634 DOI: 10.2147/prbm.s473310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
The human brain not only controls the various physiological functions but is also the prime regulator of circadian rhythms, rewards, and behaviors. Environmental factors, professional stress, and social disintegration are regarded as the initial causative factors of addiction behavior. Shift work, artificial light exposure at night, and chronic and acute jet lag influence circadian rhythm dysfunction. The result is impaired neurotransmitter release, dysfunction of neural circuits, endocrine disturbance, and metabolic disorder, leading to advancement in substance use disorder. There is a bidirectional relationship between chronodisruption and addiction behavior. Circadian rhythm dysfunction, neuroadaptation in the reward circuits, and alteration in clock gene expression in the mesolimbic areas influence substance use disorder (SUD), and chronotherapy has potential benefits in the treatment strategies. This review explores the relationship among the circadian rhythm dysfunction, reward circuit, and SUD. The impact of chronotherapy on SUD has also been discussed.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore, West Bengal, 721101, India
| | - Debasis Bagchi
- Department of Biology, College of Arts and Sciences, Adelphi University, Garden City, NY, USA and Department of Psychology, Gordon F. Derner School of Psychology, Adelphi University, Garden City, NY, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Southern University, Houston, TX, 77004, USA
| | - Mark S Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Rajendra D Badgaiyan
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Debmalya Barh
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, BeloHorizonte, 31270-901, Brazil
- Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, 721172, West Bengal, India
| | - Kenneth Blum
- Division of Addiction Research & Education, Center for Sports, Exercise, and Mental Health, Western University Health Sciences, Pomona, CA, 91766, USA
- Institute of Psychology, Eotvos Loránd University, Budapest, 1053, Hungary
- Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Center, Dayton, OH, 45435, USA
- Department of Psychiatry, University of Vermont, Burlington, VT, 05405, USA
- Division of Nutrigenomics, The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX, 78701, USA
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| |
Collapse
|
2
|
DePoy LM, Petersen KA, Zong W, Ketchesin KD, Matthaei RC, Yin R, Perez MS, Vadnie CA, Becker-Krail D, Scott MR, Tseng GC, McClung CA. Cell-type and sex-specific rhythmic gene expression in the nucleus accumbens. Mol Psychiatry 2024; 29:3117-3127. [PMID: 38678086 PMCID: PMC11449664 DOI: 10.1038/s41380-024-02569-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Circadian rhythms are critical for human health and are highly conserved across species. Disruptions in these rhythms contribute to many diseases, including psychiatric disorders. Previous results suggest that circadian genes modulate behavior through specific cell types in the nucleus accumbens (NAc), particularly dopamine D1-expressing medium spiny neurons (MSNs). However, diurnal rhythms in transcript expression have not been investigated in NAc MSNs. In this study we identified and characterized rhythmic transcripts in D1- and D2-expressing neurons and compared rhythmicity results to homogenate as well as astrocyte samples taken from the NAc of male and female mice. We find that all cell types have transcripts with diurnal rhythms and that top rhythmic transcripts are largely core clock genes, which peak at approximately the same time of day in each cell type and sex. While clock-controlled rhythmic transcripts are enriched for protein regulation pathways across cell type, cell signaling and signal transduction related processes are most commonly enriched in MSNs. In contrast to core clock genes, these clock-controlled rhythmic transcripts tend to reach their peak in expression about 2-h later in females than males, suggesting diurnal rhythms in reward may be delayed in females. We also find sex differences in pathway enrichment for rhythmic transcripts peaking at different times of day. Protein folding and immune responses are enriched in transcripts that peak in the dark phase, while metabolic processes are primarily enriched in transcripts that peak in the light phase. Importantly, we also find that several classic markers used to categorize MSNs are rhythmic in the NAc. This is critical since the use of rhythmic markers could lead to over- or under-enrichment of targeted cell types depending on the time at which they are sampled. This study greatly expands our knowledge of how individual cell types contribute to rhythms in the NAc.
Collapse
Affiliation(s)
- Lauren M DePoy
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Kaitlyn A Petersen
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Kyle D Ketchesin
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Ross C Matthaei
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA
| | - RuoFei Yin
- Department of Biostatistics, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Megan S Perez
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Chelsea A Vadnie
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, 43015, Delaware, OH, USA
| | - Darius Becker-Krail
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Madeline R Scott
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Colleen A McClung
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, 15219, Pittsburgh, PA, USA.
- Center for Neuroscience, University of Pittsburgh, 15261, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Freyberg Z, Andreazza AC, McClung CA, Phillips ML. Linking Mitochondrial Dysfunction, Neurotransmitter, and Neural Network Abnormalities and Mania: Elucidating Neurobiological Mechanisms of the Therapeutic Effect of the Ketogenic Diet in Bipolar Disorder. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024:S2451-9022(24)00199-X. [PMID: 39053576 DOI: 10.1016/j.bpsc.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
There is growing interest in the ketogenic diet as a treatment for bipolar disorder (BD), and there are promising anecdotal and small case study reports of efficacy. However, the neurobiological mechanisms by which diet-induced ketosis might ameliorate BD symptoms remain to be determined, particularly in manic and hypomanic states-defining features of BD. Identifying these mechanisms will provide new markers to guide personalized interventions and provide targets for novel treatment developments for individuals with BD. In this critical review, we describe recent findings highlighting 2 types of neurobiological abnormalities in BD: 1) mitochondrial dysfunction and 2) neurotransmitter and neural network functional abnormalities. We link these abnormalities to mania/hypomania and depression in BD and then describe the biological underpinnings by which the ketogenic diet may have a beneficial effect in individuals with BD. We end the review by describing approaches that can be employed in future studies to elucidate the neurobiology that underlies the therapeutic effect of the ketogenic diet in BD. Doing this may provide marker predictors to identify individuals who will respond well to the ketogenic diet, as well as offer neural targets for novel treatment developments for BD.
Collapse
Affiliation(s)
- Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Ana C Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Colleen A McClung
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary L Phillips
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
4
|
Freund N, Haussleiter I. Bipolar Chronobiology in Men and Mice: A Narrative Review. Brain Sci 2023; 13:738. [PMID: 37239210 PMCID: PMC10216184 DOI: 10.3390/brainsci13050738] [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: 03/22/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
In patients with bipolar disorder, we do not only see a cycling of mood episodes, but also a shift in circadian rhythm. In the present overview, the circadian rhythm, the "internal clock", and their disruptions are briefly described. In addition, influences on circadian rhythms such as sleep, genetics, and environment are discussed. This description is conducted with a translational focus covering human patients as well as animal models. Concluding the current knowledge on chronobiology and bipolar disorder, implications for specificity and the course of bipolar disorder and treatment options are given at the end of this article. Taken together, circadian rhythm disruption and bipolar disorder are strongly correlated; the exact causation, however, is still unclear.
Collapse
Affiliation(s)
- Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University, 44791 Bochum, Germany;
| | - Ida Haussleiter
- Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University, 44791 Bochum, Germany
| |
Collapse
|
5
|
Olejniczak I, Begemann K, Wilhelm I, Oster H. The circadian neurobiology of reward. Acta Physiol (Oxf) 2023; 237:e13928. [PMID: 36625310 DOI: 10.1111/apha.13928] [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: 09/19/2022] [Revised: 11/29/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
Circadian clocks are important regulators of physiology and behavior. In the brain, circadian clocks have been described in many centers of the central reward system. They affect neurotransmitter signaling, neuroendocrine circuits, and the sensitivity to external stimulation. Circadian disruption affects reward signaling, promoting the development of behavioral and substance use disorders. In this review, we summarize our current knowledge of circadian clock-reward crosstalk. We show how chronodisruption affects reward signaling in different animal models. We then translate these findings to circadian aspects of human reward (dys-) function and its clinical implications. Finally, we devise approaches to and challenges in implementing the concepts of circadian medicine in the therapy of substance use disorders.
Collapse
Affiliation(s)
- Iwona Olejniczak
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| | - Kimberly Begemann
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| | - Ines Wilhelm
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany.,Translational Psychiatry Unit, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| |
Collapse
|
6
|
Gogarnoiu ES, Vogt CD, Sanchez J, Bonifazi A, Saab E, Shaik AB, Soler-Cedeño O, Bi GH, Klein B, Xi ZX, Lane JR, Newman AH. Dopamine D 3/D 2 Receptor Ligands Based on Cariprazine for the Treatment of Psychostimulant Use Disorders That May Be Dual Diagnosed with Affective Disorders. J Med Chem 2023; 66:1809-1834. [PMID: 36661568 PMCID: PMC11100975 DOI: 10.1021/acs.jmedchem.2c01624] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Highly selective dopamine D3 receptor (D3R) partial agonists/antagonists have been developed for the treatment of psychostimulant use disorders (PSUD). However, none have reached the clinic due to insufficient potency/efficacy or potential cardiotoxicity. Cariprazine, an FDA-approved drug for the treatment of schizophrenia and bipolar disorder, is a high-affinity D3R partial agonist (Ki = 0.22 nM) with 3.6-fold selectivity over the homologous dopamine D2 receptor (D2R). We hypothesized that compounds that are moderately D3R/D2R-selective partial agonists/antagonists may be effective for the treatment of PSUD. By systematically modifying the parent molecule, we discovered partial agonists/antagonists, as measured in bioluminescence resonance energy transfer (BRET)-based assays, with high D3R affinities (Ki = 0.14-50 nM) and moderate selectivity (<100-fold) over D2R. Cariprazine and two lead analogues, 13a and 13e, decreased cocaine self-administration (FR2; 1-10 mg/kg, i.p.) in rats, suggesting that partial agonists/antagonists with modest D3R/D2R selectivity may be effective in treating PSUD and potentially comorbidities with other affective disorders.
Collapse
Affiliation(s)
- Emma S. Gogarnoiu
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Caleb D. Vogt
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Julie Sanchez
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Midlands NG2 7AG, United Kingdom
| | - Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Elizabeth Saab
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Anver Basha Shaik
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Omar Soler-Cedeño
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Guo-Hua Bi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Benjamin Klein
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Zheng-Xiong Xi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - J. Robert Lane
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Midlands NG2 7AG, United Kingdom
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|
7
|
Al-Omari A, Kecskés M, Gaszner B, Biró-Sütő T, Fazekas B, Berta G, Kuzma M, Pintér E, Kormos V. Functionally active TRPA1 ion channel is downregulated in peptidergic neurons of the Edinger-Westphal nucleus upon acute alcohol exposure. Front Cell Dev Biol 2023; 10:1046559. [PMID: 36704197 PMCID: PMC9872022 DOI: 10.3389/fcell.2022.1046559] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction: The centrally projecting Edinger-Westphal nucleus (EWcp) contributes to the control of alcohol consumption by its urocortin 1 (UCN1) and cocaine- and amphetamine-regulated transcript (CART) co-expressing peptidergic neurons. Our group recently showed that the urocortinergic centrally projecting EWcp is the primary seat of central nervous system transient receptor potential ankyrin 1 (TRPA1) cation channel mRNA expression. Here, we hypothesized that alcohol and its metabolites, that pass through the blood-brain barrier, may influence the function of urocortinergic cells in centrally projecting EWcp by activating TRPA1 ion channels. We aimed to examine the functional activity of TRPA1 in centrally projecting EWcp and its possible role in a mouse model of acute alcohol exposure. Methods: Electrophysiological measurements were performed on acute brain slices of C57BL/6J male mice containing the centrally projecting EWcp to prove the functional activity of TRPA1 using a selective, potent, covalent agonist JT010. Male TRPA1 knockout (KO) and wildtype (WT) mice were compared with each other in the morphological studies upon acute alcohol treatment. In both genotypes, half of the animals was treated intraperitoneally with 1 g/kg 6% ethanol vs. physiological saline-injected controls. Transcardial perfusion was performed 2 h after the treatment. In the centrally projecting EWcp area, FOS immunohistochemistry was performed to assess neuronal activation. TRPA1, CART, and urocortin 1 mRNA expression as well as urocortin 1 and CART peptide content was semi-quantified by RNAscope in situ hybridization combined with immunofluorescence. Results: JT010 activated TRPA1 channels of the urocortinergic cells in acute brain slices. Alcohol treatment resulted in a significant FOS activation in both genotypes. Alcohol decreased the Trpa1 mRNA expression in WT mice. The assessment of urocortin 1 peptide immunoreactivity revealed lower basal urocortin 1 in KO mice compared to WTs. The urocortin 1 peptide content was affected genotype-dependently by alcohol: the peptide content decreased in WTs while it increased in KO mice. Alcohol exposure influenced neither CART and urocortin 1 mRNA expression nor the centrally projecting EWcp/CART peptide content. Conclusion: We proved the presence of functional TRPA1 receptors on urocortin 1 neurons of the centrally projecting EWcp. Decreased Trpa1 mRNA expression upon acute alcohol treatment, associated with reduced neuronal urocortin 1 peptide content suggesting that this cation channel may contribute to the regulation of the urocortin 1 release.
Collapse
Affiliation(s)
- Ammar Al-Omari
- Department of Pharmacology and Pharmacotherapy, Centre for Neuroscience, Szentágothai Research Centre, Medical School and Molecular Pharmacology Research Group, University of Pécs, Pécs, Hungary
| | - Miklós Kecskés
- Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, Centre for Neuroscience, Medical School and Research Group for Mood Disorders, University of Pécs, Pécs, Hungary
| | - Tünde Biró-Sütő
- Department of Pharmacology and Pharmacotherapy, Centre for Neuroscience, Szentágothai Research Centre, Medical School and Molecular Pharmacology Research Group, University of Pécs, Pécs, Hungary
| | - Balázs Fazekas
- Department of Pharmacology and Pharmacotherapy, Centre for Neuroscience, Szentágothai Research Centre, Medical School and Molecular Pharmacology Research Group, University of Pécs, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology, Medical School, University of Pécs, Pécs, Hungary
| | - Mónika Kuzma
- Department of Forensic Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Centre for Neuroscience, Szentágothai Research Centre, Medical School and Molecular Pharmacology Research Group, University of Pécs, Pécs, Hungary
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Centre for Neuroscience, Szentágothai Research Centre, Medical School and Molecular Pharmacology Research Group, University of Pécs, Pécs, Hungary
| |
Collapse
|
8
|
Becker-Krail DD, Ketchesin KD, Burns JN, Zong W, Hildebrand MA, DePoy LM, Vadnie CA, Tseng GC, Logan RW, Huang YH, McClung CA. Astrocyte Molecular Clock Function in the Nucleus Accumbens Is Important for Reward-Related Behavior. Biol Psychiatry 2022; 92:68-80. [PMID: 35461698 PMCID: PMC9232937 DOI: 10.1016/j.biopsych.2022.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Substance use disorders are associated with disruptions in circadian rhythms. Both human and animal work have shown the integral role for circadian clocks in the modulation of reward behaviors. Astrocytes have emerged as key regulators of circadian rhythmicity. However, no studies to date have identified the role of circadian astrocyte function in the nucleus accumbens (NAc), a hub for reward regulation, or determined the importance of these rhythms for reward-related behavior. METHODS Using astrocyte-specific RNA sequencing across time of day, we first characterized diurnal variation of the NAc astrocyte transcriptome. We then investigated the functional significance of this circadian regulation through viral-mediated disruption of molecular clock function in NAc astrocytes, followed by assessment of reward-related behaviors, metabolic-related molecular assays, and whole-cell electrophysiology in the NAc. RESULTS Strikingly, approximately 43% of the astrocyte transcriptome has a diurnal rhythm, and key metabolic pathways were enriched among the top rhythmic genes. Moreover, mice with a viral-mediated loss of molecular clock function in NAc astrocytes show a significant increase in locomotor response to novelty, exploratory drive, operant food self-administration, and motivation. At the molecular level, these animals also show disrupted metabolic gene expression, along with significant downregulation of both lactate and glutathione levels in the NAc. Loss of NAc astrocyte clock function also significantly altered glutamatergic signaling onto neighboring medium spiny neurons, alongside upregulated glutamate-related gene expression. CONCLUSIONS Taken together, these findings demonstrate a novel role for astrocyte circadian molecular clock function in the regulation of the NAc and reward-related behaviors.
Collapse
Affiliation(s)
- Darius D Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer N Burns
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wei Zong
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mariah A Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lauren M DePoy
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chelsea A Vadnie
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Yanhua H Huang
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
9
|
Becker-Krail DD, Walker WH, Nelson RJ. The Ventral Tegmental Area and Nucleus Accumbens as Circadian Oscillators: Implications for Drug Abuse and Substance Use Disorders. Front Physiol 2022; 13:886704. [PMID: 35574492 PMCID: PMC9094703 DOI: 10.3389/fphys.2022.886704] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
Circadian rhythms convergently evolved to allow for optimal synchronization of individuals’ physiological and behavioral processes with the Earth’s 24-h periodic cycling of environmental light and temperature. Whereas the suprachiasmatic nucleus (SCN) is considered the primary pacemaker of the mammalian circadian system, many extra-SCN oscillatory brain regions have been identified to not only exhibit sustainable rhythms in circadian molecular clock function, but also rhythms in overall region activity/function and mediated behaviors. In this review, we present the most recent evidence for the ventral tegmental area (VTA) and nucleus accumbens (NAc) to serve as extra-SCN oscillators and highlight studies that illustrate the functional significance of the VTA’s and NAc’s inherent circadian properties as they relate to reward-processing, drug abuse, and vulnerability to develop substance use disorders (SUDs).
Collapse
Affiliation(s)
- Darius D Becker-Krail
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
10
|
Rizk AA, Jenkins BW, Al-Sabagh Y, Hamidullah S, Reitz CJ, Rasouli M, Martino TA, Khokhar JY. The Impact of Sex, Circadian Disruption, and the ClockΔ19/Δ19 Genotype on Alcohol Drinking in Mice. Genes (Basel) 2022; 13:genes13040701. [PMID: 35456507 PMCID: PMC9031797 DOI: 10.3390/genes13040701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/25/2022] [Accepted: 04/06/2022] [Indexed: 01/05/2023] Open
Abstract
Shift work is associated with increased alcohol drinking, more so in males than females, and is thought to be a coping mechanism for disrupted sleep cycles. However, little is presently known about the causal influence of circadian rhythm disruptions on sex differences in alcohol consumption. In this study, we disrupted circadian rhythms in female and male mice using both environmental (i.e., shifting diurnal cycles) and genetic (i.e., ClockΔ19/Δ19 mutation) manipulations, and measured changes in alcohol consumption and preference using a two-bottle choice paradigm. Alcohol consumption and preference, as well as food and water consumption, total caloric intake, and weight were assessed in adult female and male ClockΔ19/Δ19 mutant mice or wild-type (WT) litter-mates, housed under a 12-hour:12-hour light:dark (L:D) cycle or a shortened 10-hour:10-hour L:D cycle. Female WT mice (under both light cycles) increased their alcohol consumption and preference over time, a pattern not observed in male WT mice. Compared to WT mice, ClockΔ19/Δ19 mice displayed increased alcohol consumption and preference. Sex differences were not apparent in ClockΔ19/Δ19 mice, with or without shifting diurnal cycles. In conclusion, sex differences in alcohol consumption patterns are evident and increase with prolonged access to alcohol. Disrupting circadian rhythms by mutating the Clock gene greatly increases alcohol consumption and abolishes sex differences present in WT animals.
Collapse
|
11
|
Becker-Krail DD, Parekh PK, Ketchesin KD, Yamaguchi S, Yoshino J, Hildebrand MA, Dunham B, Ganapathiraiu MK, Logan RW, McClung CA. Circadian transcription factor NPAS2 and the NAD + -dependent deacetylase SIRT1 interact in the mouse nucleus accumbens and regulate reward. Eur J Neurosci 2022; 55:675-693. [PMID: 35001440 PMCID: PMC9355311 DOI: 10.1111/ejn.15596] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 12/14/2021] [Accepted: 01/06/2022] [Indexed: 02/03/2023]
Abstract
Substance use disorders are associated with disruptions to both circadian rhythms and cellular metabolic state. At the molecular level, the circadian molecular clock and cellular metabolic state may be interconnected through interactions with the nicotinamide adenine dinucleotide (NAD+ )-dependent deacetylase, sirtuin 1 (SIRT1). In the nucleus accumbens (NAc), a region important for reward, both SIRT1 and the circadian transcription factor neuronal PAS domain protein 2 (NPAS2) are highly enriched, and both are regulated by the metabolic cofactor NAD+ . Substances of abuse, like cocaine, greatly disrupt cellular metabolism and promote oxidative stress; however, their effects on NAD+ in the brain remain unclear. Interestingly, cocaine also induces NAc expression of both NPAS2 and SIRT1, and both have independently been shown to regulate cocaine reward in mice. However, whether NPAS2 and SIRT1 interact in the NAc and/or whether together they regulate reward is unknown. Here, we demonstrate diurnal expression of Npas2, Sirt1 and NAD+ in the NAc, which is altered by cocaine-induced upregulation. Additionally, co-immunoprecipitation reveals NPAS2 and SIRT1 interact in the NAc, and cross-analysis of NPAS2 and SIRT1 chromatin immunoprecipitation sequencing reveals several reward-relevant and metabolic-related pathways enriched among shared gene targets. Notably, NAc-specific Npas2 knock-down or a functional Npas2 mutation in mice attenuates SIRT1-mediated increases in cocaine preference. Together, our data reveal an interaction between NPAS2 and SIRT1 in the NAc, which may serve to integrate cocaine's effects on circadian and metabolic factors, leading to regulation of drug reward.
Collapse
Affiliation(s)
- Darius D. Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Puja K. Parekh
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Shintaro Yamaguchi
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Yoshino
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mariah A. Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Brandon Dunham
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madhavi K. Ganapathiraiu
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA,Correspondence: Colleen A. McClung,
| |
Collapse
|
12
|
Lewis RG, Florio E, Punzo D, Borrelli E. The Brain's Reward System in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1344:57-69. [PMID: 34773226 DOI: 10.1007/978-3-030-81147-1_4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rhythmic gene expression is found throughout the central nervous system. This harmonized regulation can be dependent on- and independent of- the master regulator of biological clocks, the suprachiasmatic nucleus (SCN). Substantial oscillatory activity in the brain's reward system is regulated by dopamine. While light serves as a primary time-giver (zeitgeber) of physiological clocks and synchronizes biological rhythms in 24-h cycles, nonphotic stimuli have a profound influence over circadian biology. Indeed, reward-related activities (e.g., feeding, exercise, sex, substance use, and social interactions), which lead to an elevated level of dopamine, alters rhythms in the SCN and the brain's reward system. In this chapter, we will discuss the influence of the dopaminergic reward pathways on circadian system and the implication of this interplay on human health.
Collapse
Affiliation(s)
- Robert G Lewis
- School of Medicine, Department of Microbiology and Molecular Genetics, INSERMU1233, Center for Epigenetics and Metabolism, University of California - Irvine, Irvine, CA, USA
| | - Ermanno Florio
- School of Medicine, Department of Microbiology and Molecular Genetics, INSERMU1233, Center for Epigenetics and Metabolism, University of California - Irvine, Irvine, CA, USA
| | - Daniela Punzo
- School of Medicine, Department of Microbiology and Molecular Genetics, INSERMU1233, Center for Epigenetics and Metabolism, University of California - Irvine, Irvine, CA, USA
| | - Emiliana Borrelli
- School of Medicine, Department of Microbiology and Molecular Genetics, INSERMU1233, Center for Epigenetics and Metabolism, University of California - Irvine, Irvine, CA, USA. .,University of California - Irvine, Irvine, CA, USA.
| |
Collapse
|
13
|
Circadian Rhythms in Mood Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1344:153-168. [PMID: 34773231 DOI: 10.1007/978-3-030-81147-1_9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Altered behavioral rhythms are a fundamental diagnostic feature of mood disorders. Patients report worse subjective sleep and objective measures confirm this, implicating a role for circadian rhythm disruptions in mood disorder pathophysiology. Molecular clock gene mutations are associated with increased risk of mood disorder diagnosis and/or severity of symptoms, and mouse models of clock gene mutations have abnormal mood-related behaviors. The mechanism by which circadian rhythms contribute to mood disorders remains unknown, however, circadian rhythms regulate and are regulated by various biological systems that are abnormal in mood disorders and this interaction is theorized to be a key component of mood disorder pathophysiology. A growing body of evidence has begun defining how the interaction of circadian and neurotransmitter systems influences mood and behavior, including the role of current antidepressants and mood stabilizers. Additionally, the hypothalamus-pituitary-adrenal (HPA) axis interacts with both circadian and monoaminergic systems and may facilitate the contribution of environmental stressors to mood disorder pathophysiology. The central role of circadian rhythms in mood disorders has led to the development of chronotherapeutics, which are treatments designed specifically to target circadian rhythm regulators, such as sleep, light, and melatonin, to produce an antidepressant response.
Collapse
|
14
|
Saad L, Zwiller J, Kalsbeek A, Anglard P. Epigenetic Regulation of Circadian Clocks and Its Involvement in Drug Addiction. Genes (Basel) 2021; 12:1263. [PMID: 34440437 PMCID: PMC8394526 DOI: 10.3390/genes12081263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 12/19/2022] Open
Abstract
Based on studies describing an increased prevalence of addictive behaviours in several rare sleep disorders and shift workers, a relationship between circadian rhythms and addiction has been hinted for more than a decade. Although circadian rhythm alterations and molecular mechanisms associated with neuropsychiatric conditions are an area of active investigation, success is limited so far, and further investigations are required. Thus, even though compelling evidence connects the circadian clock to addictive behaviour and vice-versa, yet the functional mechanism behind this interaction remains largely unknown. At the molecular level, multiple mechanisms have been proposed to link the circadian timing system to addiction. The molecular mechanism of the circadian clock consists of a transcriptional/translational feedback system, with several regulatory loops, that are also intricately regulated at the epigenetic level. Interestingly, the epigenetic landscape shows profound changes in the addictive brain, with significant alterations in histone modification, DNA methylation, and small regulatory RNAs. The combination of these two observations raises the possibility that epigenetic regulation is a common plot linking the circadian clocks with addiction, though very little evidence has been reported to date. This review provides an elaborate overview of the circadian system and its involvement in addiction, and we hypothesise a possible connection at the epigenetic level that could further link them. Therefore, we think this review may further improve our understanding of the etiology or/and pathology of psychiatric disorders related to drug addiction.
Collapse
Affiliation(s)
- Lamis Saad
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands;
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Andries Kalsbeek
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands;
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Patrick Anglard
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- Institut National de la Santé et de la Recherche Médicale (INSERM), 75013 Paris, France
| |
Collapse
|
15
|
Logan RW, Ozburn AR, Arey RN, Ketchesin KD, Winquist A, Crain A, Tobe BTD, Becker-Krail D, Jarpe MB, Xue X, Zong W, Huo Z, Parekh PK, Zhu X, Fitzgerald E, Zhang H, Oliver-Smith J, DePoy LM, Hildebrand MA, Snyder EY, Tseng GC, McClung CA. Valproate reverses mania-like behaviors in mice via preferential targeting of HDAC2. Mol Psychiatry 2021; 26:4066-4084. [PMID: 33235333 PMCID: PMC8141541 DOI: 10.1038/s41380-020-00958-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/20/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
Valproate (VPA) has been used in the treatment of bipolar disorder since the 1990s. However, the therapeutic targets of VPA have remained elusive. Here we employ a preclinical model to identify the therapeutic targets of VPA. We find compounds that inhibit histone deacetylase proteins (HDACs) are effective in normalizing manic-like behavior, and that class I HDACs (e.g., HDAC1 and HDAC2) are most important in this response. Using an RNAi approach, we find that HDAC2, but not HDAC1, inhibition in the ventral tegmental area (VTA) is sufficient to normalize behavior. Furthermore, HDAC2 overexpression in the VTA prevents the actions of VPA. We used RNA sequencing in both mice and human induced pluripotent stem cells (iPSCs) derived from bipolar patients to further identify important molecular targets. Together, these studies identify HDAC2 and downstream targets for the development of novel therapeutics for bipolar mania.
Collapse
Affiliation(s)
- Ryan W. Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Angela R. Ozburn
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA.,VA Portland Health Care System, Portland, OR 97239, USA
| | - Rachel N. Arey
- Department of Molecular and Cellular Biology and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Alicia Winquist
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Andrew Crain
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Brian T. D. Tobe
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.,Department of Psychiatry, Veterans Administration Medical Center, La Jolla, CA 92037, USA
| | - Darius Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Matthew B. Jarpe
- Regenacy Pharmaceuticals, 303 Wyman St, Suite 300, Waltham, MA, 02451, USA
| | - Xiangning Xue
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Wei Zong
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, FL, 32611, USA
| | - Puja K. Parekh
- Brain and Mind Research Institute, Department of Psychiatry, and Sackler Institute for Developmental Psychobiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Xiyu Zhu
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Neuroscience, University of Pittsburgh, PA, 15260, USA
| | - Ethan Fitzgerald
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Hui Zhang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Peking Union Medical College Hospital, Beijing, China 100730
| | - Jeffrey Oliver-Smith
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Lauren M. DePoy
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Mariah A. Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Evan Y. Snyder
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.,Department of Pediatrics, University of California San Diego, La Jolla, CA, 92037, USA
| | - George C. Tseng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Corresponding Author: Colleen A. McClung, Ph.D., Department of Psychiatry, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, , 412-624-5547
| |
Collapse
|
16
|
Doyle MA, Bali V, Eagle AL, Stark AR, Fallon B, Neve RL, Robison AJ, Mazei-Robison MS. Serum- and glucocorticoid-inducible kinase 1 activity in ventral tegmental area dopamine neurons regulates cocaine conditioned place preference but not cocaine self-administration. Neuropsychopharmacology 2021; 46:1574-1583. [PMID: 34007042 PMCID: PMC8280171 DOI: 10.1038/s41386-021-01032-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023]
Abstract
Drugs of abuse regulate the activity of the mesolimbic dopamine (DA) system, and drug-induced changes in ventral tegmental area (VTA) cellular activity and gene regulation are linked to behavioral outputs associated with addiction. Previous work from our lab determined that VTA serum- and glucocorticoid-inducible kinase 1 (SGK1) transcription and catalytic activity were increased by repeated cocaine administration; however, it was unknown if these biochemical changes contributed to cocaine-elicited behaviors. Using transgenic and viral-mediated manipulations, we investigated the role of VTA SGK1 catalytic activity in regulating cocaine conditioned place preference and self-administration. We showed intra-VTA infusion of a catalytically inactive SGK1 mutant (K127Q) significantly decreased cocaine conditioned place preference (CPP). Further, we found that K127Q expression in VTA DA neurons significantly decreased cocaine CPP, while this same manipulation in VTA GABA neurons had no effect. However, blunted VTA DA SGK1 catalytic activity did not alter cocaine self-administration. Altogether, these studies identify the specific VTA cells critical for SGK1-mediated effects on cocaine CPP but not self-administration.
Collapse
Affiliation(s)
- Marie A. Doyle
- grid.17088.360000 0001 2150 1785Neuroscience Program, Michigan State University, East Lansing, USA
| | - Vedrana Bali
- grid.17088.360000 0001 2150 1785Department of Physiology, Michigan State University, East Lansing, USA
| | - Andrew L. Eagle
- grid.17088.360000 0001 2150 1785Department of Physiology, Michigan State University, East Lansing, USA
| | - Ali R. Stark
- grid.17088.360000 0001 2150 1785Neuroscience Program, Michigan State University, East Lansing, USA
| | - Barbara Fallon
- grid.17088.360000 0001 2150 1785Pharmacology and Toxicology Department, Michigan State University, East Lansing, USA
| | - Rachael L. Neve
- grid.32224.350000 0004 0386 9924Gene Technology Core, Massachusetts General Hospital, Boston, USA
| | - A. J. Robison
- grid.17088.360000 0001 2150 1785Neuroscience Program, Michigan State University, East Lansing, USA ,grid.17088.360000 0001 2150 1785Department of Physiology, Michigan State University, East Lansing, USA
| | - Michelle S. Mazei-Robison
- grid.17088.360000 0001 2150 1785Neuroscience Program, Michigan State University, East Lansing, USA ,grid.17088.360000 0001 2150 1785Department of Physiology, Michigan State University, East Lansing, USA
| |
Collapse
|
17
|
López AJ, Johnson AR, Kunnath AJ, Morris AD, Zachry JE, Thibeault KC, Kutlu MG, Siciliano CA, Calipari ES. An optimized procedure for robust volitional cocaine intake in mice. Exp Clin Psychopharmacol 2021; 29:319-333. [PMID: 32658535 PMCID: PMC7890946 DOI: 10.1037/pha0000399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Substance use disorder (SUD) is a behavioral disorder characterized by volitional drug consumption. Mouse models of SUD allow for the use of molecular, genetic, and circuit-level tools, providing enormous potential for defining the underlying mechanisms of this disorder. However, the relevance of results depends on the validity of the mouse models used. Self-administration models have long been the preferred preclinical model for SUD as they allow for volitional drug consumption, thus providing strong face validity. While previous work has defined the parameters that influence intravenous cocaine self-administration in other species-such as rats and primates-many of these parameters have not been explicitly assessed in mice. In a series of experiments, we showed that commonly used mouse models of self-administration, where behavior is maintained on a fixed-ratio schedule of reinforcement, show similar levels of responding in the presence and absence of drug delivery-demonstrating that it is impossible to determine when drug consumption is and is not volitional. To address these issues, we have developed a novel mouse self-administration procedure where animals do not need to be pretrained on sucrose and behavior is maintained on a variable-ratio schedule of reinforcement. This procedure increases rates of reinforcement behavior, increases levels of drug intake, and results in clearer delineation between drug-reinforced and saline conditions. Together, these data highlight a major issue with fixed-ratio models in mice that complicates subsequent analysis and provide a simple approach to minimize these confounds with variable-ratio schedules of reinforcement. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
- Alberto J López
- Department of Pharmacology, Vanderbilt University/Vanderbilt University School of Medicine
| | - Amy R Johnson
- Department of Pharmacology, Vanderbilt University/Vanderbilt University School of Medicine
| | - Ansley J Kunnath
- Vanderbilt University Medical Scientists Training Program, Vanderbilt University/Vanderbilt University School of Medicine
| | - Allison D Morris
- Department of Pharmacology, Vanderbilt University/Vanderbilt University School of Medicine
| | - Jennifer E Zachry
- Department of Pharmacology, Vanderbilt University/Vanderbilt University School of Medicine
| | - Kimberly C Thibeault
- Department of Pharmacology, Vanderbilt University/Vanderbilt University School of Medicine
| | - Munir G Kutlu
- Department of Pharmacology, Vanderbilt University/Vanderbilt University School of Medicine
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University/Vanderbilt University School of Medicine
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University/Vanderbilt University School of Medicine
| |
Collapse
|
18
|
Circadian-Dependent and Sex-Dependent Increases in Intravenous Cocaine Self-Administration in Npas2 Mutant Mice. J Neurosci 2021; 41:1046-1058. [PMID: 33268545 DOI: 10.1523/jneurosci.1830-20.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 11/21/2022] Open
Abstract
Substance use disorder (SUD) is associated with disruptions in circadian rhythms. The circadian transcription factor neuronal PAS domain protein 2 (NPAS2) is enriched in reward-related brain regions and regulates reward, but its role in SU is unclear. To examine the role of NPAS2 in drug taking, we measured intravenous cocaine self-administration (acquisition, dose-response, progressive ratio, extinction, cue-induced reinstatement) in wild-type (WT) and Npas2 mutant mice at different times of day. In the light (inactive) phase, cocaine self-administration, reinforcement, motivation and extinction responding were increased in all Npas2 mutants. Sex differences emerged during the dark (active) phase with Npas2 mutation increasing self-administration, extinction responding, and reinstatement only in females as well as reinforcement and motivation in males and females. To determine whether circulating hormones are driving these sex differences, we ovariectomized WT and Npas2 mutant females and confirmed that unlike sham controls, ovariectomized mutant mice showed no increase in self-administration. To identify whether striatal brain regions are activated in Npas2 mutant females, we measured cocaine-induced ΔFosB expression. Relative to WT, ΔFosB expression was increased in D1+ neurons in the nucleus accumbens (NAc) core and dorsolateral (DLS) striatum in Npas2 mutant females after dark phase self-administration. We also identified potential target genes that may underlie the behavioral responses to cocaine in Npas2 mutant females. These results suggest NPAS2 regulates reward and activity in specific striatal regions in a sex and time of day (TOD)-specific manner. Striatal activation could be augmented by circulating sex hormones, leading to an increased effect of Npas2 mutation in females.SIGNIFICANCE STATEMENT Circadian disruptions are a common symptom of substance use disorders (SUDs) and chronic exposure to drugs of abuse alters circadian rhythms, which may contribute to subsequent SU. Diurnal rhythms are commonly found in behavioral responses to drugs of abuse with drug sensitivity and motivation peaking during the dark (active) phase in nocturnal rodents. Emerging evidence links disrupted circadian genes to SU vulnerability and drug-induced alterations to these genes may augment drug-seeking. The circadian transcription factor neuronal PAS domain protein 2 (NPAS2) is enriched in reward-related brain regions and regulates reward, but its role in SU is unclear. To examine the role of NPAS2 in drug taking, we measured intravenous cocaine self-administration in wild-type (WT) and Npas2 mutant mice at different times of day.
Collapse
|
19
|
Tamura EK, Oliveira-Silva KS, Ferreira-Moraes FA, Marinho EAV, Guerrero-Vargas NN. Circadian rhythms and substance use disorders: A bidirectional relationship. Pharmacol Biochem Behav 2021; 201:173105. [PMID: 33444601 DOI: 10.1016/j.pbb.2021.173105] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 01/23/2023]
Abstract
The circadian system organizes circadian rhythms (biological cycles that occur around 24 h) that couple environmental cues (zeitgebers) with internal functions of the organism. The misalignment between circadian rhythms and external cues is known as chronodisruption and contributes to the development of mental, metabolic and other disorders, including cancer, cardiovascular diseases and addictive disorders. Drug addiction represents a global public health concern and affects the health and well-being of individuals, families and communities. In this manuscript, we reviewed evidence indicating a bidirectional relationship between the circadian system and the development of addictive disorders. We provide information on the interaction between the circadian system and drug addiction for each drug or drug class (alcohol, cannabis, hallucinogens, psychostimulants and opioids). We also describe evidence showing that drug use follows a circadian pattern, which changes with the progression of addiction. Furthermore, clock gene expression is also altered during the development of drug addiction in many brain areas related to drug reward, drug seeking and relapse. The regulation of the glutamatergic and dopaminergic neurocircuitry by clock genes is postulated to be the main circadian mechanism underlying the escalation of drug addiction. The bidirectional interaction between the circadian system and drug addiction seems to be mediated by the effects caused by each drug or class of drugs of abuse. These studies provide new insights on the development of successful strategies aimed at restoring/stabilizing circadian rhythms to reduce the risk for addiction development and relapse.
Collapse
Affiliation(s)
- Eduardo K Tamura
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil.
| | - Kallyane S Oliveira-Silva
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Felipe A Ferreira-Moraes
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Eduardo A V Marinho
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Natalí N Guerrero-Vargas
- Department of Anatomy, Faculty of Medicine, Universidad Nacional Autonóma de México, Av Universidad 3000, Ciudad Universitaria, México City 04510, Mexico
| |
Collapse
|
20
|
Liu S, Wei J, Ni R, Gao T, Ni P, Zhao L, Duan X, Ma X, Sham PC, Li T. Valproate Reverses Mania-Like Behavior of Clock delta19 Mouse and Alters Monoamine Neurotransmitters Metabolism in the Hippocampus. Neuropsychiatr Dis Treat 2021; 17:471-480. [PMID: 33603383 PMCID: PMC7884953 DOI: 10.2147/ndt.s293482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/18/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Mice with a deletion at exon 19 of the circadian locomotor output cycles Kaput gene (Clock delta19) exhibit mania-like behavior and have been one of the most common animal models for bipolar disorder (BD). The predictive validity of the Clock delta19 was investigated via studies using lithium previously. Determination of effects of other mood stabilizers on Clock delta19 mouse would be helpful for better understanding of the mechanism underlined. METHODS Wildtype (WT) and Clock delta19 mice were treated with saline (n = 10 for WT and n=10 for Clock delta19) or valproate (VPA) (n = 10 for WT and n=10 for Clock delta19) for 10 days. The hyperactivity, anxiety-like behaviors and depression-like behaviors were tested. The concentration of monoamine neurotransmitters and their metabolites in the hippocampus of saline or VPA treated WT and Clock delta19 mouse (n = 8 for each) were also determined. RESULTS VPA can reverse hyperactivity, lower level of anxiety-like and depression-like behaviors of the Clock delta19 mouse. Clock delta19 mouse exhibited lower levels of serotonin (5-HT) and dopamine (DA) in right hippocampus compared to WT mouse. Chronic VPA treatment did not affect the levels of 5-HT and DA, but can reduce the level of levodopa (L-DOPA) in the right hippocampus of Clock delta19 mouse. CONCLUSION Our results indicated that chronic VPA treatment can reverse the mania-like behaviors of the Clock delta19 mouse and further consolidate the validity of the Clock delta19 mouse as a model of BD. Monoamine neurotransmitters and their metabolites in the hippocampus are partly regulated by mutation of the Clock gene or VPA treatment.
Collapse
Affiliation(s)
- Shasha Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| | - Jinxue Wei
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Rongjun Ni
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Tianhao Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| | - Peiyan Ni
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Liansheng Zhao
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Xiyu Duan
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Xiaohong Ma
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Pokfulam, Hong Kong, People's Republic of China.,State Key Laboratory for Cognitive and Brain Sciences, The University of Hong Kong, Pokfulam, Hong Kong, People's Republic of China
| | - Tao Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People's Republic of China.,Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China.,Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, People's Republic of China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, People's Republic of China
| |
Collapse
|
21
|
von Schantz M, Leocadio-Miguel MA, McCarthy MJ, Papiol S, Landgraf D. Genomic perspectives on the circadian clock hypothesis of psychiatric disorders. ADVANCES IN GENETICS 2020; 107:153-191. [PMID: 33641746 DOI: 10.1016/bs.adgen.2020.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Circadian rhythm disturbances are frequently described in psychiatric disorders such as major depressive disorder, bipolar disorder, and schizophrenia. Growing evidence suggests a biological connection between mental health and circadian rhythmicity, including the circadian influence on brain function and mood and the requirement for circadian entrainment by external factors, which is often impaired in mental illness. Mental (as well as physical) health is also adversely affected by circadian misalignment. The marked interindividual differences in this combined susceptibility, in addition to the phenotypic spectrum in traits related both to circadian rhythms and mental health, suggested the possibility of a shared genetic background and that circadian clock genes may also be candidate genes for psychiatric disorders. This hypothesis was further strengthened by observations in animal models where clock genes had been knocked out or mutated. The introduction of genome-wide association studies (GWAS) enabled hypothesis-free testing. GWAS analysis of chronotype confirmed the prominent role of circadian genes in these phenotypes and their extensive polygenicity. However, in GWAS on psychiatric traits, only one clock gene, ARNTL (BMAL1) was identified as one of the few loci differentiating bipolar disorder from schizophrenia, and macaque monkeys where the ARNTL gene has been knocked out display symptoms similar to schizophrenia. Another lesson from genomic analyses is that chronotype has an important genetic correlation with several psychiatric disorders and that this effect is unidirectional. We conclude that the effect of circadian disturbances on psychiatric disorders probably relates to modulation of rhythm parameters and extend beyond the core clock genes themselves.
Collapse
Affiliation(s)
- Malcolm von Schantz
- Faculty of Health and Medical Sciences, University of Surrey, Surrey, United Kingdom; Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | - Mario A Leocadio-Miguel
- Faculty of Health and Medical Sciences, University of Surrey, Surrey, United Kingdom; Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Michael J McCarthy
- Department of Psychiatry, University of California San Diego, San Diego, CA, United States
| | - Sergi Papiol
- Department of Psychiatry, University Hospital, Munich, Germany; Institute of Psychiatric Phenomics and Genomics (IPPG), Munich, Germany
| | - Dominic Landgraf
- Circadian Biology Group, Department of Molecular Neurobiology, Clinic of Psychiatry and Psychotherapy, University Hospital, Munich, Germany
| |
Collapse
|
22
|
Hühne A, Volkmann P, Stephan M, Rossner M, Landgraf D. An in-depth neurobehavioral characterization shows anxiety-like traits, impaired habituation behavior, and restlessness in male Cryptochrome-deficient mice. GENES, BRAIN, AND BEHAVIOR 2020; 19:e12661. [PMID: 32348614 DOI: 10.1111/gbb.12661] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022]
Abstract
Many psychiatric disorders, for example, anxiety, are accompanied by disturbances of circadian rhythms, including disturbed sleep/wake cycles, changes in locomotor activity, and abnormal endocrine function. Conversely, alternations of circadian rhythms are a risk factor for the development of psychiatric disorders. This assumption is supported by animals with clock gene mutations which often display behaviors that resemble human psychiatric disorders. In this study, we performed an in-depth behavioral analysis with male mice lacking the central clock genes Cryptochrome 1 and 2 (Cry1/2-/- ), which are thus unable to express endogenous circadian rhythms. With wild-type and Cry1/2-/- mice, we performed an extensive behavioral analysis to study their cognitive abilities, social behavior, and their expression of depression-like and anxiety-like behavior. While Cry1/2-/- mice showed only mild abnormalities at cognitive and social behavioral levels, they were consistently more anxious than wildtype mice. Anxiety-like behavior was particularly evident in reduced mobility in new environments, altered ability to habituate, compensatory behavior, and consistent restless behavior across many behavioral tests. In line with their anxiety-like behavioral phenotype, Cry1/2-/- mice have higher c-Fos activity in the amygdala after exposure to an anxiogenic stressor than wild-type mice. In our study, we identified Cry1/2-/- mice as animals that qualify as a translational mouse model for anxiety disorder in humans because of its consistent behavior of restlessness, increased immobility, and dysfunctional habituation in new environments.
Collapse
Affiliation(s)
- Anisja Hühne
- Circadian Biology Group, Department of Molecular Neurobiology, Clinic of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
- Munich Medical Research School, Ludwig Maximilian University, Munich, Germany
| | - Paul Volkmann
- Department of Molecular Neurobiology, Clinic of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - Marius Stephan
- Department of Molecular Neurobiology, Clinic of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Moritz Rossner
- Department of Molecular Neurobiology, Clinic of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - Dominic Landgraf
- Circadian Biology Group, Department of Molecular Neurobiology, Clinic of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
23
|
Characterization of genetically complex Collaborative Cross mouse strains that model divergent locomotor activating and reinforcing properties of cocaine. Psychopharmacology (Berl) 2020; 237:979-996. [PMID: 31897574 PMCID: PMC7542678 DOI: 10.1007/s00213-019-05429-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/09/2019] [Indexed: 12/25/2022]
Abstract
RATIONALE Few effective treatments exist for cocaine use disorders due to gaps in knowledge about its complex etiology. Genetically defined animal models provide a useful tool for advancing our understanding of the biological and genetic underpinnings of addiction-related behavior and evaluating potential treatments. However, many attempts at developing mouse models of behavioral disorders were based on overly simplified single gene perturbations, often leading to inconsistent and misleading results in pre-clinical pharmacology studies. A genetically complex mouse model may better reflect disease-related behaviors. OBJECTIVES Screening defined, yet genetically complex, intercrosses of the Collaborative Cross (CC) mice revealed two lines, RIX04/17 and RIX41/51, with extreme high and low behavioral responses to cocaine. We characterized these lines as well as their CC parents, CC004/TauUnc and CC041/TauUnc, to evaluate their utility as novel model systems for studying the biological and genetic mechanisms underlying behavioral responses to cocaine. METHODS Behavioral responses to acute (initial locomotor sensitivity) and repeated (behavioral sensitization, conditioned place preference, intravenous self-administration) exposures to cocaine were assessed. We also examined the monoaminergic system (striatal tissue content and in vivo fast scan cyclic voltammetry), HPA axis reactivity, and circadian rhythms as potential mechanisms for the divergent phenotypic behaviors observed in the two strains, as these systems have a previously known role in mediating addiction-related behaviors. RESULTS RIX04/17 and 41/51 show strikingly divergent initial locomotor sensitivity to cocaine with RIX04/17 exhibiting very high and RIX41/51 almost no response. The lines also differ in the emergence of behavioral sensitization with RIX41/51 requiring more exposures to exhibit a sensitized response. Both lines show conditioned place preference for cocaine. We determined that the cocaine sensitivity phenotype in each RIX line was largely driven by the genetic influence of one CC parental strain, CC004/TauUnc and CC041/TauUnc. CC004 demonstrates active operant cocaine self-administration and CC041 is unable to acquire under the same testing conditions, a deficit which is specific to cocaine as both strains show operant response for a natural food reward. Examination of potential mechanisms driving differential responses to cocaine show strain differences in molecular and behavioral circadian rhythms. Additionally, while there is no difference in striatal dopamine tissue content or dynamics, there are selective differences in striatal norepinephrine and serotonergic tissue content. CONCLUSIONS These CC strains offer a complex polygenic model system to study underlying mechanisms of cocaine response. We propose that CC041/TauUnc and CC004/TauUnc will be useful for studying genetic and biological mechanisms underlying resistance or vulnerability to the stimulatory and reinforcing effects of cocaine.
Collapse
|
24
|
Liu L, Luo T, Dong H, Zhang C, Liu T, Zhang X, Hao W. Genome-Wide DNA Methylation Analysis in Male Methamphetamine Users With Different Addiction Qualities. Front Psychiatry 2020; 11:588229. [PMID: 33192735 PMCID: PMC7645035 DOI: 10.3389/fpsyt.2020.588229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/24/2020] [Indexed: 12/20/2022] Open
Abstract
This paper aimed to explore the genome-wide DNA methylation status of methamphetamine (MA) abusers with different qualities to addiction and to identify differentially methylated candidate genes. A total of 207 male MA abusers with an MA abuse frequency of ≥10 times and an MA abuse duration of ≥1 year were assigned to the high MA addiction quality group (HMAQ group; 168 subjects who met the diagnostic criteria for MA dependence according to the DSM-IV) or to the low MA addictive quality group (LMAQ group; 39 subjects who did not meet the criteria for MA dependence). In addition 105 healthy controls were recruited. Eight HMAQ subjects, eight LMAQ subjects, and eight healthy controls underwent genome-wide DNA methylation scans with an Infinium Human Methylation 450 array (Illumina). The differentially methylated region (DMR) data were entered into pathway analysis, and the differentially methylated position (DMP) data were screened for candidate genes and verified by MethyLight qPCR with all samples. Seven specific pathways with an abnormal methylation status were identified, including the circadian entrainment, cholinergic synapse, glutamatergic synapse, retrograde endocannabinoid signaling, GABAergic synapse, morphine addiction and PI3K-Akt signaling pathways. SLC1A6, BHLHB9, LYNX1, CAV2, and PCSK9 showed differences in their methylation levels in the three groups. Only the number of methylated copies of CAV2 was significantly higher in the LMAQ group than in the HMAQ group. Our findings suggest that the circadian entrainment pathway and the caveolin-2 gene may play key roles in MA addiction quality. Further studies on their functions and mechanisms will help us to better understand the pathogenesis of MA addiction and to explore new targets for drug intervention.
Collapse
Affiliation(s)
- Liang Liu
- Department of Geriatric Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| | - Tao Luo
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China.,Department of Clinic Psychiatry, Jiangxi Mental Hospital, Nanchang University, Nanchang, China
| | - Huixi Dong
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| | - Chenxi Zhang
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| | - Tieqiao Liu
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| | - Xiangyang Zhang
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Wei Hao
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| |
Collapse
|
25
|
Barbosa-Méndez S, Salazar-Juárez A. Melatonin decreases cocaine-induced locomotor activity in pinealectomized rats. ACTA ACUST UNITED AC 2019; 42:295-308. [PMID: 31859790 PMCID: PMC7236171 DOI: 10.1590/1516-4446-2018-0400] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 09/13/2019] [Indexed: 12/22/2022]
Abstract
Objective: Several studies have shown that the time of day regulates the reinforcing effects of cocaine. Additionally, melatonin and its MT1 and MT2 receptors have been found to participate in modulation of the reinforcing effects of such addictive drugs as cocaine. Loss of the diurnal variation in cocaine-induced locomotor sensitization and cocaine-induced place preference has been identified in pinealectomized mice. In addition, several studies in rodents have shown that administration of melatonin decreased the reinforcing effects of cocaine. The objective of this study was to evaluate the effect of melatonin on cocaine-induced locomotor activity in pinealectomized rats at different times of day (zeitgeber time [ZT]4, ZT10, ZT16, and ZT22). Methods: Naïve, pinealectomized Wistar rats received cocaine at different times of day. Melatonin was administered 30 min before cocaine; luzindole was administered 15 min prior to melatonin and 45 min before cocaine. After administration of each treatment, locomotor activity for each animal was recorded for a total of 30 min. Pinealectomy was confirmed at the end of the experiment through melatonin quantitation by ELISA. Results: Cocaine-induced locomotor activity varied according to the time of day. Continuous lighting and pinealectomy increased cocaine-induced locomotor activity. Melatonin administration decreased cocaine-induced locomotor activity in naïve and pinealectomized rats at different times of day. Luzindole blocked the melatonin-induced reduction in cocaine-induced locomotor activity in pinealectomized rats. Conclusion: Given its ability to mitigate various reinforcing effects of cocaine, melatonin could be a useful therapy for cocaine abuse.
Collapse
Affiliation(s)
- Susana Barbosa-Méndez
- Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría, Ciudad de México, Mexico
| | - Alberto Salazar-Juárez
- Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría, Ciudad de México, Mexico
| |
Collapse
|
26
|
Ahrens AM, Ahmed OJ. Neural circuits linking sleep and addiction: Animal models to understand why select individuals are more vulnerable to substance use disorders after sleep deprivation. Neurosci Biobehav Rev 2019; 108:435-444. [PMID: 31756346 DOI: 10.1016/j.neubiorev.2019.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 10/26/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
Abstract
Individuals differ widely in their drug-craving behaviors. One reason for these differences involves sleep. Sleep disturbances lead to an increased risk of substance use disorders and relapse in only some individuals. While animal studies have examined the impact of sleep on reward circuitry, few have addressed the role of individual differences in the effects of altered sleep. There does, however, exist a rodent model of individual differences in reward-seeking behavior: the sign/goal-tracker model of Pavlovian conditioned approach. In this model, only some rats show the key behavioral traits associated with addiction, including impulsivity and poor attentional control, making this an ideal model system to examine individually distinct sleep-reward interactions. Here, we describe how the limbic neural circuits responsible for individual differences in incentive motivation overlap with those involved in sleep-wake regulation, and how this model can elucidate the common underlying mechanisms. Consideration of individual differences in preclinical models would improve our understanding of how sleep interacts with motivational systems, and why sleep deprivation contributes to addiction in only select individuals.
Collapse
Affiliation(s)
| | - Omar J Ahmed
- Dept. of Psychology, United States; Neuroscience Graduate Program, United States; Michigan Center for Integrative Research in Critical Care, United States; Kresge Hearing Research Institute, United States; Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
27
|
Logan RW, Parekh PK, Kaplan G, Becker-Krail D, Williams W, Yamaguchi S, Yoshino J, Shelton MA, Zhu X, Zhang H, Waplinger S, Fitzgerald E, Oliver-Smith J, Sundarvelu P, Enwright JF, Huang YH, McClung CA. NAD+ cellular redox and SIRT1 regulate the diurnal rhythms of tyrosine hydroxylase and conditioned cocaine reward. Mol Psychiatry 2019; 24:1668-1684. [PMID: 29728703 PMCID: PMC6215755 DOI: 10.1038/s41380-018-0061-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 01/12/2018] [Accepted: 02/19/2018] [Indexed: 12/21/2022]
Abstract
The diurnal regulation of dopamine is important for normal physiology and diseases such as addiction. Here we find a novel role for the CLOCK protein to antagonize CREB-mediated transcriptional activity at the tyrosine hydroxylase (TH) promoter, which is mediated by the interaction with the metabolic sensing protein, Sirtuin 1 (SIRT1). Additionally, we demonstrate that the transcriptional activity of TH is modulated by the cellular redox state, and daily rhythms of redox balance in the ventral tegmental area (VTA), along with TH transcription, are highly disrupted following chronic cocaine administration. Furthermore, CLOCK and SIRT1 are important for regulating cocaine reward and dopaminergic (DAergic) activity, with interesting differences depending on whether DAergic activity is in a heightened state and if there is a functional CLOCK protein. Taken together, we find that rhythms in cellular metabolism and circadian proteins work together to regulate dopamine synthesis and the reward value for drugs of abuse.
Collapse
Affiliation(s)
- Ryan W. Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA,Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609
| | - Puja K. Parekh
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Gabrielle Kaplan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Darius Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Wilbur Williams
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Shintaro Yamaguchi
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609
| | - Jun Yoshino
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609
| | - Micah A. Shelton
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Xiyu Zhu
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Hui Zhang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,School of Medicine, Peking Union Medical College, Tsinghua University, Beijing, China
| | - Spencer Waplinger
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Ethan Fitzgerald
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Jeffrey Oliver-Smith
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - Poornima Sundarvelu
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | - John F. Enwright
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA
| | | | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, 15219, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA,Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609,Correspondence: (C.A.M.)
| |
Collapse
|
28
|
Lafaye G, Desterke C, Marulaz L, Benyamina A. Cannabidiol affects circadian clock core complex and its regulation in microglia cells. Addict Biol 2019; 24:921-934. [PMID: 30307084 DOI: 10.1111/adb.12660] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 12/27/2022]
Abstract
Cannabis is often used by consumers for sleep disorders. Studies show that circadian rhythm could be affected by a misuse of cannabis. Recent research has connected the role of microglial cells with psychiatric disorders such as substance abuse. The aim was to show the effect of two major components of cannabis on circadian genes regulation in microglial cells. In BV-2 microglial cells, cannabidiol (CBD) induces a deregulation of circadian genes with (P-value = 0.039) or without (P-value = 0.0015) lipopolisaccharides stimulation. CBD up regulated Arntl (P = 9.72E-5) and down regulated Clock (P = 0.0034) in BV-2 cells. Temporal expression of Arntl (light and dark P = 0.0054) and Clock (light and dark P = 0.047) was confirmed to have 24 hours light and dark rhythmic regulation in dissected suprachiasmatic nucleus as well as of Cb1 cannabinoid receptor (light and dark P = 0.019). In BV-2 microglia cells, CBD also up regulated CRY2 (P = 0.0473) and PER1 (P = 0.0131). Other nuclear molecules show a deregulation of circadian rhythm in microglial cells by CBD, such as RORA, RevErbα, RORB, CREBBP, AFT4, AFT5 and NFIL3. Our study suggests that circadian rhythm in microglial cells is deregulated by CBD but not by THC. It is consistent with clinical observations of the use of therapeutic cannabis to treat insomnia.
Collapse
Affiliation(s)
- Geneviève Lafaye
- Dpt Addictologie, AP-HP, GH Paris-Sud, Hôpital Paul Brousse, Villejuif, France
- INSERM U1178, Villejuif, France
| | | | - Laurent Marulaz
- Dpt Addictologie, AP-HP, GH Paris-Sud, Hôpital Paul Brousse, Villejuif, France
- INSERM U1178, Villejuif, France
| | - Amine Benyamina
- Dpt Addictologie, AP-HP, GH Paris-Sud, Hôpital Paul Brousse, Villejuif, France
- INSERM U1178, Villejuif, France
| |
Collapse
|
29
|
Parekh PK, Logan RW, Ketchesin KD, Becker-Krail D, Shelton MA, Hildebrand MA, Barko K, Huang YH, McClung CA. Cell-Type-Specific Regulation of Nucleus Accumbens Synaptic Plasticity and Cocaine Reward Sensitivity by the Circadian Protein, NPAS2. J Neurosci 2019; 39:4657-4667. [PMID: 30962277 PMCID: PMC6561687 DOI: 10.1523/jneurosci.2233-18.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 03/16/2019] [Accepted: 04/03/2019] [Indexed: 12/23/2022] Open
Abstract
The circadian transcription factor neuronal PAS domain 2 (NPAS2) is linked to psychiatric disorders associated with altered reward sensitivity. The expression of Npas2 is preferentially enriched in the mammalian forebrain, including the nucleus accumbens (NAc), a major neural substrate of motivated and reward behavior. Previously, we demonstrated that downregulation of NPAS2 in the NAc reduces the conditioned behavioral response to cocaine in mice. We also showed that Npas2 is preferentially enriched in dopamine receptor 1 containing medium spiny neurons (D1R-MSNs) of the striatum. To extend these studies, we investigated the impact of NPAS2 disruption on accumbal excitatory synaptic transmission and strength, along with the behavioral sensitivity to cocaine reward in a cell-type-specific manner. Viral-mediated knockdown of Npas2 in the NAc of male and female C57BL/6J mice increased the excitatory drive onto MSNs. Using Drd1a-tdTomato mice in combination with viral knockdown, we determined these synaptic adaptations were specific to D1R-MSNs relative to non-D1R-MSNs. Interestingly, NAc-specific knockdown of Npas2 blocked cocaine-induced enhancement of synaptic strength and glutamatergic transmission specifically onto D1R-MSNs. Last, we designed, validated, and used a novel Cre-inducible short-hairpin RNA virus for MSN-subtype-specific knockdown of Npas2 Cell-type-specific Npas2 knockdown in D1R-MSNs, but not D2R-MSNs, in the NAc reduced cocaine conditioned place preference. Together, our results demonstrate that NPAS2 regulates excitatory synapses of D1R-MSNs in the NAc and cocaine reward-related behavior.SIGNIFICANCE STATEMENT Drug addiction is a widespread public health concern often comorbid with other psychiatric disorders. Disruptions of the circadian clock can predispose or exacerbate substance abuse in vulnerable individuals. We demonstrate a role for the core circadian protein, NPAS2, in mediating glutamatergic neurotransmission at medium spiny neurons (MSNs) in the nucleus accumbens (NAc), a region critical for reward processing. We find that NPAS2 negatively regulates functional excitatory synaptic plasticity in the NAc and is necessary for cocaine-induced plastic changes in MSNs expressing the dopamine 1 receptor (D1R). We further demonstrate disruption of NPAS2 in D1R-MSNs produces augmented cocaine preference. These findings highlight the significance of cell-type-specificity in mechanisms underlying reward regulation by NPAS2 and extend our knowledge of its function.
Collapse
Affiliation(s)
- Puja K Parekh
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
- Center for Systems Genetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine 04609
| | - Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Darius Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Micah A Shelton
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Mariah A Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Kelly Barko
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Yanhua H Huang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219, and
- Center for Systems Genetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine 04609
| |
Collapse
|
30
|
Wang DQ, Wang XL, Wang CY, Wang Y, Li SX, Liu KZ. Effects of chronic cocaine exposure on the circadian rhythmic expression of the clock genes in reward-related brain areas in rats. Behav Brain Res 2019; 363:61-69. [DOI: 10.1016/j.bbr.2019.01.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/09/2019] [Accepted: 01/22/2019] [Indexed: 12/15/2022]
|
31
|
Abstract
Many processes in the human body - including brain function - are regulated over the 24-hour cycle, and there are strong associations between disrupted circadian rhythms (for example, sleep-wake cycles) and disorders of the CNS. Brain disorders such as autism, depression and Parkinson disease typically develop at certain stages of life, and circadian rhythms are important during each stage of life for the regulation of processes that may influence the development of these disorders. Here, we describe circadian disruptions observed in various brain disorders throughout the human lifespan and highlight emerging evidence suggesting these disruptions affect the brain. Currently, much of the evidence linking brain disorders and circadian dysfunction is correlational, and so whether and what kind of causal relationships might exist are unclear. We therefore identify remaining questions that may direct future research towards a better understanding of the links between circadian disruption and CNS disorders.
Collapse
Affiliation(s)
- Ryan W Logan
- University of Pittsburgh School of Medicine, Department of Psychiatry, Pittsburgh, PA, USA
| | - Colleen A McClung
- University of Pittsburgh School of Medicine, Department of Psychiatry, Pittsburgh, PA, USA.
| |
Collapse
|
32
|
Parekh PK, Becker-Krail D, Sundaravelu P, Ishigaki S, Okado H, Sobue G, Huang Y, McClung CA. Altered GluA1 (Gria1) Function and Accumbal Synaptic Plasticity in the ClockΔ19 Model of Bipolar Mania. Biol Psychiatry 2018; 84:817-826. [PMID: 28780133 PMCID: PMC5745309 DOI: 10.1016/j.biopsych.2017.06.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Disruptions in circadian rhythms are associated with an increased risk for bipolar disorder. Moreover, studies show that the circadian protein CLOCK (circadian locomotor output cycles kaput) is involved in regulating monoaminergic systems and mood-related behavior. However, the molecular and synaptic mechanisms underlying this relationship remain poorly understood. METHODS Using ex vivo whole-cell patch-clamp electrophysiology in ClockΔ19 mutant and wild-type mice we characterized alterations in excitatory synaptic transmission, strength, and intrinsic excitability of nucleus accumbens (NAc) neurons. We performed protein crosslinking and Western blot analysis to examine surface and intracellular levels and rhythm of the glutamate receptor subunit, GluA1, in the NAc. Viral-mediated overexpression of Gria1 in the NAc and behavioral assays were also used. RESULTS Compared with wild-type mice, ClockΔ19 mice display reduced alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor-mediated excitatory synaptic responses at NAc medium spiny neurons. These alterations are likely postsynaptic, as presynaptic release of glutamate onto medium spiny neurons is unaltered in mutant mice. Additionally, NAc surface protein levels and the rhythm of GRIA1 are decreased in ClockΔ19 mice diurnally, consistent with reduced functional synaptic response. Furthermore, we observed a significantly hyperpolarized resting membrane potential of ClockΔ19 medium spiny neurons, suggesting lowered intrinsic excitability. Last, overexpression of functional Gria1 in the NAc of mutant mice was able to normalize increased exploratory drive and reward sensitivity behavior when mice are in a manic-like state. CONCLUSIONS Together, our findings demonstrate that NAc excitatory signaling via Gria1 expression is integral to the effects of Clock gene disruption on manic-like behaviors.
Collapse
Affiliation(s)
- Puja K. Parekh
- University of Pittsburgh Medical Center, Department of Psychiatry, Pittsburgh, Pennsylvania 15219
| | - Darius Becker-Krail
- University of Pittsburgh Medical Center, Department of Psychiatry, Pittsburgh, Pennsylvania 15219
| | - Poornima Sundaravelu
- University of Pittsburgh Medical Center, Department of Psychiatry, Pittsburgh, Pennsylvania 15219
| | - Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Haruo Okado
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yanhua Huang
- University of Pittsburgh Medical Center, Department of Psychiatry, Pittsburgh, Pennsylvania 15219
| | - Colleen A. McClung
- University of Pittsburgh Medical Center, Department of Psychiatry, Pittsburgh, Pennsylvania 15219
| |
Collapse
|
33
|
Abstract
Disruption of circadian clocks is strongly associated with mood disorders. Chronotherapies targeting circadian rhythms have been shown to be very effective treatments of mood disorders, but still are not widely used in clinical practice. The mechanisms by which circadian disruption leads to mood disorders are poorly characterized and, therefore, may not convince clinicians to apply chronotherapies. Hence, in this review, we describe specific potential mechanisms, in order to make this connection more credible to clinicians. We believe that four major features of disrupted clocks may contribute to the development of mood disorders: (1) loss of synchronization to environmental 24-h rhythms, (2) internal desynchronization among body clocks, (3) low rhythm amplitude, and (4) changes in sleep architecture. Discussing these attributes and giving plausible examples, we will discuss prospects for relatively simple chronotherapies addressing these features that are easy to implement in clinical practice. Key messages In this review, we describe specific potential mechanisms by which disrupted clocks may contribute to the development of mood disorders: (1) loss of synchronization to environmental 24-h rhythms, (2) internal desynchronization among body clocks, (3) low rhythm amplitude, and (4) changes in sleep architecture. We provide prospects for relatively simple chronotherapies addressing these features that are easy to implement in clinical practice.
Collapse
Affiliation(s)
- Anisja Hühne
- a Circadian Biology Group, Department of Psychiatry , Ludwig Maximilian University , Munich , Germany
| | - David K Welsh
- b Veterans Affairs San Diego Healthcare System , San Diego , CA , USA.,c Department of Psychiatry & Center for Circadian Biology , University of California San Diego , La Jolla , CA , USA
| | - Dominic Landgraf
- a Circadian Biology Group, Department of Psychiatry , Ludwig Maximilian University , Munich , Germany
| |
Collapse
|
34
|
Yaw AM, Woodruff RW, Prosser RA, Glass JD. Paternal Cocaine Disrupts Offspring Circadian Clock Function in a Sex-Dependent Manner in Mice. Neuroscience 2018; 379:257-268. [DOI: 10.1016/j.neuroscience.2018.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/20/2018] [Accepted: 03/09/2018] [Indexed: 10/17/2022]
|
35
|
Excitatory and inhibitory synaptic dysfunction in mania: an emerging hypothesis from animal model studies. Exp Mol Med 2018; 50:1-11. [PMID: 29628501 PMCID: PMC5938027 DOI: 10.1038/s12276-018-0028-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 12/26/2022] Open
Abstract
Bipolar disorder (BD) is a common psychiatric disorder characterized by recurrent mood swings between depression and mania, and is associated with high treatment costs. The existence of manic episodes is the defining feature of BD, during which period, patients experience extreme elevation in activity, energy, and mood, with changes in sleep patterns that together severely impair their ability to function in daily life. Despite some limitations in recapitulating the complex features of human disease, several rodent models of mania have been generated and characterized, which have provided important insights toward understanding its underlying pathogenic mechanisms. Among the mechanisms, neuronal excitatory and inhibitory (E/I) synaptic dysfunction in some brain regions, including the frontal cortex, hippocampus, and striatum, is an emerging hypothesis explaining mania. In this review, we highlight recent studies of rodent manic models having impairments in the E/I synaptic development and function. We also summarize the molecular and functional changes of E/I synapses by some mood stabilizers that may contribute to the therapeutic efficacy of drugs. Furthermore, we discuss potential future directions in the study of this emerging hypothesis to better connect the outcomes of basic research to the treatment of patients with this devastating mental illness. Studies in rodents offer insights into bipolar disorder that may help understanding and treatment of this common and debilitating condition. Kihoon Han and colleagues at Korea University in Seoul review research using mice and rats to model the episodes of mania in patients with bipolar disorder. The research supports an emerging hypothesis implicating specific problems with nervous transmission in the brain in the onset of mania. The hypothesis suggests that the transmission of signals between particular nerve cells whose normal function is either to excite or to inhibit other nerve cells may be involved. It also indicates regions of the brain most involved in manic episodes. Changes at the affected nerve junctions—called synapses—brought about by mood-stabilizing drugs are examined. The hypothesis suggests new approaches to treatment options for researchers to explore.
Collapse
|
36
|
Schuch JB, Genro JP, Bastos CR, Ghisleni G, Tovo-Rodrigues L. The role of CLOCK gene in psychiatric disorders: Evidence from human and animal research. Am J Med Genet B Neuropsychiatr Genet 2018; 177:181-198. [PMID: 28902457 DOI: 10.1002/ajmg.b.32599] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
The circadian clock system drives daily rhythms in physiology, metabolism, and behavior in mammals. Molecular mechanisms of this system consist of multiple clock genes, with Circadian Locomotor Output Cycles Kaput (CLOCK) as a core member that plays an important role in a wide range of behaviors. Alterations in the CLOCK gene are associated with common psychiatric disorders as well as with circadian disturbances comorbidities. This review addresses animal, molecular, and genetic studies evaluating the role of the CLOCK gene on many psychiatric conditions, namely autism spectrum disorder, schizophrenia, attention-deficit/hyperactivity disorder, major depressive disorder, bipolar disorder, anxiety disorder, and substance use disorder. Many animal experiments focusing on the effects of the Clock gene in behavior related to psychiatric conditions have shown consistent biological plausibility and promising findings. In humans, genetic and gene expression studies regarding disorder susceptibility, sleep disturbances related comorbidities, and response to pharmacological treatment, in general, are in agreement with animal studies. However, the number of controversial results is high. Literature suggests that the CLOCK gene exerts important influence on these conditions, and influences the susceptibility to phenotypes of psychiatric disorders.
Collapse
Affiliation(s)
- Jaqueline B Schuch
- Laboratory of Immunosenescence, Graduate Program in Biomedical Gerontology, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Julia P Genro
- Graduate Program in Bioscience, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Clarissa R Bastos
- Laboratory of Clinical Neuroscience, Graduate Program in Health and Behavior, Universidade Católica de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Gabriele Ghisleni
- Laboratory of Clinical Neuroscience, Graduate Program in Health and Behavior, Universidade Católica de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Luciana Tovo-Rodrigues
- Graduate Program in Epidemiology, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| |
Collapse
|
37
|
Kristensen M, Nierenberg AA, Østergaard SD. Face and predictive validity of the ClockΔ19 mouse as an animal model for bipolar disorder: a systematic review. Mol Psychiatry 2018; 23:70-80. [PMID: 29112195 DOI: 10.1038/mp.2017.192] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Mice carrying the circadian locomotor output cycles Kaput delta 19 N-ethyl-N-nitrosoure (ENU) mutation (ClockΔ19) are used as an animal model for bipolar disorder (BD). We aimed to systematically review the face validity (phenotypical and pathophysiological resemblance with BD) and predictive validity (responsiveness to treatments used in BD) of this model in adherence with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. We carried out a systematic search of the databases PubMed and Embase, combining search terms covering BD and ClockΔ19. The 22 studies included in the review (from a total of 1281 identified records) show that the behavioral phenotype of the ClockΔ19 mouse is characterized by hyperactivity, decreased anxiety-like behavior, decreased depression-like behavior and increased preference for rewarding stimuli. This is highly consistent with mania in humans. Moreover, the ClockΔ19 mouse exhibits rapid mood cycling (a manic-like phenotype during the day followed by euthymia at night), which is consistent with BD. Chronic administration of lithium, a drug with well established mood-stabilizing effect in humans with BD, reverses the majority of the bipolar-like traits and most of the neurobiological abnormalities observed in the ClockΔ19 mouse. In conclusion, the ClockΔ19 mouse has substantial face validity as an animal model for BD. The predictive validity of the ClockΔ19 mouse has primarily been investigated via studies using lithium challenge. Therefore, further studies are needed to determine how the ClockΔ19 mouse responds to other mood-stabilizing treatments of BD such as valproate, lamotrigine, carbamazepine, oxcarbazepine, antipsychotics, electroconvulsive therapy and various light interventions.
Collapse
Affiliation(s)
- M Kristensen
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - A A Nierenberg
- Bipolar Clinic and Research Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - S D Østergaard
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| |
Collapse
|
38
|
De Nobrega AK, Lyons LC. Drosophila: An Emergent Model for Delineating Interactions between the Circadian Clock and Drugs of Abuse. Neural Plast 2017; 2017:4723836. [PMID: 29391952 PMCID: PMC5748135 DOI: 10.1155/2017/4723836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/13/2017] [Indexed: 01/12/2023] Open
Abstract
Endogenous circadian oscillators orchestrate rhythms at the cellular, physiological, and behavioral levels across species to coordinate activity, for example, sleep/wake cycles, metabolism, and learning and memory, with predictable environmental cycles. The 21st century has seen a dramatic rise in the incidence of circadian and sleep disorders with globalization, technological advances, and the use of personal electronics. The circadian clock modulates alcohol- and drug-induced behaviors with circadian misalignment contributing to increased substance use and abuse. Invertebrate models, such as Drosophila melanogaster, have proven invaluable for the identification of genetic and molecular mechanisms underlying highly conserved processes including the circadian clock, drug tolerance, and reward systems. In this review, we highlight the contributions of Drosophila as a model system for understanding the bidirectional interactions between the circadian system and the drugs of abuse, alcohol and cocaine, and illustrate the highly conserved nature of these interactions between Drosophila and mammalian systems. Research in Drosophila provides mechanistic insights into the corresponding behaviors in higher organisms and can be used as a guide for targeted inquiries in mammals.
Collapse
Affiliation(s)
- Aliza K. De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Lisa C. Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
39
|
Neural Mechanisms of Circadian Regulation of Natural and Drug Reward. Neural Plast 2017; 2017:5720842. [PMID: 29359051 PMCID: PMC5735684 DOI: 10.1155/2017/5720842] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/07/2017] [Accepted: 10/11/2017] [Indexed: 01/26/2023] Open
Abstract
Circadian rhythms are endogenously generated near 24-hour variations of physiological and behavioral functions. In humans, disruptions to the circadian system are associated with negative health outcomes, including metabolic, immune, and psychiatric diseases, such as addiction. Animal models suggest bidirectional relationships between the circadian system and drugs of abuse, whereby desynchrony, misalignment, or disruption may promote vulnerability to drug use and the transition to addiction, while exposure to drugs of abuse may entrain, disrupt, or perturb the circadian timing system. Recent evidence suggests natural (i.e., food) and drug rewards may influence overlapping neural circuitry, and the circadian system may modulate the physiological and behavioral responses to these stimuli. Environmental disruptions, such as shifting schedules or shorter/longer days, influence food and drug intake, and certain mutations of circadian genes that control cellular rhythms are associated with altered behavioral reward. We highlight the more recent findings associating circadian rhythms to reward function, linking environmental and genetic evidence to natural and drug reward and related neural circuitry.
Collapse
|
40
|
Ozburn AR, Kern J, Parekh PK, Logan RW, Liu Z, Falcon E, Becker-Krail D, Purohit K, Edgar NM, Huang Y, McClung CA. NPAS2 Regulation of Anxiety-Like Behavior and GABAA Receptors. Front Mol Neurosci 2017; 10:360. [PMID: 29163035 PMCID: PMC5675889 DOI: 10.3389/fnmol.2017.00360] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/19/2017] [Indexed: 12/26/2022] Open
Abstract
Abnormal circadian rhythms and circadian genes are strongly associated with several psychiatric disorders. Neuronal PAS Domain Protein 2 (NPAS2) is a core component of the molecular clock that acts as a transcription factor and is highly expressed in reward- and stress-related brain regions such as the striatum. However, the mechanism by which NPAS2 is involved in mood-related behaviors is still unclear. We measured anxiety-like behaviors in mice with a global null mutation in Npas2 (Npas2 null mutant mice) and found that Npas2 null mutant mice exhibit less anxiety-like behavior than their wild-type (WT) littermates (in elevated plus maze, light/dark box and open field assay). We assessed the effects of acute or chronic stress on striatal Npas2 expression, and found that both stressors increased levels of Npas2. Moreover, knockdown of Npas2 in the ventral striatum resulted in a similar reduction of anxiety-like behaviors as seen in the Npas2 null mutant mouse. Additionally, we identified Gabra genes as transcriptional targets of NPAS2, found that Npas2 null mutant mice exhibit reduced sensitivity to the GABAa positive allosteric modulator, diazepam and that knockdown of Npas2 reduced Gabra1 expression and response to diazepam in the ventral striatum. These results: (1) implicate Npas2 in the response to stress and the development of anxiety; and (2) provide functional evidence for the regulation of GABAergic neurotransmission by NPAS2 in the ventral striatum.
Collapse
Affiliation(s)
- Angela R Ozburn
- Portland Veterans Affairs Medical Center, Research and Development Service, Portland, OR, United States.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States.,Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Joseph Kern
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Puja K Parekh
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Ryan W Logan
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Zheng Liu
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Edgardo Falcon
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Darius Becker-Krail
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Kush Purohit
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Nicole M Edgar
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Yanhua Huang
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Colleen A McClung
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
41
|
Abstract
Reward-related learning, including that associated with drugs of abuse, is largely mediated by the dopaminergic mesolimbic pathway. Mesolimbic neurophysiology and motivated behavior, in turn, are modulated by the circadian timing system which generates ∼24-h rhythms in cellular activity. Both drug taking and seeking and mesolimbic dopaminergic neurotransmission can vary widely over the day. Moreover, circadian clock genes are expressed in ventral tegmental area dopaminergic cells and in mesolimbic target regions where they can directly modulate reward-related neurophysiology and behavior. There also exists a reciprocal influence between drug taking and circadian timing as the administration of drugs of abuse can alter behavioral rhythms and circadian clock gene expression in mesocorticolimbic structures. These interactions suggest that manipulations of the circadian timing system may have some utility in the treatment of substance abuse disorders. Here, the literature on bidirectional interactions between the circadian timing system and drug taking is briefly reviewed, and potential chronotherapeutic considerations for the treatment of addiction are discussed.
Collapse
|
42
|
RETRACTED ARTICLE: A Clock mutation enhances light-phase responsiveness to cocaine in locomotor activity and self-administration with impulsive-like responding in mice. Psychopharmacology (Berl) 2017; 234:185. [PMID: 27068482 DOI: 10.1007/s00213-016-4284-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
|
43
|
Abstract
In the face of chronic stress, some individuals can maintain normal function while others go on to develop mental illness. Addiction, affecting one in every twelve people in America, is a substance use disorder long associated with stressful life events and disruptions in the sleep/wake cycle. The circadian and stress response systems have evolved to afford adaptability to environmental changes and allow for maintenance of functional stability, or homeostasis. This mini-review will discuss how circadian rhythms and stress individually affect drug response, affect each other, and how their interactions may regulate reward-related behavior. In particular, we will focus on the interactions between the circadian clock and the regulation of glucocorticoids by the hypothalamic-pituitary-adrenal (HPA) axis. Determining how these two systems act on dopaminergic reward circuitry may not only reveal the basis for vulnerability to addiction, but may also illuminate potential therapeutic targets for future investigation.
Collapse
Affiliation(s)
- Darius Becker-Krail
- School of Medicine, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Colleen McClung
- School of Medicine, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
44
|
Parekh PK, McClung CA. Circadian Mechanisms Underlying Reward-Related Neurophysiology and Synaptic Plasticity. Front Psychiatry 2016; 6:187. [PMID: 26793129 PMCID: PMC4709415 DOI: 10.3389/fpsyt.2015.00187] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/21/2015] [Indexed: 12/18/2022] Open
Abstract
Evidence from clinical and preclinical research provides an undeniable link between disruptions in the circadian clock and the development of psychiatric diseases, including mood and substance abuse disorders. The molecular clock, which controls daily patterns of physiological and behavioral activity in living organisms, when desynchronized, may exacerbate or precipitate symptoms of psychiatric illness. One of the outstanding questions remaining in this field is that of cause and effect in the relationship between circadian rhythm disruption and psychiatric disease. Focus has recently turned to uncovering the role of circadian proteins beyond the maintenance of homeostatic systems and outside of the suprachiasmatic nucleus (SCN), the master pacemaker region of the brain. In this regard, several groups, including our own, have sought to understand how circadian proteins regulate mechanisms of synaptic plasticity and neurotransmitter signaling in mesocorticolimbic brain regions, which are known to be critically involved in reward processing and mood. This regulation can come in the form of direct transcriptional control of genes central to mood and reward, including those associated with dopaminergic activity in the midbrain. It can also be seen at the circuit level through indirect connections of mesocorticolimbic regions with the SCN. Circadian misalignment paradigms as well as genetic models of circadian disruption have helped to elucidate some of the complex interactions between these systems and neural activity influencing behavior. In this review, we explore findings that link circadian protein function with synaptic adaptations underlying plasticity as it may contribute to the development of mood disorders and addiction. In light of recent advances in technology and sophisticated methods for molecular and circuit-level interrogation, we propose future directions aimed at teasing apart mechanisms through which the circadian system modulates mood and reward-related behavior.
Collapse
Affiliation(s)
- Puja K. Parekh
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Colleen A. McClung
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Ozburn AR, Purohit K, Parekh PK, Kaplan GN, Falcon E, Mukherjee S, Cates HM, McClung CA. Functional Implications of the CLOCK 3111T/C Single-Nucleotide Polymorphism. Front Psychiatry 2016; 7:67. [PMID: 27148095 PMCID: PMC4838618 DOI: 10.3389/fpsyt.2016.00067] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 04/04/2016] [Indexed: 01/01/2023] Open
Abstract
Circadian rhythm disruptions are prominently associated with bipolar disorder (BD). Circadian rhythms are regulated by the molecular clock, a family of proteins that function together in a transcriptional-translational feedback loop. The CLOCK protein is a key transcription factor of this feedback loop, and previous studies have found that manipulations of the Clock gene are sufficient to produce manic-like behavior in mice (1). The CLOCK 3111T/C single-nucleotide polymorphism (SNP; rs1801260) is a genetic variation of the human CLOCK gene that is significantly associated with increased frequency of manic episodes in BD patients (2). The 3111T/C SNP is located in the 3'-untranslated region of the CLOCK gene. In this study, we sought to examine the functional implications of the human CLOCK 3111T/C SNP by transfecting a mammalian cell line (mouse embryonic fibroblasts isolated from Clock(-/-) knockout mice) with pcDNA plasmids containing the human CLOCK gene with either the T or C SNP at position 3111. We then measured circadian gene expression over a 24-h time period. We found that the CLOCK3111C SNP resulted in higher mRNA levels than the CLOCK 3111T SNP. Furthermore, we found that Per2, a transcriptional target of CLOCK, was also more highly expressed with CLOCK 3111C expression, indicating that the 3'-UTR SNP affects the expression, function, and stability of CLOCK mRNA.
Collapse
Affiliation(s)
- Angela R Ozburn
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; Portland Alcohol Research Center, VA Medical Center, Portland, OR, USA
| | - Kush Purohit
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Puja K Parekh
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Gabrielle N Kaplan
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Edgardo Falcon
- Department of Pharmacology, University of Pennsylvania , Philadelphia, PA , USA
| | - Shibani Mukherjee
- Department of Psychiatry, University of Texas Southwestern Medical Center , Dallas, TX , USA
| | - Hannah M Cates
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Colleen A McClung
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| |
Collapse
|
46
|
Radwan B, Liu H, Chaudhury D. Regulation and Modulation of Depression-Related Behaviours: Role of Dopaminergic Neurons. DOPAMINE AND SLEEP 2016:147-190. [DOI: 10.1007/978-3-319-46437-4_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
|
47
|
Ozburn AR, Janowsky AJ, Crabbe JC. Commonalities and Distinctions Among Mechanisms of Addiction to Alcohol and Other Drugs. Alcohol Clin Exp Res 2015; 39:1863-77. [PMID: 26431116 PMCID: PMC4594192 DOI: 10.1111/acer.12810] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/10/2015] [Indexed: 01/25/2023]
Abstract
BACKGROUND Alcohol abuse is comorbid with abuse of many other drugs, some with similar pharmacology and others quite different. This leads to the hypothesis of an underlying, unitary dysfunctional neurobiological basis for substance abuse risk and consequences. METHODS In this review, we discuss commonalities and distinctions of addiction to alcohol and other drugs. We focus on recent advances in preclinical studies using rodent models of drug self-administration. RESULTS While there are specific behavioral and molecular manifestations common to alcohol, psychostimulant, opioid, and nicotine dependence, attempts to propose a unifying theory of the addictions inevitably face details where distinctions are found among classes of drugs. CONCLUSIONS For alcohol, versus other drugs of abuse, we discuss and compare advances in: (i) neurocircuitry important for the different stages of drug dependence; (ii) transcriptomics and genetical genomics; and (iii) enduring effects, noting in particular the contributions of behavioral genetics and animal models.
Collapse
Affiliation(s)
- Angela R. Ozburn
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Aaron J. Janowsky
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Department of Psychiatry, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - John C. Crabbe
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
48
|
Logan RW, McClung CA. Animal models of bipolar mania: The past, present and future. Neuroscience 2015; 321:163-188. [PMID: 26314632 DOI: 10.1016/j.neuroscience.2015.08.041] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 12/19/2022]
Abstract
Bipolar disorder (BD) is the sixth leading cause of disability in the world according to the World Health Organization and affects nearly six million (∼2.5% of the population) adults in the United State alone each year. BD is primarily characterized by mood cycling of depressive (e.g., helplessness, reduced energy and activity, and anhedonia) and manic (e.g., increased energy and hyperactivity, reduced need for sleep, impulsivity, reduced anxiety and depression), episodes. The following review describes several animal models of bipolar mania with a focus on more recent findings using genetically modified mice, including several with the potential of investigating the mechanisms underlying 'mood' cycling (or behavioral switching in rodents). We discuss whether each of these models satisfy criteria of validity (i.e., face, predictive, and construct), while highlighting their strengths and limitations. Animal models are helping to address critical questions related to pathophysiology of bipolar mania, in an effort to more clearly define necessary targets of first-line medications, lithium and valproic acid, and to discover novel mechanisms with the hope of developing more effective therapeutics. Future studies will leverage new technologies and strategies for integrating animal and human data to reveal important insights into the etiology, pathophysiology, and treatment of BD.
Collapse
Affiliation(s)
- R W Logan
- University of Pittsburgh School of Medicine, Department of Psychiatry, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, United States
| | - C A McClung
- University of Pittsburgh School of Medicine, Department of Psychiatry, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, United States.
| |
Collapse
|
49
|
Doyle SE, Feng H, Garber G, Menaker M, Lynch WJ. Effects of circadian disruption on methamphetamine consumption in methamphetamine-exposed rats. Psychopharmacology (Berl) 2015; 232:2169-79. [PMID: 25543849 PMCID: PMC4433617 DOI: 10.1007/s00213-014-3845-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 01/06/2023]
Abstract
RATIONALE A substantial number of clinical studies indicate associations between sleep abnormalities and drug abuse; however, the role played by the circadian system in the development of addiction is largely unknown. OBJECTIVE The aim of this study was to examine the effects of experimentally induced chronic jet lag on methamphetamine consumption in a rat model of methamphetamine drinking. METHODS Male Sprague-Dawley rats (n = 32) were housed in running wheel cages in a 12:12 h light:dark cycle. One group of rats (n = 16) was given 2 weeks of forced methamphetamine consumption (0.01 % in drinking water; meth pre-exposed) while a second group (n = 16, not pre-exposed) received water only. This was followed by a 2-week abstinence period during which half of the animals from each group were exposed to four consecutive 6-h advancing phase shifts of the light:dark cycle, while the other half remained on the original light:dark cycle. Methamphetamine consumption was assessed in all rats following the deprivation period using a two-bottle choice paradigm. RESULTS Methamphetamine consumption was initially lower in methamphetamine pre-exposed versus not pre-exposed rats. However, during the second week following abstinence, consumption was significantly higher in phase-shifted rats of the methamphetamine pre-exposed group compared to all other groups. CONCLUSIONS These data reveal an effect of circadian rhythm disturbance on methamphetamine consumption and suggest that dysregulation of the circadian system be considered in the etiology of relapse and addiction.
Collapse
Affiliation(s)
- Susan E. Doyle
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22904, USA
| | - Hanting Feng
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Garrett Garber
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22904, USA
| | - Michael Menaker
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Wendy J. Lynch
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22904, USA
| |
Collapse
|
50
|
Perreau-Lenz S, Spanagel R. Clock genes × stress × reward interactions in alcohol and substance use disorders. Alcohol 2015; 49:351-7. [PMID: 25943583 PMCID: PMC4457607 DOI: 10.1016/j.alcohol.2015.04.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 04/13/2015] [Indexed: 12/31/2022]
Abstract
Adverse life events and highly stressful environments have deleterious consequences for mental health. Those environmental factors can potentiate alcohol and drug abuse in vulnerable individuals carrying specific genetic risk factors, hence producing the final risk for alcohol- and substance-use disorders development. The nature of these genes remains to be fully determined, but studies indicate their direct or indirect relation to the stress hypothalamo-pituitary-adrenal (HPA) axis and/or reward systems. Over the past decade, clock genes have been revealed to be key-players in influencing acute and chronic alcohol/drug effects. In parallel, the influence of chronic stress and stressful life events in promoting alcohol and substance use and abuse has been demonstrated. Furthermore, the reciprocal interaction of clock genes with various HPA-axis components, as well as the evidence for an implication of clock genes in stress-induced alcohol abuse, have led to the idea that clock genes, and Period genes in particular, may represent key genetic factors to consider when examining gene × environment interaction in the etiology of addiction. The aim of the present review is to summarize findings linking clock genes, stress, and alcohol and substance abuse, and to propose potential underlying neurobiological mechanisms.
Collapse
Affiliation(s)
- Stéphanie Perreau-Lenz
- Institute of Psychopharmacology, Central Institute for Mental Health, Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany; SRI International, Center for Neuroscience, Biosciences Division, Menlo Park, CA, USA.
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute for Mental Health, Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|