101
|
Salaberry NL, Mateo M, Mendoza J. The Clock Gene Rev-Erbα Regulates Methamphetamine Actions on Circadian Timekeeping in the Mouse Brain. Mol Neurobiol 2016; 54:5327-5334. [PMID: 27581301 DOI: 10.1007/s12035-016-0076-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/23/2016] [Indexed: 12/21/2022]
Abstract
Circadian rhythms are strongly affected by drugs. In rodents, chronic methamphetamine (METH) intake changes circadian activity rhythms, mainly by altering light synchronization that generates the expression of a free-running rhythm with a period longer than 24 h and a second behavioral component that is independent of the main suprachiasmatic (SCN) clock. Although a number of clock genes do not appear to be involved in the effects of METH on circadian behavior, the molecular clockwork controlling these changes is still unclear. Therefore, we investigated the role of the clock gene Rev-Erbα in METH-induced behavioral and molecular responses using knockout mice and their wild-type littermates. Chronic intake of METH alters period circadian behavior of wild-type mice. However, in mice lacking the clock gene Rev-Erbα METH had no effect on their behavioral rhythms. Furthermore, PER2 bioluminescence rhythms in two extra-SCN brain oscillators, the dorsomedial hypothalamus and the habenula, were altered by METH in wild type but not in KO mice. Together, the present results implicate Rev-Erbα in the modulation of the circadian responses to METH and may provide a better comprehension into the mechanisms underlying circadian alterations provoked by drug addiction.
Collapse
Affiliation(s)
- Nora L Salaberry
- CNRS UPR-3212, Institute of Cellular and Integrative Neurosciences, 5 rue Blaise Pascal, 67084, Strasbourg, Cedex, France
| | - Maria Mateo
- CNRS UPR-3212, Institute of Cellular and Integrative Neurosciences, 5 rue Blaise Pascal, 67084, Strasbourg, Cedex, France
| | - Jorge Mendoza
- CNRS UPR-3212, Institute of Cellular and Integrative Neurosciences, 5 rue Blaise Pascal, 67084, Strasbourg, Cedex, France.
| |
Collapse
|
102
|
Verwey M, Dhir S, Amir S. Circadian influences on dopamine circuits of the brain: regulation of striatal rhythms of clock gene expression and implications for psychopathology and disease. F1000Res 2016; 5. [PMID: 27635233 PMCID: PMC5007753 DOI: 10.12688/f1000research.9180.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2016] [Indexed: 12/18/2022] Open
Abstract
Circadian clock proteins form an autoregulatory feedback loop that is central to the endogenous generation and transmission of daily rhythms in behavior and physiology. Increasingly, circadian rhythms in clock gene expression are being reported in diverse tissues and brain regions that lie outside of the suprachiasmatic nucleus (SCN), the master circadian clock in mammals. For many of these extra-SCN rhythms, however, the region-specific implications are still emerging. In order to gain important insights into the potential behavioral, physiological, and psychological relevance of these daily oscillations, researchers have begun to focus on describing the neurochemical, hormonal, metabolic, and epigenetic contributions to the regulation of these rhythms. This review will highlight important sites and sources of circadian control within dopaminergic and striatal circuitries of the brain and will discuss potential implications for psychopathology and disease
. For example, rhythms in clock gene expression in the dorsal striatum are sensitive to changes in dopamine release, which has potential implications for Parkinson’s disease and drug addiction. Rhythms in the ventral striatum and limbic forebrain are sensitive to psychological and physical stressors, which may have implications for major depressive disorder. Collectively, a rich circadian tapestry has emerged that forces us to expand traditional views and to reconsider the psychopathological, behavioral, and physiological importance of these region-specific rhythms in brain areas that are not immediately linked with the regulation of circadian rhythms.
Collapse
Affiliation(s)
- Michael Verwey
- Center for Studies in Behavioural Neurobiology, FRQS Groupe de Recherche en Neurobiologie Comportementale, Concorida University, Montreal, Quebec, Canada
| | | | - Shimon Amir
- Center for Studies in Behavioural Neurobiology, FRQS Groupe de Recherche en Neurobiologie Comportementale, Concorida University, Montreal, Quebec, Canada
| |
Collapse
|
103
|
Rawashdeh O, Jilg A, Maronde E, Fahrenkrug J, Stehle JH. Period1gates the circadian modulation of memory-relevant signaling in mouse hippocampus by regulating the nuclear shuttling of the CREB kinase pP90RSK. J Neurochem 2016; 138:731-45. [DOI: 10.1111/jnc.13689] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Oliver Rawashdeh
- Institute of Cellular and Molecular Anatomy; Dr. Senckenbergische Anatomie; Goethe-University; Frankfurt Germany
- School of Biomedical Sciences; University of Queensland; St Lucia Qld Australia
| | - Antje Jilg
- Institute of Cellular and Molecular Anatomy; Dr. Senckenbergische Anatomie; Goethe-University; Frankfurt Germany
| | - Erik Maronde
- Institute of Cellular and Molecular Anatomy; Dr. Senckenbergische Anatomie; Goethe-University; Frankfurt Germany
| | - Jan Fahrenkrug
- Department of Clinical Chemistry; Bispebjerg Hospital, Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Jörg H. Stehle
- Institute of Cellular and Molecular Anatomy; Dr. Senckenbergische Anatomie; Goethe-University; Frankfurt Germany
| |
Collapse
|
104
|
Jones JR, McMahon DG. The core clock gene Per1 phases molecular and electrical circadian rhythms in SCN neurons. PeerJ 2016; 4:e2297. [PMID: 27602274 PMCID: PMC4991845 DOI: 10.7717/peerj.2297] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/06/2016] [Indexed: 11/20/2022] Open
Abstract
The brain’s biological clock, the suprachiasmatic nucleus (SCN), exhibits endogenous 24-hour rhythms in gene expression and spontaneous firing rate; however, the functional relationship between these neuronal rhythms is not fully understood. Here, we used a Per1::GFP transgenic mouse line that allows for the simultaneous quantification of molecular clock state and firing rate in SCN neurons to examine the relationship between these key components of the circadian clock. We find that there is a stable, phased relationship between E-box-driven clock gene expression and spontaneous firing rate in SCN neurons and that these relationships are independent of light input onto the system or of GABAA receptor-mediated synaptic activity. Importantly, the concordant phasing of gene and neural rhythms is disrupted in the absence of the homologous clock gene Per1, but persists in the absence of the core clock gene Per2. These results suggest that Per1 plays a unique, non-redundant role in phasing gene expression and firing rate rhythms in SCN neurons to increase the robustness of cellular timekeeping.
Collapse
Affiliation(s)
- Jeff R Jones
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, United States; Current affiliation: Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Douglas G McMahon
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, United States; Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
105
|
Tataroglu O, Davidson AJ, Benvenuto LJ, Menaker M. The Methamphetamine-Sensitive Circadian Oscillator (MASCO) in Mice. J Biol Rhythms 2016; 21:185-94. [PMID: 16731658 DOI: 10.1177/0748730406287529] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The suprachiasmatic nucleus (SCN) orchestrates synchrony among many peripheral oscillators and is required for circadian rhythms of locomotor activity and many physiological processes. However, the unique effects of methamphetamine (MAP) on circadian behavior suggest the presence of an SCN-independent, methamphetamine-sensitive circadian oscillator (MASCO). Substantial data collected using rat models show that chronic methamphetamine dramatically lengthens circadian period of locomotor activity rhythms and induces rhythms in animals lacking an SCN. However, the anatomical substrate and the molecular components of the MASCO are unknown. The response to MAP is less well studied in mice, a model that would provide the genetic tools to probe the molecular components of this extra-SCN oscillator. The authors tested the effects of chronic MAP on 2 strains of intact and SCN-lesioned mice in constant dark and constant light. Furthermore, they applied various MAP availability schedules to SCN-lesioned mice to confirm the circadian nature of the underlying oscillator. The results indicate that this oscillator has circadian properties. In intact mice, the MASCO interacts with the SCN in a manner that is strain, sex, and dose dependent. In SCN-lesioned mice, it induces robust free-running locomotor rhythmicity, which persists for up to 14 cycles after methamphetamine is withdrawn. In the future, localization of the MASCO and characterization of its underlying molecular mechanism, as well as its interactions with other oscillators in the body, will be essential to a complete understanding of the organization of the mammalian circadian system.
Collapse
Affiliation(s)
- Ozgür Tataroglu
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | | | | | | |
Collapse
|
106
|
Baranger DAA, Ifrah C, Prather AA, Carey CE, Corral-Frías NS, Drabant Conley E, Hariri AR, Bogdan R. PER1 rs3027172 Genotype Interacts with Early Life Stress to Predict Problematic Alcohol Use, but Not Reward-Related Ventral Striatum Activity. Front Psychol 2016; 7:464. [PMID: 27065929 PMCID: PMC4814479 DOI: 10.3389/fpsyg.2016.00464] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 03/15/2016] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence suggests that the circadian and stress regulatory systems contribute to alcohol use disorder (AUD) risk, which may partially arise through effects on reward-related neural function. The C allele of the PER1 rs3027172 single nucleotide polymorphism (SNP) reduces PER1 expression in cells incubated with cortisol and has been associated with increased risk for adult AUD and problematic drinking among adolescents exposed to high levels of familial psychosocial adversity. Using data from undergraduate students who completed the ongoing Duke Neurogenetics Study (DNS) (n = 665), we tested whether exposure to early life stress (ELS; Childhood Trauma Questionnaire) moderates the association between rs3027172 genotype and later problematic alcohol use (Alcohol Use Disorders Identification Test) as well as ventral striatum (VS) reactivity to reward (card-guessing task while functional magnetic resonance imaging data were acquired). Initial analyses found that PER1 rs3027172 genotype interacted with ELS to predict both problematic drinking and VS reactivity; minor C allele carriers, who were also exposed to elevated ELS reported greater problematic drinking and exhibited greater ventral striatum reactivity to reward-related stimuli. When gene × covariate and environment × covariate interactions were controlled for, the interaction predicting problematic alcohol use remained significant (p < 0.05, corrected) while the interaction predicting VS reactivity was no longer significant. These results extend our understanding of relationships between PER1 genotype, ELS, and problematic alcohol use, and serve as a cautionary tale on the importance of controlling for potential confounders in studies of moderation including gene × environment interactions.
Collapse
Affiliation(s)
- David A. A. Baranger
- Brain Laboratory, Department of Psychological and Brain Sciences, Washington University in St. LouisSt. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University in St. LouisSt. Louis, MO, USA
| | - Chloé Ifrah
- Brain Laboratory, Department of Psychological and Brain Sciences, Washington University in St. LouisSt. Louis, MO, USA
| | - Aric A. Prather
- Department of Psychiatry, University of California, San FranciscoSan Francisco, CA, USA
| | - Caitlin E. Carey
- Brain Laboratory, Department of Psychological and Brain Sciences, Washington University in St. LouisSt. Louis, MO, USA
| | | | | | - Ahmad R. Hariri
- Laboratory of NeuroGenetics, Department of Psychology and Neuroscience, Duke UniversityDurham, NC, USA
| | - Ryan Bogdan
- Brain Laboratory, Department of Psychological and Brain Sciences, Washington University in St. LouisSt. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University in St. LouisSt. Louis, MO, USA
- Department of Psychiatry, Washington University in St. LouisSt. Louis, MO, USA
| |
Collapse
|
107
|
Effect of circadian rhythm disturbance on morphine preference and addiction in male rats: Involvement of period genes and dopamine D1 receptor. Neuroscience 2016; 322:104-14. [PMID: 26892296 DOI: 10.1016/j.neuroscience.2016.02.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 01/16/2016] [Accepted: 02/10/2016] [Indexed: 12/17/2022]
Abstract
It is claimed that a correlation exists between disturbance of circadian rhythms by factors such as alteration of normal light-dark cycle and the development of addiction. However, the exact mechanisms involved in this relationship are not much understood. Here we have studied the effect of constant light on morphine voluntary consumption and withdrawal symptoms and also investigated the involvement of Per1, Per2 and dopamine D1 receptor in these processes. Male wistar rats were kept under standard (LD) or constant light (LL) conditions for one month. The plasma concentration of melatonin was evaluated by enzyme-linked immunosorbent assay (ELISA). Real-time PCR was used to determine the mRNA expression of Per1, Per2 and dopamine D1 receptor in the striatum and prefrontal cortex. Morphine preference (50mg/L) was evaluated in a two-bottle-choice paradigm for 10 weeks and withdrawal symptoms were recorded after administration of naloxone (3mg/kg). One month exposure to constant light resulted in a significant decrease of melatonin concentration in the LL group. In addition, mRNA levels of Per2 and dopamine D1 receptor were up-regulated in both the striatum and prefrontal cortex of the LL group. However, expression of Per1 gene was only up-regulated in the striatum of LL rats in comparison to LD animals. Furthermore, after one month exposure to constant light, morphine consumption and preference ratio and also severity of naloxone-induced withdrawal syndrome were significantly greater in LL animals. It is concluded that exposure to constant light by up-regulation of Per2 and dopamine D1 receptor in the striatum and prefrontal cortex and up-regulation of Per1 in the striatum and the possible involvement of melatonin makes animals vulnerable to morphine preference and addiction.
Collapse
|
108
|
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
|
109
|
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: 3.8] [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
|
110
|
Impact of Impulse Control Disorders on Sleep-Wake Regulation in Parkinson's Disease. PARKINSONS DISEASE 2015; 2015:970862. [PMID: 26664825 PMCID: PMC4667074 DOI: 10.1155/2015/970862] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/03/2015] [Indexed: 01/21/2023]
Abstract
Sleep disturbances are common in patients with Parkinson's disease (PD) and are even more prevalent in patients with behavioural addictions, such as pathological gambling, compulsive sexual behaviour, compulsive buying, binge eating, punding, and the compulsive use of dopamine replacement therapy. An overview of the relationship between these impulse control disorders and sleep disturbances is given and potential underlying mechanisms and treatment strategies are covered.
Collapse
|
111
|
Barandas R, Landgraf D, McCarthy MJ, Welsh DK. Circadian Clocks as Modulators of Metabolic Comorbidity in Psychiatric Disorders. Curr Psychiatry Rep 2015; 17:98. [PMID: 26483181 DOI: 10.1007/s11920-015-0637-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Psychiatric disorders such as schizophrenia, bipolar disorder, and major depressive disorder are often accompanied by metabolic dysfunction symptoms, including obesity and diabetes. Since the circadian system controls important brain systems that regulate affective, cognitive, and metabolic functions, and neuropsychiatric and metabolic diseases are often correlated with disturbances of circadian rhythms, we hypothesize that dysregulation of circadian clocks plays a central role in metabolic comorbidity in psychiatric disorders. In this review paper, we highlight the role of circadian clocks in glucocorticoid, dopamine, and orexin/melanin-concentrating hormone systems and describe how a dysfunction of these clocks may contribute to the simultaneous development of psychiatric and metabolic symptoms.
Collapse
Affiliation(s)
- Rita Barandas
- Department of Psychiatry, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- VA San Diego Healthcare System Psychiatry Service, La Jolla, CA, USA
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 9500 Gilman Drive MC-0603, La Jolla, CA, 92093-0603, USA
| | - Dominic Landgraf
- VA San Diego Healthcare System Psychiatry Service, La Jolla, CA, USA.
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 9500 Gilman Drive MC-0603, La Jolla, CA, 92093-0603, USA.
| | - Michael J McCarthy
- VA San Diego Healthcare System Psychiatry Service, La Jolla, CA, USA
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 9500 Gilman Drive MC-0603, La Jolla, CA, 92093-0603, USA
| | - David K Welsh
- VA San Diego Healthcare System Psychiatry Service, La Jolla, CA, USA
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 9500 Gilman Drive MC-0603, La Jolla, CA, 92093-0603, USA
| |
Collapse
|
112
|
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: 4.0] [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
|
113
|
Abstract
Multiple lines of evidence suggest that psychopathological symptoms of bipolar disorder arise in part from a malfunction of the circadian system, linking the disease with an abnormal internal timing. Alterations in circadian rhythms and sleep are core elements in the disorders, characterizing both mania and depression and having recently been shown during euthymia. Several human genetic studies have implicated specific genes that make up the genesis of circadian rhythms in the manifestation of mood disorders with polymorphisms in molecular clock genes not only showing an association with the disorder but having also been linked to its phenotypic particularities. Many medications used to treat the disorder, such as antidepressant and mood stabilizers, affect the circadian clock. Finally, circadian rhythms and sleep researches have been the starting point of the developing of chronobiological therapies. These interventions are safe, rapid and effective and they should be considered first-line strategies for bipolar depression.
Collapse
Affiliation(s)
- Sara Dallaspezia
- Department of Clinical Neurosciences, Scientific Institute and University Vita-Salute San Raffaele, Milan, Italy,
| | | |
Collapse
|
114
|
Horton WJ, Gissel HJ, Saboy JE, Wright KP, Stitzel JA. Melatonin administration alters nicotine preference consumption via signaling through high-affinity melatonin receptors. Psychopharmacology (Berl) 2015; 232:2519-30. [PMID: 25704105 PMCID: PMC4482784 DOI: 10.1007/s00213-015-3886-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 02/08/2015] [Indexed: 12/13/2022]
Abstract
RATIONALE While it is known that tobacco use varies across the 24-h day, the time-of-day effects are poorly understood. Findings from several previous studies indicate a potential role for melatonin in these time-of-day effects; however, the specific underlying mechanisms have not been well characterized. Understanding of these mechanisms may lead to potential novel smoking cessation treatments. OBJECTIVE The objective of this study is examine the role of melatonin and melatonin receptors in nicotine free-choice consumption METHODS A two-bottle oral nicotine choice paradigm was utilized with melatonin supplementation in melatonin-deficient mice (C57BL/6J) or without melatonin supplementation in mice proficient at melatonin synthesis (C3H/Ibg) compared to melatonin-proficient mice lacking both or one of the high-affinity melatonin receptors (MT1 and MT2; double-null mutant DM, or MT1 or MT2). Preference for bitter and sweet tastants also was assessed in wild-type and MT1 and MT2 DM mice. Finally, home cage locomotor monitoring was performed to determine the effect of melatonin administration on activity patterns. RESULTS Supplemental melatonin in drinking water significantly reduced free-choice nicotine consumption in C57BL/6J mice, which do not produce endogenous melatonin, while not altering activity patterns. Independently, genetic deletion of both MT1 and MT2 receptors in a melatonin-proficient mouse strain (C3H) resulted in significantly more nicotine consumption than controls. However, single genetic deletion of either the MT1 or MT2 receptor alone did not result in increased nicotine consumption. Deletion of MT1 and MT2 did not impact taste preference. CONCLUSIONS This study demonstrates that nicotine consumption can be affected by exogenous or endogenous melatonin and requires at least one of the high-affinity melatonin receptors. The fact that expression of either the MT1 or MT2 melatonin receptor is sufficient to maintain lower nicotine consumption suggests functional overlap and potential mechanistic explanations.
Collapse
Affiliation(s)
- William J. Horton
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80303,Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303
| | - Hannah J. Gissel
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80303
| | - Jennifer E. Saboy
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80303
| | - Kenneth P. Wright
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303
| | - Jerry A. Stitzel
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80303,Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303
| |
Collapse
|
115
|
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: 1.8] [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
|
116
|
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: 4.0] [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
|
117
|
Parekh PK, Ozburn AR, McClung CA. Circadian clock genes: effects on dopamine, reward and addiction. Alcohol 2015; 49:341-9. [PMID: 25641765 DOI: 10.1016/j.alcohol.2014.09.034] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 12/17/2022]
Abstract
Addiction is a widespread public health issue with social and economic ramifications. Substance abuse disorders are often accompanied by disruptions in circadian rhythms including sleep/wake cycles, which can exacerbate symptoms of addiction and dependence. Additionally, genetic disturbance of circadian molecular mechanisms can predispose some individuals to substance abuse disorders. In this review, we will discuss how circadian genes can regulate midbrain dopaminergic activity and subsequently, drug intake and reward. We will also suggest future directions for research on circadian genes and drugs of abuse.
Collapse
|
118
|
Ozburn AR, Falcon E, Twaddle A, Nugent AL, Gillman AG, Spencer SM, Arey RN, Mukherjee S, Lyons-Weiler J, Self DW, McClung CA. Direct regulation of diurnal Drd3 expression and cocaine reward by NPAS2. Biol Psychiatry 2015; 77:425-433. [PMID: 25444159 PMCID: PMC4315729 DOI: 10.1016/j.biopsych.2014.07.030] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 07/31/2014] [Accepted: 07/31/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Circadian gene disruptions are associated with the development of psychiatric disorders, including addiction. However, the mechanisms by which circadian genes regulate reward remain poorly understood. METHODS We used mice with a mutation in Npas2 and adeno-associated virus-short hairpin RNA mediated knockdown of Npas2 and Clock in the nucleus accumbens (NAc). We performed conditioned place preference assays. We utilized cell sorting quantitative real-time polymerase chain reaction, and chromatin immunoprecipitation followed by deep sequencing. RESULTS Npas2 mutants exhibit decreased sensitivity to cocaine reward, which is recapitulated with a knockdown of neuronal PAS domain protein 2 (NPAS2) specifically in the NAc, demonstrating the importance of NPAS2 in this region. Interestingly, reducing circadian locomotor output cycles kaput (CLOCK) (a homologue of NPAS2) in the NAc had no effect, suggesting an important distinction in NPAS2 and CLOCK function. Furthermore, we found that NPAS2 expression is restricted to Drd1 expressing neurons while CLOCK is ubiquitous. Moreover, NPAS2 and CLOCK have distinct temporal patterns of DNA binding, and we identified novel and unique binding sites for each protein. We identified the Drd3 dopamine receptor as a direct transcriptional target of NPAS2 and found that NPAS2 knockdown in the NAc disrupts its diurnal rhythm in expression. Chronic cocaine treatment likewise disrupts the normal rhythm in Npas2 and Drd3 expression in the NAc, which may underlie behavioral plasticity in response to cocaine. CONCLUSIONS Together, these findings identify an important role for the circadian protein, NPAS2, in the NAc in the regulation of dopamine receptor expression and drug reward.
Collapse
Affiliation(s)
- Angela R. Ozburn
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
| | - Edgardo Falcon
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390-9070
| | - Alan Twaddle
- Bioinformatics Analysis Core, Clinical and Translational Science Institute at the University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Andrea G. Gillman
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
| | - Sade M. Spencer
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390-9070
| | - Rachel N. Arey
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390-9070
| | - Shibani Mukherjee
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390-9070
| | - James Lyons-Weiler
- Bioinformatics Analysis Core, Clinical and Translational Science Institute at the University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - David W. Self
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390-9070
| | - Colleen A. McClung
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
| |
Collapse
|
119
|
Webb IC, Lehman MN, Coolen LM. Diurnal and circadian regulation of reward-related neurophysiology and behavior. Physiol Behav 2015; 143:58-69. [PMID: 25708277 DOI: 10.1016/j.physbeh.2015.02.034] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 12/16/2022]
Abstract
Here, we review work over the past two decades that has indicated drug reward is modulated by the circadian system that generates daily (i.e., 24h) rhythms in physiology and behavior. Specifically, drug-self administration, psychomotor stimulant-induced conditioned place preference, and locomotor sensitization vary widely across the day in various species. These drug-related behavioral rhythms are associated with rhythmic neural activity and dopaminergic signaling in the mesocorticolimbic pathways, with a tendency toward increased activity during the species typical wake period. While the mechanisms responsible for such cellular rhythmicity remain to be fully identified, circadian clock genes are expressed in these brain areas and can function locally to modulate both dopaminergic neurotransmission and drug-associated behavior. In addition, neural and endocrine inputs to these brain areas contribute to cellular and reward-related behavioral rhythms, with the medial prefrontal cortex playing a pivotal role. Acute or chronic administration of drugs of abuse can also alter clock gene expression in reward-related brain regions. Emerging evidence suggests that drug craving in humans is under a diurnal regulation and that drug reward may be influenced by clock gene polymorphisms. These latter findings, in particular, indicate that the development of therapeutic strategies to modulate the circadian influence on drug reward may prove beneficial in the treatment of substance abuse disorders.
Collapse
Affiliation(s)
- Ian C Webb
- Dept. of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Michael N Lehman
- Dept. of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lique M Coolen
- Dept. of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA; Dept. of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
120
|
Salaberry NL, Mendoza J. Insights into the Role of the Habenular Circadian Clock in Addiction. Front Psychiatry 2015; 6:179. [PMID: 26779042 PMCID: PMC4700272 DOI: 10.3389/fpsyt.2015.00179] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/07/2015] [Indexed: 12/15/2022] Open
Abstract
Drug addiction is a brain disease involving alterations in anatomy and functional neural communication. Drug intake and toxicity show daily rhythms in both humans and rodents. Evidence concerning the role of clock genes in drug intake has been previously reported. However, the implication of a timekeeping brain locus is much less known. The epithalamic lateral habenula (LHb) is now emerging as a key nucleus in drug intake and addiction. This brain structure modulates the activity of dopaminergic neurons from the ventral tegmental area, a central part of the reward system. Moreover, the LHb has circadian properties: LHb cellular activity (i.e., firing rate and clock genes expression) oscillates in a 24-h range, and the nucleus is affected by photic stimulation and has anatomical connections with the main circadian pacemaker, the suprachiasmatic nucleus. Here, we describe the current insights on the role of the LHb as a circadian oscillator and its possible implications on the rhythmic regulation of the dopaminergic activity and drug intake. These data could inspire new strategies to treat drug addiction, considering circadian timing as a principal factor.
Collapse
Affiliation(s)
- Nora L Salaberry
- CNRS UPR-3212, Institute of Cellular and Integrative Neurosciences, University of Strasbourg , Strasbourg , France
| | - Jorge Mendoza
- CNRS UPR-3212, Institute of Cellular and Integrative Neurosciences, University of Strasbourg , Strasbourg , France
| |
Collapse
|
121
|
Banerjee S, Wang Y, Solt LA, Griffett K, Kazantzis M, Amador A, El-Gendy BM, Huitron-Resendiz S, Roberts AJ, Shin Y, Kamenecka TM, Burris TP. Pharmacological targeting of the mammalian clock regulates sleep architecture and emotional behaviour. Nat Commun 2014; 5:5759. [PMID: 25536025 PMCID: PMC4495958 DOI: 10.1038/ncomms6759] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 11/04/2014] [Indexed: 12/20/2022] Open
Abstract
Synthetic drug-like molecules that directly modulate the activity of key clock proteins offer the potential to directly modulate the endogenous circadian rhythm and treat diseases associated with clock dysfunction. Here we demonstrate that synthetic ligands targeting a key component of the mammalian clock, the nuclear receptors REV-ERBα and β, regulate sleep architecture and emotional behaviour in mice. REV-ERB agonists induce wakefulness and reduce REM and slow-wave sleep. Interestingly, REV-ERB agonists also reduce anxiety-like behaviour. These data are consistent with increased anxiety-like behaviour of REV-ERBβ-null mice, in which REV-ERB agonists have no effect. These results indicate that pharmacological targeting of REV-ERB may lead to the development of novel therapeutics to treat sleep disorders and anxiety.
Collapse
MESH Headings
- ARNTL Transcription Factors/genetics
- ARNTL Transcription Factors/metabolism
- Animals
- Anxiety/drug therapy
- Anxiety/genetics
- Anxiety/metabolism
- Anxiety/physiopathology
- Behavior, Animal/drug effects
- CLOCK Proteins/genetics
- CLOCK Proteins/metabolism
- Circadian Clocks/drug effects
- Circadian Clocks/genetics
- Circadian Rhythm/genetics
- Cryptochromes/genetics
- Cryptochromes/metabolism
- Feedback, Physiological
- Gene Expression Regulation
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- Period Circadian Proteins/genetics
- Period Circadian Proteins/metabolism
- Pyrrolidines/pharmacology
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Repressor Proteins/agonists
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Reward
- Signal Transduction
- Sleep, REM/drug effects
- Thiophenes/pharmacology
Collapse
Affiliation(s)
- Subhashis Banerjee
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter FL 33458
| | - Yongjun Wang
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Laura A. Solt
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter FL 33458
| | - Kristine Griffett
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Melissa Kazantzis
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter FL 33458
| | - Ariadna Amador
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter FL 33458
| | - Bahaa M. El-Gendy
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter FL 33458
| | - Salvador Huitron-Resendiz
- Department of Molecular and Integrative Neurosciences, The Scripps Research Institute, La Jolla, CA 92037
| | - Amanda J. Roberts
- Department of Molecular and Integrative Neurosciences, The Scripps Research Institute, La Jolla, CA 92037
| | - Youseung Shin
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter FL 33458
| | - Theodore M. Kamenecka
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter FL 33458
| | - Thomas P. Burris
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
122
|
Wager TT, Chandrasekaran RY, Bradley J, Rubitski D, Berke H, Mente S, Butler T, Doran A, Chang C, Fisher K, Knafels J, Liu S, Ohren J, Marconi M, DeMarco G, Sneed B, Walton K, Horton D, Rosado A, Mead A. Casein kinase 1δ/ε inhibitor PF-5006739 attenuates opioid drug-seeking behavior. ACS Chem Neurosci 2014; 5:1253-65. [PMID: 25299732 DOI: 10.1021/cn500201x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Casein kinase 1 delta (CK1δ) and casein kinase 1 epsilon (CK1ε) inhibitors are potential therapeutic agents for a range of psychiatric disorders. The feasibility of developing a CNS kinase inhibitor has been limited by an inability to identify safe brain-penetrant compounds with high kinome selectivity. Guided by structure-based drug design, potent and selective CK1δ/ε inhibitors have now been identified that address this gap, through the design and synthesis of novel 4-[4-(4-fluorophenyl)-1-(piperidin-4-yl)-1H-imidazol-5-yl]pyrimidin-2-amine derivatives. PF-5006739 (6) possesses a desirable profile, with low nanomolar in vitro potency for CK1δ/ε (IC50 = 3.9 and 17.0 nM, respectively) and high kinome selectivity. In vivo, 6 demonstrated robust centrally mediated circadian rhythm phase-delaying effects in both nocturnal and diurnal animal models. Further, 6 dose-dependently attenuated opioid drug-seeking behavior in a rodent operant reinstatement model in animals trained to self-administer fentanyl. Collectively, our data supports further development of 6 as a promising candidate to test the hypothesis of CK1δ/ε inhibition in treating multiple indications in the clinic.
Collapse
Affiliation(s)
- Travis T. Wager
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | | | - Jenifer Bradley
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - David Rubitski
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Helen Berke
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Scot Mente
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Todd Butler
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Angela Doran
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Cheng Chang
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Katherine Fisher
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - John Knafels
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Shenping Liu
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jeff Ohren
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Michael Marconi
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - George DeMarco
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Blossom Sneed
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kevin Walton
- Division
of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, NIH, Rockville, Maryland 20852, United States
| | - David Horton
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amy Rosado
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andy Mead
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
123
|
Blancas A, González-García S, Rodríguez K, Escobar C. Progressive anticipation in behavior and brain activation of rats exposed to scheduled daily palatable food. Neuroscience 2014; 281:44-53. [DOI: 10.1016/j.neuroscience.2014.09.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/11/2014] [Accepted: 09/12/2014] [Indexed: 01/09/2023]
|
124
|
Søvik E, Even N, Radford CW, Barron AB. Cocaine affects foraging behaviour and biogenic amine modulated behavioural reflexes in honey bees. PeerJ 2014; 2:e662. [PMID: 25405075 PMCID: PMC4232840 DOI: 10.7717/peerj.662] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/20/2014] [Indexed: 11/20/2022] Open
Abstract
In humans and other mammals, drugs of abuse alter the function of biogenic amine pathways in the brain leading to the subjective experience of reward and euphoria. Biogenic amine pathways are involved in reward processing across diverse animal phyla, however whether cocaine acts on these neurochemical pathways to cause similar rewarding behavioural effects in animal phyla other than mammals is unclear. Previously, it has been shown that bees are more likely to dance (a signal of perceived reward) when returning from a sucrose feeder after cocaine treatment. Here we examined more broadly whether cocaine altered reward-related behaviour, and biogenic amine modulated behavioural responses in bees. Bees developed a preference for locations at which they received cocaine, and when foraging at low quality sucrose feeders increase their foraging rate in response to cocaine treatment. Cocaine also increased reflexive proboscis extension to sucrose, and sting extension to electric shock. Both of these simple reflexes are modulated by biogenic amines. This shows that systemic cocaine treatment alters behavioural responses that are modulated by biogenic amines in insects. Since insect reward responses involve both octopamine and dopamine signalling, we conclude that cocaine treatment altered diverse reward-related aspects of behaviour in bees. We discuss the implications of these results for understanding the ecology of cocaine as a plant defence compound. Our findings further validate the honey bee as a model system for understanding the behavioural impacts of cocaine, and potentially other drugs of abuse.
Collapse
Affiliation(s)
- Eirik Søvik
- Department of Biological Sciences, Macquarie University , Sydney , Australia ; Department of Biology, Washington University in St. Louis , St. Louis , USA
| | - Naïla Even
- Department of Biological Sciences, Macquarie University , Sydney , Australia
| | - Catherine W Radford
- Department of Biological Sciences, Macquarie University , Sydney , Australia
| | - Andrew B Barron
- Department of Biological Sciences, Macquarie University , Sydney , Australia
| |
Collapse
|
125
|
Li SX, Wei YM, Shi HS, Luo YX, Ding ZB, Xue YX, Lu L, Yu CX. Glycogen synthase kinase-3β in the ventral tegmental area mediates diurnal variations in cocaine-induced conditioned place preference in rats. Addict Biol 2014; 19:996-1005. [PMID: 23750993 DOI: 10.1111/adb.12068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cocaine sensitization and reward are reported to be under the influence of diurnal rhythm. However, no previous studies have reported brain areas that play a role as modulators and underlie the mechanism of diurnal variations in cocaine reward. We examined (1) the diurnal rhythm of glycogen synthase kinase-3β (GSK-3β) activity in the suprachiasmatic nucleus (SCN) and reward-related brain areas in naive rats; (2) the effect of day and night on the acquisition of cocaine-induced conditioned place preference (CPP); (3) the influence of cocaine-induced CPP on GSK-3β activity in the SCN and reward-related brain areas; and (4) the effect of the GSK-3β inhibitor SB216763 microinjected bilaterally into the ventral tegmental area (VTA) on cocaine-induced CPP. A significant diurnal rhythm of GSK-3β activity was found in the SCN and reward-related brain areas, with diurnal variations in cocaine-induced CPP. GSK-3β activity in the SCN and reward-related brain areas exhibited marked diurnal variations in rats treated with saline. GSK-3β activity in rats treated with cocaine exhibited distinct diurnal variations only in the prefrontal cortex and VTA. Cocaine decreased the expression of phosphorylated GSK-3β (i.e. increased GSK-3β activity) only in the VTA in rats trained and tested at ZT4 and ZT16. SB216763 microinjected into the VTA bilaterally eliminated the diurnal variations in cocaine-induced CPP, but did not affect the acquisition of cocaine-induced CPP. These findings suggest that the VTA may be a critical area involved in the diurnal variations in cocaine-induced CPP, and GSK-3β may be a regulator of diurnal variations in cocaine-induced CPP.
Collapse
Affiliation(s)
- Su-Xia Li
- National Institute on Drug Dependence, Peking University, China
| | | | | | | | | | | | | | | |
Collapse
|
126
|
Al-Safadi S, Al-Safadi A, Branchaud M, Rutherford S, Dayanandan A, Robinson B, Amir S. Stress-induced changes in the expression of the clock protein PERIOD1 in the rat limbic forebrain and hypothalamus: role of stress type, time of day, and predictability. PLoS One 2014; 9:e111166. [PMID: 25338089 PMCID: PMC4206498 DOI: 10.1371/journal.pone.0111166] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/25/2014] [Indexed: 11/23/2022] Open
Abstract
Stressful events can disrupt circadian rhythms in mammals but mechanisms underlying this disruption remain largely unknown. One hypothesis is that stress alters circadian protein expression in the forebrain, leading to functional dysregulation of the brain circadian network and consequent disruption of circadian physiological and behavioral rhythms. Here we characterized the effects of several different stressors on the expression of the core clock protein, PER1 and the activity marker, FOS in select forebrain and hypothalamic nuclei in rats. We found that acute exposure to processive stressors, restraint and forced swim, elevated PER1 and FOS expression in the paraventricular and dorsomedial hypothalamic nuclei and piriform cortex but suppressed PER1 and FOS levels exclusively in the central nucleus of the amygdala (CEAl) and oval nucleus of the bed nucleus of the stria terminalis (BNSTov). Conversely, systemic stressors, interleukin-1β and 2-Deoxy-D-glucose, increased PER1 and FOS levels in all regions studied, including the CEAl and BNSTov. PER1 levels in the suprachiasmatic nucleus (SCN), the master pacemaker, were unaffected by any of the stress manipulations. The effect of stress on PER1 and FOS was modulated by time of day and, in the case of daily restraint, by predictability. These results demonstrate that the expression of PER1 in the forebrain is modulated by stress, consistent with the hypothesis that PER1 serves as a link between stress and the brain circadian network. Furthermore, the results show that the mechanisms that control PER1 and FOS expression in CEAl and BNSTov are uniquely sensitive to differences in the type of stressor. Finally, the finding that the effect of stress on PER1 parallels its effect on FOS supports the idea that Per1 functions as an immediate-early gene. Our observations point to a novel role for PER1 as a key player in the interface between stress and circadian rhythms.
Collapse
Affiliation(s)
- Sherin Al-Safadi
- Department of Biology, Concordia University, Montréal, Quebéc, Canada
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montréal, Quebéc, Canada
| | - Aya Al-Safadi
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montréal, Quebéc, Canada
| | - Marie Branchaud
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montréal, Quebéc, Canada
| | - Spencer Rutherford
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montréal, Quebéc, Canada
| | - Arun Dayanandan
- Department of Biology, Concordia University, Montréal, Quebéc, Canada
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montréal, Quebéc, Canada
| | - Barry Robinson
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montréal, Quebéc, Canada
| | - Shimon Amir
- Department of Biology, Concordia University, Montréal, Quebéc, Canada
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montréal, Quebéc, Canada
- * E-mail:
| |
Collapse
|
127
|
Stowie AC, Amicarelli MJ, Prosser RA, Glass JD. Chronic cocaine causes long-term alterations in circadian period and photic entrainment in the mouse. Neuroscience 2014; 284:171-179. [PMID: 25301751 DOI: 10.1016/j.neuroscience.2014.08.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 11/17/2022]
Abstract
The disruptive effects of cocaine on physiological, behavioral and genetic processes are well established. However, few studies have focused on the actions of cocaine on the adult circadian timekeeping system, and none have explored the circadian implications of long-term (weeks to months) cocaine exposure. The present study was undertaken to explore the actions of such long-term cocaine administration on core circadian parameters in mice, including rhythm period, length of the nocturnal activity period and photic entrainment. For cocaine dosing over extended periods, cocaine was provided in drinking water using continuous and scheduled regimens. The impact of chronic cocaine on circadian regulation was evidenced by disruptions of the period of circadian entrainment and intrinsic free-running circadian period. Specifically, mice under a skeleton photoperiod (1-min pulse of dim light delivered daily) receiving continuous ad libitum cocaine entrained rapidly to the light pulse at activity onset. Conversely, water controls entrained more slowly at activity offset through a process of phase-delays, which resulted in their activity rhythms being entrained 147° out of phase with the cocaine group. This pattern persisted after cocaine withdrawal. Next, mice exposed to scheduled daily cocaine presentations exhibited free-running periods under constant darkness that were significantly longer than water controls and which also persisted after cocaine withdrawal. These cocaine-induced perturbations of clock timing could produce chronic psychological and physiological stress, contributing to increased cocaine use and dependence.
Collapse
Affiliation(s)
- A C Stowie
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - M J Amicarelli
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - R A Prosser
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - J D Glass
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA.
| |
Collapse
|
128
|
Mendoza J, Challet E. Circadian insights into dopamine mechanisms. Neuroscience 2014; 282:230-42. [PMID: 25281877 DOI: 10.1016/j.neuroscience.2014.07.081] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 07/22/2014] [Accepted: 07/24/2014] [Indexed: 01/11/2023]
Abstract
Almost every physiological or behavioral process in mammals follows rhythmic patterns, which depend mainly on a master circadian clock located in the hypothalamic suprachiasmatic nucleus (SCN). The dopaminergic (DAergic) system in the brain is principally implicated in motor functions, motivation and drug intake. Interestingly, DA-related parameters and behaviors linked to the motivational and arousal states, show daily rhythms that could be regulated by the SCN or by extra-SCN circadian oscillator(s) modulating DAergic systems. Here we examine what is currently understood about the anatomical and functional central multi-oscillatory circadian system, highlighting how the main SCN clock communicates timing information with other brain clocks to regulate the DAergic system and conversely, how DAergic cues may have feedback effects on the SCN. These studies give new insights into the role of the brain circadian system in DA-related neurologic pathologies, such as Parkinson's disease, attention deficit/hyperactive disorder and drug addiction.
Collapse
Affiliation(s)
- J Mendoza
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, University of Strasbourg, 5 rue Blaise Pascal, 67084 Strasbourg cedex, France.
| | - E Challet
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, University of Strasbourg, 5 rue Blaise Pascal, 67084 Strasbourg cedex, France
| |
Collapse
|
129
|
Landgraf D, McCarthy MJ, Welsh DK. Circadian clock and stress interactions in the molecular biology of psychiatric disorders. Curr Psychiatry Rep 2014; 16:483. [PMID: 25135782 DOI: 10.1007/s11920-014-0483-7] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Many psychiatric disorders are characterized by circadian rhythm abnormalities, including disturbed sleep/wake cycles, changes in locomotor activity, and abnormal endocrine function. Animal models with mutations in circadian "clock genes" commonly show disturbances in reward processing, locomotor activity and novelty seeking behaviors, further supporting the idea of a connection between the circadian clock and psychiatric disorders. However, if circadian clock dysfunction is a common risk factor for multiple psychiatric disorders, it is unknown if and how these putative clock abnormalities could be expressed differently, and contribute to multiple, distinct phenotypes. One possible explanation is that the circadian clock modulates the biological responses to stressful environmental factors that vary with an individual's experience. It is known that the circadian clock and the stress response systems are closely related: Circadian clock genes regulate the physiological sensitivity to and rhythmic release of glucocorticoids (GC). In turn, GCs have reciprocal effects on the clock. Since stressful life events or increased vulnerability to stress are risk factors for multiple psychiatric disorders, including post-traumatic stress disorder (PTSD), attention deficit hyperactivity disorder (ADHD), bipolar disorder (BD), major depressive disorder (MDD), alcohol use disorder (AUD) and schizophrenia (SCZ), we propose that modulation of the stress response is a common mechanism by which circadian clock genes affect these illnesses. Presently, we review how molecular components of the circadian clock may contribute to these six psychiatric disorders, and present the hypothesis that modulation of the stress response may constitute a common mechanism by which the circadian clock affects multiple psychiatric disorders.
Collapse
Affiliation(s)
- Dominic Landgraf
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | | | | |
Collapse
|
130
|
Sun Y, Meng S, Li J, Shi J, Lu L. Advances in genetic studies of substance abuse in China. SHANGHAI ARCHIVES OF PSYCHIATRY 2014; 25:199-211. [PMID: 24991158 PMCID: PMC4054556 DOI: 10.3969/j.issn.1002-0829.2013.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Summary The importance of genetic factors in substance addiction has long been established. The rationale for this work is that understanding of the function of addiction genes and delineation of the key molecular pathways of these genes would enhance the development of novel therapeutic targets and biomarkers that could be used in the prevention and management of substance abuse. Over the past few years, there has been a substantial increase in the number of genetic studies conducted on addiction in China; these studies have primarily focused on heroin, alcohol, and nicotine dependence. Most studies of candidate genes have concentrated on the dopamine, opioid, and serotonin systems. A number of genes associated with substance abuse in Caucasians are also risk factors in Chinese, but several novel genes and genetic risk factors associated with substance abuse in Chinese subjects have also been identified. This paper reviews the genetic studies of substance abuse performed by Chinese researchers. Genotypes and alleles related to addictive behavior in Chinese individuals are discussed and the contributions of Chinese researchers to the international corpus of knowledge about the genetic understanding of substance abuse are described.
Collapse
Affiliation(s)
- Yan Sun
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jiali Li
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lin Lu
- Institute of Mental Health, Peking University, Beijing, China
| |
Collapse
|
131
|
Prosser RA, Stowie A, Amicarelli M, Nackenoff AG, Blakely RD, Glass JD. Cocaine modulates mammalian circadian clock timing by decreasing serotonin transport in the SCN. Neuroscience 2014; 275:184-93. [PMID: 24950119 DOI: 10.1016/j.neuroscience.2014.06.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 12/15/2022]
Abstract
Cocaine abuse disrupts reward and homeostatic processes through diverse processes, including those involved in circadian clock regulation. Recently we showed that cocaine administration to mice disrupts nocturnal photic phase resetting of the suprachiasmatic (SCN) circadian clock, whereas administration during the day induces non-photic phase shifts. Importantly, the same effects are seen when cocaine is applied to the SCN in vitro, where it blocks photic-like (glutamate-induced) phase shifts at night and induces phase advances during the day. Furthermore, our previous data suggest that cocaine acts in the SCN by enhancing 5-HT signaling. For example, the in vitro actions of cocaine mimic those of 5-HT and are blocked by the 5-HT antagonist, metergoline, but not the dopamine receptor antagonist, fluphenazine. Although our data are consistent with cocaine acting through enhanced 5-HT signaling, the nonselective actions of cocaine as an antagonist of monoamine transporters raises the question of whether inhibition of the 5-HT transporter (SERT) is key to its circadian effects. Here we investigate this issue using transgenic mice expressing a SERT that exhibits normal 5-HT recognition and transport but significantly reduced cocaine potency (SERT Met172). Circadian patterns of SCN behavioral and neuronal activity did not differ between wild-type (WT) and SERT Met172 mice, nor did they differ in the ability of the 5-HT1A,2,7 receptor agonist, 8-OH-DPAT to reset SCN clock phase, consistent with the normal SERT expression and activity in the transgenic mice. However, (1) cocaine administration does not induce phase advances when administered in vivo or in vitro in SERT Met172 mice; (2) cocaine does not block photic or glutamate-induced phase shifts in SERT Met172 mice; and (3) cocaine does not induce long-term changes in free-running period in SERT Met172 mice. We conclude that SERT antagonism is required for the phase shifting of the SCN circadian clock induced by cocaine.
Collapse
Affiliation(s)
- R A Prosser
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA.
| | - A Stowie
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - M Amicarelli
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - A G Nackenoff
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA; Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - R D Blakely
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA; Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - J D Glass
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
132
|
Clough SJ, Hutchinson AJ, Hudson RL, Dubocovich ML. Genetic deletion of the MT1 or MT2 melatonin receptors abrogates methamphetamine-induced reward in C3H/HeN mice. Physiol Behav 2014; 132:79-86. [PMID: 24813704 DOI: 10.1016/j.physbeh.2014.04.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 03/26/2014] [Accepted: 04/29/2014] [Indexed: 01/09/2023]
Abstract
The drug of abuse methamphetamine (METH) is known for its ability to enhance reward responses. The rewarding properties of psychostimulants have been shown to vary across time of day in mice. The goal of this study was to determine the role of the MT1 and MT2 melatonin receptors in METH-induced reward, as measured by the conditioned place preference (CPP) paradigm during the light and dark phases. C3H/HeN wild-type mice were trained for METH-induced CPP at either ZT 6-8 (ZT: Zeitgeber time; ZT 0=lights on), when endogenous melatonin levels are low, or ZT 19-21, when melatonin levels are high. These time points also correspond to the high and low points for expression of the circadian gene Period1, respectively. The locomotor response to METH (1.2mg/kg, ip) treatment was of similar magnitude at both times; however only C3H/HeN mice conditioned to METH at ZT 6-8 developed a place preference. C3H/HeN mice with a genetic deletion of either the MT1 (MT1KO) or MT2 (MT2KO) receptor tested at ZT 6-8 or ZT 19-21 did not develop a place preference for METH, though both showed a similar increase in locomotor activity following METH treatment when compared to wild-type mice. We conclude that in our mouse model METH-induced CPP is dependent on time of day and the presence of the MT1 or MT2 receptors, suggesting a role for melatonin in METH-induced reward.
Collapse
Affiliation(s)
- Shannon J Clough
- Department of Pharmacology & Toxicology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, United States
| | - Anthony J Hutchinson
- Department of Pharmacology & Toxicology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, United States
| | - Randall L Hudson
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, United States
| | - Margarita L Dubocovich
- Department of Pharmacology & Toxicology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, United States.
| |
Collapse
|
133
|
Logan RW, Williams WP, McClung CA. Circadian rhythms and addiction: mechanistic insights and future directions. Behav Neurosci 2014; 128:387-412. [PMID: 24731209 DOI: 10.1037/a0036268] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Circadian rhythms are prominent in many physiological and behavioral functions. Circadian disruptions either by environmental or molecular perturbation can have profound health consequences, including the development and progression of addiction. Both animal and humans studies indicate extensive bidirectional relationships between the circadian system and drugs of abuse. Addicted individuals display disrupted rhythms, and chronic disruption or particular chronotypes may increase the risk for substance abuse and relapse. Moreover, polymorphisms in circadian genes and an evening chronotype have been linked to mood and addiction disorders, and recent efforts suggest an association with the function of reward neurocircuitry. Animal studies are beginning to determine how altered circadian gene function results in drug-induced neuroplasticity and behaviors. Many studies suggest a critical role for circadian rhythms in reward-related pathways in the brain and indicate that drugs of abuse directly affect the central circadian pacemaker. In this review, we highlight key findings demonstrating the importance of circadian rhythms in addiction and how future studies will reveal important mechanistic insights into the involvement of circadian rhythms in drug addiction.
Collapse
Affiliation(s)
- Ryan W Logan
- Department of Psychiatry, University of Pittsburgh School of Medicine
| | - Wilbur P Williams
- Department of Psychiatry, University of Pittsburgh School of Medicine
| | - Colleen A McClung
- Department of Psychiatry, University of Pittsburgh School of Medicine
| |
Collapse
|
134
|
Rawashdeh O, Jilg A, Jedlicka P, Slawska J, Thomas L, Saade A, Schwarzacher SW, Stehle JH. PERIOD1 coordinates hippocampal rhythms and memory processing with daytime. Hippocampus 2014; 24:712-23. [DOI: 10.1002/hipo.22262] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/26/2014] [Accepted: 02/13/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Oliver Rawashdeh
- Institute of Cellular and Molecular Anatomy, Dr. Senckenbergische Anatomie; Goethe-University; Theodor-Stern-Kai 7 Frankfurt Germany
| | - Antje Jilg
- Institute of Cellular and Molecular Anatomy, Dr. Senckenbergische Anatomie; Goethe-University; Theodor-Stern-Kai 7 Frankfurt Germany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Dr. Senckenbergische Anatomie; Goethe-University; Theodor-Stern-Kai 7 Frankfurt Germany
| | - Jolanta Slawska
- Institute of Cellular and Molecular Anatomy, Dr. Senckenbergische Anatomie; Goethe-University; Theodor-Stern-Kai 7 Frankfurt Germany
| | - Lukas Thomas
- Institute of Cellular and Molecular Anatomy, Dr. Senckenbergische Anatomie; Goethe-University; Theodor-Stern-Kai 7 Frankfurt Germany
| | - Anastasia Saade
- Institute of Cellular and Molecular Anatomy, Dr. Senckenbergische Anatomie; Goethe-University; Theodor-Stern-Kai 7 Frankfurt Germany
| | - Stephan W. Schwarzacher
- Institute of Clinical Neuroanatomy, Dr. Senckenbergische Anatomie; Goethe-University; Theodor-Stern-Kai 7 Frankfurt Germany
| | - Jörg H. Stehle
- Institute of Cellular and Molecular Anatomy, Dr. Senckenbergische Anatomie; Goethe-University; Theodor-Stern-Kai 7 Frankfurt Germany
| |
Collapse
|
135
|
Bi J, Gelernter J, Sun J, Kranzler HR. Comparing the utility of homogeneous subtypes of cocaine use and related behaviors with DSM-IV cocaine dependence as traits for genetic association analysis. Am J Med Genet B Neuropsychiatr Genet 2014; 165B:148-56. [PMID: 24339190 PMCID: PMC4152729 DOI: 10.1002/ajmg.b.32216] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 11/20/2013] [Indexed: 11/08/2022]
Abstract
Because DSM-IV cocaine dependence (CD) is heterogeneous, it is not an optimal phenotype to identify genetic variation contributing to risk for cocaine use and related behaviors (CRBs). We used a cluster analytic method to differentiate homogeneous, highly heritable subtypes of CRBs and to compare their utility with that of the DSM-IV CD as traits for genetic association analysis. Clinical features of CRBs and co-occurring disorders were obtained via a poly-diagnostic interview administered to 9,965 participants in genetic studies of substance dependence. A subsample of subjects (N = 3,443) were genotyped for 1,350 single nucleotide polymorphisms (SNPs) selected from 130 candidate genes related to addiction. Cluster analysis of clinical features of the sample yielded five subgroups, two of which were characterized by heavy cocaine use and high heritability: a heavy cocaine use, infrequent intravenous injection group and an early-onset, heavy cocaine use, high comorbidity group. The utility of these traits was compared with the CD diagnosis through association testing of 2,320 affected subjects and 480 cocaine-exposed controls. Analyses examined both single SNP (main) and SNP-SNP interaction (epistatic) effects, separately for African-Americans and European-Americans. The two derived subtypes showed more significant P values for 6 of 8 main effects and 7 of 8 epistatic effects. Variants in the CLOCK gene were significantly associated with the heavy cocaine use, infrequent intravenous injection group, but not with the DSM-IV diagnosis of CD. These results support the utility of subtypes based on CRBs to detect risk variants for cocaine addiction.
Collapse
Affiliation(s)
- Jinbo Bi
- Department of Computer Science and Engineering, School of Engineering, University of Connecticut, Storrs, Connecticut
| | - Joel Gelernter
- Departments of Psychiatry, Genetics, and Neurobiology, Yale University School of Medicine, New Haven, Connecticut,Department of Psychiatry, VA CT Healthcare Center, West Haven, Connecticut
| | - Jiangwen Sun
- Department of Computer Science and Engineering, School of Engineering, University of Connecticut, Storrs, Connecticut
| | - Henry R. Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania,VISN 4 Mental Illness Research, Education, and Clinical Center, Philadelphia, Pennsylvania,Correspondence to: Henry R. Kranzler, M.D., Department of Psychiatry, Treatment Research Center, University of Pennsylvania Perelman School of Medicine, 3900 Chestnut Street, Philadelphia, PA 19104-6178.
| |
Collapse
|
136
|
Sidor MM, McClung CA. Timing matters: using optogenetics to chronically manipulate neural circuitry and rhythms. Front Behav Neurosci 2014; 8:41. [PMID: 24592222 PMCID: PMC3924037 DOI: 10.3389/fnbeh.2014.00041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/27/2014] [Indexed: 11/19/2022] Open
Abstract
The ability to probe defined neural circuits with both the spatial and temporal resolution imparted by optogenetics has transformed the field of neuroscience. Although much attention has been paid to the advantages of manipulating neural activity at millisecond timescales in order to elicit time-locked neural responses, little consideration has been given to the manipulation of circuit activity at physiologically relevant times of day, across multiple days. Nearly all biological events are governed by the circadian clock and exhibit 24 h rhythms in activity. Indeed, neural circuit activity itself exhibits a daily rhythm with distinct temporal peaks in activity occurring at specific times of the day. Therefore, experimentally probing circuit function within and across physiologically relevant time windows (minutes to hours) in behaving animals is fundamental to understanding the function of any one particular circuit within the intact brain. Furthermore, understanding how circuit function changes with repeated manipulation is important for modeling the circuit-wide disruptions that occur with chronic disease states. Here, we review recent advances in optogenetic technology that allow for chronic, temporally specific, control of circuit activity and provide examples of chronic optogenetic paradigms that have been utilized in the search for the neural circuit basis of behaviors relevant to human neuropsychiatric disease.
Collapse
Affiliation(s)
- Michelle M Sidor
- 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
|
137
|
Golombek DA, Bussi IL, Agostino PV. Minutes, days and years: molecular interactions among different scales of biological timing. Philos Trans R Soc Lond B Biol Sci 2014; 369:20120465. [PMID: 24446499 DOI: 10.1098/rstb.2012.0465] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Biological clocks are genetically encoded oscillators that allow organisms to keep track of their environment. Among them, the circadian system is a highly conserved timing structure that regulates several physiological, metabolic and behavioural functions with periods close to 24 h. Time is also crucial for everyday activities that involve conscious time estimation. Timing behaviour in the second-to-minutes range, known as interval timing, involves the interaction of cortico-striatal circuits. In this review, we summarize current findings on the neurobiological basis of the circadian system, both at the genetic and behavioural level, and also focus on its interactions with interval timing and seasonal rhythms, in order to construct a multi-level biological clock.
Collapse
Affiliation(s)
- Diego A Golombek
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, , Roque Sáenz Peña 352, Bernal, Buenos Aires B1876BXD, Argentina
| | | | | |
Collapse
|
138
|
Hutchinson AJ, Ma J, Liu J, Hudson RL, Dubocovich ML. Role of MT1 melatonin receptors in methamphetamine-induced locomotor sensitization in C57BL/6 mice. Psychopharmacology (Berl) 2014; 231:257-67. [PMID: 23934259 PMCID: PMC4696604 DOI: 10.1007/s00213-013-3228-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 07/26/2013] [Indexed: 02/07/2023]
Abstract
RATIONALE Melatonin modifies physiological and behavioral responses to psychostimulants, with the MT₁ and MT₂ melatonin receptors specifically implicated in facilitating methamphetamine (METH)-induced sensitization in melatonin-proficient mice. OBJECTIVE The objective of the study is to assess differences in locomotor sensitization after a single dose of methamphetamine in low-melatonin-expressing C57BL/6 wild-type and MT₁ receptor knockout (MT₁KO) mice, comparing with melatonin-expressing C3H/HeN mice. METHODS Mice received a vehicle or methamphetamine (1.2 mg/kg, i.p.) pretreatment (day 1) during the light (ZT5-9) or dark (ZT 19-21) periods in novel test arenas. Locomotor sensitization was assessed by methamphetamine challenge after an eight-day abstinence (day 9). TH protein expression was evaluated by immunofluorescence and Western blot analysis. RESULTS Methamphetamine pretreatment induced statistically significant locomotor sensitization upon challenge after eight-day abstinence in C3H and C57 wild-type mice during the light period. The magnitude of sensitization in C57 mice was diminished in the dark period and completely abrogated in MT₁KO mice. No differences were observed in tyrosine hydroxylase immunoreactivity in the mesolimbic dopamine system. Additional exposures to the test arenas after methamphetamine pretreatment (nights 2-6) enhanced sensitization. CONCLUSIONS Deletion of the MT₁ melatonin receptor abolishes sensitization induced by a single METH pretreatment. The magnitude of sensitization is also altered by time of day and contextual cues. We conclude that the MT₁ melatonin receptor is emerging as a novel target of therapeutic intervention for drug abuse disorders.
Collapse
Affiliation(s)
- Anthony J. Hutchinson
- Department of Pharmacology & Toxicology, School of Medicine and Biomedical Sciences, 3435 Main Street, University at Buffalo, Buffalo, NY 14221
| | - Jason Ma
- Department of Pharmacology & Toxicology, School of Medicine and Biomedical Sciences, 3435 Main Street, University at Buffalo, Buffalo, NY 14221
| | - Jiabei Liu
- Department of Pharmacology & Toxicology, School of Medicine and Biomedical Sciences, 3435 Main Street, University at Buffalo, Buffalo, NY 14221
| | - Randall L. Hudson
- Department of Physiology & Biophysics, School of Medicine and Biomedical Sciences, 3435 Main Street, University at Buffalo, Buffalo, NY 14221
| | - Margarita L. Dubocovich
- Department of Pharmacology & Toxicology, School of Medicine and Biomedical Sciences, 3435 Main Street, University at Buffalo, Buffalo, NY 14221
- Corresponding Author. Margarita L. Dubocovich, Ph.D., Department of Pharmacology & Toxicology, School of Medicine and Biomedical Sciences, 3435 Main Street (Farber Hall 102), University at Buffalo; Buffalo, NY 14221, Phone: 1-716-829-3048, Fax: 1-716-829-2801,
| |
Collapse
|
139
|
Unterwald EM, Page ME, Brown TB, Miller JS, Ruiz M, Pescatore KA, Xu B, Reichardt LF, Beverley J, Tang B, Steiner H, Thomas EA, Ehrlich ME. Behavioral and transcriptome alterations in male and female mice with postnatal deletion of TrkB in dorsal striatal medium spiny neurons. Mol Neurodegener 2013; 8:47. [PMID: 24369067 PMCID: PMC3880973 DOI: 10.1186/1750-1326-8-47] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/19/2013] [Indexed: 01/05/2023] Open
Abstract
Background The high affinity tyrosine kinase receptor, TrkB, is the primary receptor for brain derived neurotrophic factor (BDNF) and plays an important role in development, maintenance and plasticity of the striatal output medium size spiny neuron. The striatal BDNF/TrkB system is thereby implicated in many physiologic and pathophysiologic processes, the latter including mood disorders, addiction, and Huntington’s disease. We crossed a mouse harboring a transgene directing cre-recombinase expression primarily to postnatal, dorsal striatal medium spiny neurons, to a mouse containing a floxed TrkB allele (fB) mouse designed for deletion of TrkB to determine its role in the adult striatum. Results We found that there were sexually dimorphic alterations in behaviors in response to stressful situations and drugs of abuse. Significant sex and/or genotype differences were found in the forced swim test of depression-like behaviors, anxiety-like behaviors on the elevated plus maze, and cocaine conditioned reward. Microarray analysis of dorsal striatum revealed significant dysregulation in individual and groups of genes that may contribute to the observed behavioral responses and in some cases, represent previously unidentified downstream targets of TrkB. Conclusions The data point to a set of behaviors and changes in gene expression following postnatal deletion of TrkB in the dorsal striatum distinct from those in other brain regions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Michelle E Ehrlich
- Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
140
|
The ClockΔ19 mutation in mice fails to alter the primary and secondary reinforcing properties of nicotine. Drug Alcohol Depend 2013; 133:733-9. [PMID: 24054990 DOI: 10.1016/j.drugalcdep.2013.08.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 08/27/2013] [Accepted: 08/27/2013] [Indexed: 01/26/2023]
Abstract
BACKGROUND Clock genes have been demonstrated to play a role in behavioral responses to a variety of drugs of abuse, including cocaine, amphetamine, morphine, and ethanol. However, no studies to date have examined the role of Clock genes on nicotine-mediated behaviors. We examined the involvement of Clock, one of several Clock genes, on the effects of nicotine by examining mice with the ClockΔ19 mutation in behaviors commonly used to assess drug effects in rodents. METHODS We first measured the locomotor effects of nicotine in mutants and wild type mice in response to repeated nicotine injections (0.175 mg/kg, IP). To assess the secondary properties of nicotine, we measured the ability of nicotine (0.175 mg/kg, IP) to induce a conditioned place preference. Finally, we measured the primary reinforcing properties of nicotine at two doses (0.01 and 0.03 mg/kg/infusion, IV) using the self-administration paradigm. RESULTS Mutant mice demonstrated no difference in magnitude of the sensitized response to nicotine as compared to wild-type controls. In the conditioned place preference paradigm, mutant and wild-type mice demonstrated a similar preference for a nicotine-paired environment. And finally, mutant and wild-type mice demonstrated a similar acquisition of nicotine self-administration, as indicated by the number of responses on a nicotine-paired lever and the number of nicotine reinforcers achieved during sessions. CONCLUSIONS The ClockΔ19 mutation appears to have no effect on the reinforcing properties of nicotine, in contrast to its demonstrated role in cocaine reinforcement. Further studies are needed to determine the effect of other Clock genes on nicotine reinforcement.
Collapse
|
141
|
Egr1 regulates lithium-induced transcription of the Period 2 (PER2) gene. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1969-79. [DOI: 10.1016/j.bbadis.2013.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 05/22/2013] [Accepted: 06/19/2013] [Indexed: 11/19/2022]
|
142
|
Baltazar RM, Coolen LM, Webb IC. Medial prefrontal cortex inactivation attenuates the diurnal rhythm in amphetamine reward. Neuroscience 2013; 258:204-10. [PMID: 24239716 DOI: 10.1016/j.neuroscience.2013.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 11/06/2013] [Accepted: 11/06/2013] [Indexed: 10/26/2022]
Abstract
Psychostimulant reward, as assessed via the conditioned place preference (CPP) paradigm, exhibits a daily rhythm with peaks in the late dark and early light periods, and a nadir near the light-to-dark transition. While this diurnal rhythm is correlated with neural activity in several corticolimbic structures, the brain regions mediating this behavioral rhythm remain unknown. Here, we examine the role of the ventral medial prefrontal cortex (mPFC). The effects of excitotoxic mPFC lesions on daily rhythms in amphetamine CPP were examined at previously observed peak (zeitgeber time [ZT] 23) and nadir times (ZT11). mPFC lesions encompassing the prelimbic and infralimbic subregions increased the CPP for amphetamine at the nadir time, thereby eliminating the daily rhythm in amphetamine reward. To examine the effects of transient mPFC inactivation, rats received intra-mPFC infusions of GABA receptor agonists during the acquisition or expression phases of CPP testing. Inactivation of the ventral mPFC at either of these phases also eliminated the daily rhythm in amphetamine-induced CPP via an increase in drug-paired chamber dwell time at the baseline nadir. Together, these results indicate that the ventral mPFC plays a critical role in mediating the diurnal rhythm in amphetamine CPP during both the acquisition and expression of learned reward-context associations. Moreover, as the loss of rhythmicity occurs via an increase at the nadir point, these results suggest that excitatory output from the ventral mPFC normally inhibits context-elicited reward seeking prior to the light-to-dark transition.
Collapse
Affiliation(s)
- R M Baltazar
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - L M Coolen
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - I C Webb
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
143
|
The role of clock in ethanol-related behaviors. Neuropsychopharmacology 2013; 38:2393-400. [PMID: 23722243 PMCID: PMC3799058 DOI: 10.1038/npp.2013.138] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 02/08/2023]
Abstract
Mice with a mutation in the Clock gene (ClockΔ19) exhibit increased preference for stimulant rewards and sucrose. They also have an increase in dopaminergic activity in the ventral tegmental area (VTA) and a general increase in glutamatergic tone that might underlie these behaviors. However, it is unclear if their phenotype would extend to a very different class of drug (ethanol), and if so, whether these systems might be involved in their response. Continuous access voluntary ethanol intake was evaluated in ClockΔ19 mutants and wild-type (WT) mice. We found that ClockΔ19 mice exhibited significantly increased ethanol intake in a two-bottle choice paradigm. Interestingly, this effect was more robust in female mice. Moreover, chronic ethanol experience resulted in a long-lasting decrease in VTA Clock expression. To determine the importance of VTA Clock expression in ethanol intake, we knocked down Clock expression in the VTA of WT mice via RNA interference. We found that reducing Clock expression in the VTA resulted in significantly increased ethanol intake similar to the ClockΔ19 mice. Interestingly, we also discovered that ClockΔ19 mice exhibit significantly augmented responses to the sedative effects of ethanol and ketamine, but not pentobarbital. However, their drinking behavior was not affected by acamprosate, an FDA-approved drug for the treatment of alcoholism, suggesting that their increased glutamatergic tone might underlie the increased sensitivity to the sedative/hypnotic properties of ethanol but not the rewarding properties of ethanol. Taken together, we have identified a significant role for Clock in the VTA as a negative regulator of ethanol intake and implicate the VTA dopamine system in this response.
Collapse
|
144
|
Pohl JB, Ghezzi A, Lew LK, Robles RB, Cormack L, Atkinson NS. Circadian genes differentially affect tolerance to ethanol in Drosophila. Alcohol Clin Exp Res 2013; 37:1862-71. [PMID: 23808628 PMCID: PMC4861224 DOI: 10.1111/acer.12173] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 03/29/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND There is a strong relationship between circadian rhythms and ethanol (EtOH) responses. EtOH consumption has been shown to disrupt physiological and behavioral circadian rhythms in mammals (Alcohol Clin Exp Res 2005b, 29, 1550). The Drosophila central circadian pacemaker is composed of proteins encoded by the per, tim, cyc, and Clk genes. Using Drosophila mutant analysis, we asked whether these central components of the circadian clock make the equivalent contribution toward EtOH tolerance and whether rhythmicity itself is necessary for tolerance. METHODS We tested flies carrying mutations in core clock genes for the capacity to acquire EtOH tolerance. Tolerance was assayed by comparing the sedation curves of populations during their first and second sedation. Animals that had acquired tolerance sedated more slowly. Movement was also monitored as the flies breathe the EtOH vapor to determine if other facets of the EtOH response were affected by the mutations. Gas chromatography was used to measure internal EtOH concentration. Constant light was used to nongenetically destabilize the PER and TIM proteins. RESULTS A group of circadian mutations, all of which eliminate circadian rhythms, do not disrupt tolerance identically. Mutations in per, tim, and cyc completely block tolerance. However, a mutation in Clk does not interfere with tolerance. Constant light also disrupts the capacity to acquire tolerance. These lines did not differ in EtOH absorption. CONCLUSIONS Mutations affecting different parts of the intracellular circadian clock can block the capacity to acquire rapid EtOH tolerance. However, the role of circadian genes in EtOH tolerance is independent of their role in producing circadian rhythmicity. The interference in the capacity to acquire EtOH tolerance by some circadian mutations is not merely a downstream effect of a nonfunctional circadian clock; instead, these circadian genes play an independent role in EtOH tolerance.
Collapse
Affiliation(s)
- Jascha B. Pohl
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, The University of Texas at Austin, 1 University Station C0920, Austin, Texas, 78712 USA
| | - Alfredo Ghezzi
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, The University of Texas at Austin, 1 University Station C0920, Austin, Texas, 78712 USA
| | - Linda K. Lew
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, The University of Texas at Austin, 1 University Station C0920, Austin, Texas, 78712 USA
| | - Roseanna B. Robles
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, The University of Texas at Austin, 1 University Station C0920, Austin, Texas, 78712 USA
| | - Lawrence Cormack
- Department of Psychology & Section of Neurobiology, The University of Texas at Austin, Texas 78712
| | - Nigel S. Atkinson
- The Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology, The University of Texas at Austin, 1 University Station C0920, Austin, Texas, 78712 USA
| |
Collapse
|
145
|
Søvik E, Barron AB. Invertebrate models in addiction research. BRAIN, BEHAVIOR AND EVOLUTION 2013; 82:153-65. [PMID: 24192516 DOI: 10.1159/000355506] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/03/2013] [Indexed: 11/19/2022]
Abstract
While drug addiction is a uniquely human problem, most research examining the biological mechanisms of the transition from substance use to addiction is conducted with vertebrate animal models. Many other fields of neuroscience have greatly benefitted from contributions from simple and manipulable invertebrate model systems. However, the potential of invertebrate research has yet to be fully capitalised on in the field of addiction neuroscience. This may be because of the complexity of addiction and the clinical imperative of addiction research. We argue that the homocentric diagnostic criteria of addiction are no more a hindrance to the use of invertebrate models than they are to vertebrate models. We highlight the strengths of the diversity of different invertebrate model systems in terms of neuroanatomy and molecular machinery, and stress that working with a range of different models will aid in understanding addiction and not be a disadvantage. Finally, we discuss the specific advantages of utilising invertebrate animals for addiction research and highlight key areas in which invertebrates are suited for making unique and meaningful contributions to this field.
Collapse
Affiliation(s)
- Eirik Søvik
- Department of Biological Sciences, Macquarie University, Sydney, N.S.W., Australia
| | | |
Collapse
|
146
|
De Bundel D, Gangarossa G, Biever A, Bonnefont X, Valjent E. Cognitive dysfunction, elevated anxiety, and reduced cocaine response in circadian clock-deficient cryptochrome knockout mice. Front Behav Neurosci 2013; 7:152. [PMID: 24187535 PMCID: PMC3807562 DOI: 10.3389/fnbeh.2013.00152] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 10/04/2013] [Indexed: 01/09/2023] Open
Abstract
The circadian clock comprises a set of genes involved in cell-autonomous transcriptional feedback loops that orchestrate the expression of a range of downstream genes, driving circadian patterns of behavior. Cognitive dysfunction, mood disorders, anxiety disorders, and substance abuse disorders have been associated with disruptions in circadian rhythm and circadian clock genes, but the causal relationship of these associations is still poorly understood. In the present study, we investigate the effect of genetic disruption of the circadian clock, through deletion of both paralogs of the core gene cryptochrome (Cry1 and Cry2). Mice lacking Cry1 and Cry2 (Cry1(-/-)Cry2(-/-) ) displayed attenuated dark phase and novelty-induced locomotor activity. Moreover, they showed impaired recognition memory but intact fear memory. Depression-related behaviors in the forced swim test or sucrose preference tests were unaffected but Cry1(-/-)Cry2(-/-) mice displayed increased anxiety in the open field and elevated plus maze tests. Finally, hyperlocomotion and striatal phosphorylation of extracellular signal-regulated kinase (ERK) induced by a single cocaine administration are strongly reduced in Cry1(-/-)Cry2(-/-) mice. Interestingly, only some behavioral measures were affected in mice lacking either Cry1 or Cry2. Notably, recognition memory was impaired in both Cry1(-/-)Cry2(+/+) and Cry1(+/+)Cry2(-/-) mice. Moreover, we further observed elevated anxiety in Cry1(-/-)Cry2(+/+) and Cry1(+/+)Cry2(-/-) mice. Our data indicate that beyond their role in the control of circadian rhythm, cryptochrome genes have a direct influence in cognitive function, anxiety-related behaviors and sensitivity to psychostimulant drugs.
Collapse
Affiliation(s)
- Dimitri De Bundel
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle Montpellier, France ; INSERM, U661 Montpellier, France ; Universités de Montpellier 1 and 2, UMR-5203 Montpellier, France
| | | | | | | | | |
Collapse
|
147
|
Adult female rats' altered diurnal locomotor activity pattern following chronic methylphenidate treatment. J Neural Transm (Vienna) 2013; 120:1717-31. [PMID: 23893293 DOI: 10.1007/s00702-013-1063-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 06/25/2013] [Indexed: 12/25/2022]
Abstract
Methylphenidate (MPD) is one of the most prescribed pharmacological agents, which is also used for cognitive enhancement and recreational purposes. The objective of this study was to investigate the repetitive dose-response effects of MPD on circadian rhythm of locomotor activity pattern of female WKY rats. The hypothesis is that a change in the circadian activity pattern indicates a long-lasting effect of the drug. Four animal groups (saline control, 0.6, 2.5, and 10.0 mg/kg MPD dose groups) were housed in a sound-controlled room at 12:12 light/dark cycle. All received saline injections on experimental day 1 (ED 1). On EDs 2-7, the control group received saline injection; the other groups received 0.6, 2.5, or 10.0 mg/kg MPD, respectively. On ED 8-10, injections were withheld. On ED 11, each group received the same dose as EDs 2-7. Hourly histograms and cosine statistical analyses calculating the acrophase (ϕ), amplitude (A), and MESOR (M) were applied to assess the 24-h circadian activity pattern. The 0.6 and 2.5 mg/kg MPD groups exhibited significant (p < 0.05) change in their circadian activity pattern on ED 11. The 10.0 mg/kg MPD group exhibited tolerance on ED 11 and also a significant change in activity pattern on ED 8 compared to ED 1, consistent with withdrawal behavior (p < 0.007). In conclusion, chronic MPD administration alters circadian locomotor activity of adult female WKY rats and confirms that chronic MPD use elicits long-lasting effects.
Collapse
|
148
|
Differential regulation of the period genes in striatal regions following cocaine exposure. PLoS One 2013; 8:e66438. [PMID: 23776671 PMCID: PMC3679086 DOI: 10.1371/journal.pone.0066438] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/09/2013] [Indexed: 11/19/2022] Open
Abstract
Several studies have suggested that disruptions in circadian rhythms contribute to the pathophysiology of multiple psychiatric diseases, including drug addiction. In fact, a number of the genes involved in the regulation of circadian rhythms are also involved in modulating the reward value for drugs of abuse, like cocaine. Thus, we wanted to determine the effects of chronic cocaine on the expression of several circadian genes in the Nucleus Accumbens (NAc) and Caudate Putamen (CP), regions of the brain known to be involved in the behavioral responses to drugs of abuse. Moreover, we wanted to explore the mechanism by which these genes are regulated following cocaine exposure. Here we find that after repeated cocaine exposure, expression of the Period (Per) genes and Neuronal PAS Domain Protein 2 (Npas2) are elevated, in a somewhat regionally selective fashion. Moreover, NPAS2 (but not CLOCK (Circadian Locomotor Output Cycles Kaput)) protein binding at Per gene promoters was enhanced following cocaine treatment. Mice lacking a functional Npas2 gene failed to exhibit any induction of Per gene expression after cocaine, suggesting that NPAS2 is necessary for this cocaine-induced regulation. Examination of Per gene and Npas2 expression over twenty-four hours identified changes in diurnal rhythmicity of these genes following chronic cocaine, which were regionally specific. Taken together, these studies point to selective disruptions in Per gene rhythmicity in striatial regions following chronic cocaine treatment, which are mediated primarily by NPAS2.
Collapse
|
149
|
Abstract
Increasingly invertebrates are being used to investigate the molecular and cellular effects of drugs of abuse to explore basic mechanisms of addiction. However, in mammals the principle factors contributing to addiction are long-term adaptive responses to repeated drug use. Here we examined whether adaptive responses to cocaine are also seen in invertebrates using the honey bee model system. Repeated topical treatment with a low dose of cocaine rendered bees resistant to the deleterious motor effects of a higher cocaine dose, indicating the development of physiological tolerance to cocaine in bees. Cocaine inhibits biogenic amine reuptake transporters, but neither acute nor repeated cocaine treatments caused measurable changes in levels of biogenic amines measured in whole bee brains. Our data show clear short and long-term behavioural responses of bees to cocaine administration, but caution that, despite the small size of the bee brain, measures of biogenic amines conducted at the whole-brain level may not reveal neurochemical effects of the drug.
Collapse
|
150
|
Gillman AG, Leffel JK, Kosobud AEK, Timberlake W. Behavioral characteristics and pharmacological manipulations of a nicotine-entrainable circadian oscillator. Chronobiol Int 2013; 30:855-69. [PMID: 23697901 DOI: 10.3109/07420528.2013.782314] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic daily administration of nicotine and other drugs of abuse has been found to entrain pre- and post-drug circadian locomotor activity episodes that oscillate on a 24-h schedule and persist for several days after administration ceases. This drug-entrainable oscillator system could conceivably lead to circadian rhythms of drug seeking and drug use in human drug addicts. The present study (1) characterizes the ability of daily nicotine administration to entrain circadian wheel-running activity episodes in rats across a range of doses, lighting schedules, and food access; and (2) tests whether pre- and post-nicotine episodes can be altered through pharmacological manipulations. Adult female rats were housed in wheel boxes for 35-60 d, and both wheel-running and feeding-related behaviors were measured continuously. Following acclimation, nicotine or saline was administered for 16-24 d, and the rats were left undisturbed for several test days to observe the persistence of nicotine-entrained activity. The results showed that nicotine dose-dependently entrains wheel-running activity, and the highest dose of 1.0 mg/kg produces robust pre- and post-nicotine circadian activity episodes under constant, fixed, and variable light/dark schedules. In the pharmacological manipulation experiment, nicotine-entrained rats were administered one of seven pharmacological treatments (varenicline, mecamylamine, acamprosate, topiramate, naltrexone, SB-334867, or bupropion) in place of the nicotine injection for 2 d, and the rats were not disturbed for four subsequent days. Most of the treatment drugs significantly reduced post-nicotine activity episodes, but only three treatments affected pre-nicotine episodes: the μ- and κ-opioid receptor antagonist naltrexone, the orexin-1 receptor antagonist SB-334867, and the AMPA (2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic acid)/kainate antagonist topiramate. These results show that chronic daily nicotine administration is a robust zeitgeber that dose-dependently entrains a nonphotic oscillator system that includes opioid, orexin, and glutamate pathways.
Collapse
Affiliation(s)
- Andrea G Gillman
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15219-3143, USA.
| | | | | | | |
Collapse
|