1
|
Ahmed T, Haque MA, Aunto TK, Hasan MK. Prevalence and associated risk factors of anxiety-induced sleep disturbance: Sex-stratified comparison among school-going adolescents in Paraguay. Sleep Med 2025; 127:91-99. [PMID: 39832431 DOI: 10.1016/j.sleep.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVES This study aimed to assess the prevalence and correlates of anxiety-induced sleep disturbance (AISD) in Paraguayan adolescents through sex-stratified analysis. METHODS This study used the cross-sectional data from Paraguay's 2017 Global School-Based Student Health Survey (GSHS). A total of 3149 in-school adolescents (12-17 years; 51.2 % female) were included in the final analysis. Anxiety-induced sleep disturbance (AISD) was evaluated using the survey question: "During the past 12 months, how often have you been so anxious about something that you could not sleep at night?" Participants could respond on a scale from 1 (never) to 5 (always). For analysis, responses were categorized as follows: "never/rarely/occasionally" indicated no AISD, while "often/always" indicated the presence of AISD. Age-adjusted univariate and multiple logistic analyses were used to determine the correlates of AISD. Regression analysis findings were presented using adjusted odds ratios (AOR) and 95 % confidence intervals (CI), with a p-value <0.05. RESULTS The overall prevalence of AISD was 9.5 % among the respondents, significantly higher in girls (12 %) than boys (6.7 %). Loneliness, suicidal ideation, not attending physical education classes, and sitting activities were significantly associated with respondents' AISD. In both sexes, those with loneliness and those who had suicidal ideation were more likely to suffer from AISD than their counterparts. Among girls, those who did not attend physical education class and those who engaged in sedentary activities for three hours or more had significantly higher odds of AISD than boys. CONCLUSION This study found differences in patterns of significant associations between risk factors of AISD for boys and girls. Therefore, different sex-based strategies are recommended to reduce the prevalence of AISD among adolescents in Paraguay.
Collapse
Affiliation(s)
- Taufique Ahmed
- Institute of Disaster Management and Vulnerability Studies, University of Dhaka, Dhaka, Bangladesh
| | - Muhammad Azimul Haque
- Institute of Disaster Management and Vulnerability Studies, University of Dhaka, Dhaka, Bangladesh
| | - Tanjin Kabir Aunto
- Institute of Disaster Management and Vulnerability Studies, University of Dhaka, Dhaka, Bangladesh
| | - Md Khalid Hasan
- Institute of Disaster Management and Vulnerability Studies, University of Dhaka, Dhaka, Bangladesh; Department of Human Development and Family Sciences, Texas Tech University, Lubbock, TX, United States.
| |
Collapse
|
2
|
DePoy LM, Vadnie CA, Petersen KA, Scott MR, Zong W, Yin R, Matthaei RC, Anaya FJ, Kampe CI, Tseng GC, McClung CA. Adolescent circadian rhythm disruption increases reward and risk-taking. Front Neurosci 2024; 18:1478508. [PMID: 39737435 PMCID: PMC11683121 DOI: 10.3389/fnins.2024.1478508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Circadian rhythm disturbances have long been associated with the development of psychiatric disorders, including mood and substance use disorders. Adolescence is a particularly vulnerable time for the onset of psychiatric disorders and for circadian rhythm and sleep disruptions. Preclinical studies have found that circadian rhythm disruption (CRD) impacts the brain and behavior, but this research is largely focused on adult disruptions. Here, we hypothesized that adolescent CRD would have a greater effect on psychiatric-related behaviors, relative to adult disruption. Methods We determined the long-term behavioral and neurobiological effects of CRD during early adolescence by exposing mice to 12 h shifts in the light/dark cycle. Adult mice were exposed to the same CRD paradigm. Behavior testing began approximately 4 weeks later for both groups. To identify possible mechanisms, we also measured gene expression in brain regions relevant to circadian rhythms, mood and reward. Results CRD during early adolescence, but not adulthood, persistently increased exploratory drive (risk-taking behavior) and cocaine preference when tested later in life. Interestingly, we found sex differences when intravenous cocaine self-administration was tested. While female mice with a history of adolescent CRD had a greater propensity to self-administer cocaine, as well as increased motivation and cue-induced reinstatement, male adolescent CRD mice had reduced motivation and extinction responding. Importantly, we found that transcripts in the SCN were affected by adolescent CRD and these were largely distinct across sex. Conclusion Overall, adolescent CRD in mice caused persistent increases in risky behavior, cocaine reward and cocaine self-administration, which suggests that CRD during adolescence may predispose individuals toward substance use disorders. Future research is required to elucidate how adolescent CRD affects behaviors relevant to mood-and substance use-related disorders across the 24-h day, as well as to identify intervention strategies to alleviate disruption during adolescence and novel therapeutic approaches once symptoms have begun.
Collapse
Affiliation(s)
- Lauren M. DePoy
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Chelsea A. Vadnie
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH, United States
| | - Kaitlyn A. Petersen
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Madeline R. Scott
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - RuoFei Yin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ross C. Matthaei
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Callie I. Kampe
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH, United States
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Colleen A. McClung
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH, United States
| |
Collapse
|
3
|
Ma C, Shen B, Chen L, Yang G. Impacts of circadian disruptions on behavioral rhythms in mice. FASEB J 2024; 38:e70183. [PMID: 39570004 DOI: 10.1096/fj.202401536r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Circadian rhythms are fundamental biological processes that recur approximately every 24 h, with the sleep-wake cycle or circadian behavior being a well-known example. In the field of chronobiology, mice serve as valuable model animals for studying mammalian circadian rhythms due to their genetic similarity to humans and the availability of various genetic tools for manipulation. Monitoring locomotor activity in mice provides valuable insights into the impact of various conditions or disturbances on circadian behavior. In this review, we summarized the effects of disturbance of biological rhythms on circadian behavior in mice. External factors, especially light exert a significant impact on circadian behavior. Additionally, feeding timing, food composition, ambient temperature, and physical exercise contribute to variations in the behavior of the mouse. Internal factors, including gender, age, genetic background, and clock gene mutation or deletion, are effective as well. Understanding the effects of circadian disturbances on murine behavior is essential for gaining insights into the underlying mechanisms of circadian regulation and developing potential therapeutic interventions for circadian-related disorders in humans.
Collapse
Affiliation(s)
- Changxiao Ma
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Bingyi Shen
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Lihong Chen
- Health Science Center, East China Normal University, Shanghai, China
| | - Guangrui Yang
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
4
|
Samanta S, Bagchi D, Gold MS, Badgaiyan RD, Barh D, Blum K. A Complex Relationship Among the Circadian Rhythm, Reward Circuit and Substance Use Disorder (SUD). Psychol Res Behav Manag 2024; 17:3485-3501. [PMID: 39411118 PMCID: PMC11479634 DOI: 10.2147/prbm.s473310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
The human brain not only controls the various physiological functions but is also the prime regulator of circadian rhythms, rewards, and behaviors. Environmental factors, professional stress, and social disintegration are regarded as the initial causative factors of addiction behavior. Shift work, artificial light exposure at night, and chronic and acute jet lag influence circadian rhythm dysfunction. The result is impaired neurotransmitter release, dysfunction of neural circuits, endocrine disturbance, and metabolic disorder, leading to advancement in substance use disorder. There is a bidirectional relationship between chronodisruption and addiction behavior. Circadian rhythm dysfunction, neuroadaptation in the reward circuits, and alteration in clock gene expression in the mesolimbic areas influence substance use disorder (SUD), and chronotherapy has potential benefits in the treatment strategies. This review explores the relationship among the circadian rhythm dysfunction, reward circuit, and SUD. The impact of chronotherapy on SUD has also been discussed.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore, West Bengal, 721101, India
| | - Debasis Bagchi
- Department of Biology, College of Arts and Sciences, Adelphi University, Garden City, NY, USA and Department of Psychology, Gordon F. Derner School of Psychology, Adelphi University, Garden City, NY, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Southern University, Houston, TX, 77004, USA
| | - Mark S Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Rajendra D Badgaiyan
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Debmalya Barh
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, BeloHorizonte, 31270-901, Brazil
- Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, 721172, West Bengal, India
| | - Kenneth Blum
- Division of Addiction Research & Education, Center for Sports, Exercise, and Mental Health, Western University Health Sciences, Pomona, CA, 91766, USA
- Institute of Psychology, Eotvos Loránd University, Budapest, 1053, Hungary
- Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Center, Dayton, OH, 45435, USA
- Department of Psychiatry, University of Vermont, Burlington, VT, 05405, USA
- Division of Nutrigenomics, The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX, 78701, USA
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| |
Collapse
|
5
|
Georgakopoulou VE, Sklapani P, Trakas N, Reiter RJ, Spandidos DA. Exploring the association between melatonin and nicotine dependence (Review). Int J Mol Med 2024; 54:82. [PMID: 39092582 PMCID: PMC11315657 DOI: 10.3892/ijmm.2024.5406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
Due to the addictive qualities of tobacco products and the compulsive craving and dependence associated with their use, nicotine dependence continues to be a serious public health concern on a global scale. Despite awareness of the associated health risks, nicotine addiction contributes to numerous acute and chronic medical conditions, including cardiovascular disease, respiratory disorders and cancer. The nocturnal secretion of pineal melatonin, known as the 'hormone of darkness', influences circadian rhythms and is implicated in addiction‑related behaviors. Melatonin receptors are found throughout the brain, influencing dopaminergic neurotransmission and potentially attenuating nicotine‑seeking behavior. Additionally, the antioxidant properties of melatonin may mitigate oxidative stress from chronic nicotine exposure, reducing cellular damage and lowering the risk of nicotine‑related health issues. In addition to its effects on circadian rhythmicity, melatonin acting via specific neural receptors influences sleep and mood, and provides neuroprotection. Disruptions in melatonin signaling may contribute to sleep disturbances and mood disorders, highlighting the potential therapeutic role of melatonin in addiction and psychiatric conditions. Melatonin may influence neurotransmitter systems involved in addiction, such as the dopaminergic, glutamatergic, serotonergic and endogenous opioid systems. Preclinical studies suggest the potential of melatonin in modulating reward processing, attenuating drug‑induced hyperactivity and reducing opioid withdrawal symptoms. Chronotherapeutic approaches targeting circadian rhythms and melatonin signaling show promise in smoking cessation interventions. Melatonin supplementation during periods of heightened nicotine cravings may alleviate withdrawal symptoms and reduce the reinforcing effects of nicotine. Further research is required however, to examine the molecular mechanisms underlying the melatonin‑nicotine association and the optimization of therapeutic interventions. Challenges include variability in individual responses to melatonin, optimal dosing regimens and identifying biomarkers of treatment response. Understanding these complexities could lead to personalized treatment strategies and improve smoking cessation outcomes.
Collapse
Affiliation(s)
| | - Pagona Sklapani
- Department of Biochemistry, Sismanogleio Hospital, 15126 Athens, Greece
| | - Nikolaos Trakas
- Department of Biochemistry, Sismanogleio Hospital, 15126 Athens, Greece
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health Science Center, San Antonio, TX 78229, USA
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
6
|
Sharma P, Nelson RJ. Disrupted Circadian Rhythms and Substance Use Disorders: A Narrative Review. Clocks Sleep 2024; 6:446-467. [PMID: 39189197 PMCID: PMC11348162 DOI: 10.3390/clockssleep6030030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024] Open
Abstract
Substance use disorder is a major global health concern, with a high prevalence among adolescents and young adults. The most common substances of abuse include alcohol, marijuana, cocaine, nicotine, and opiates. Evidence suggests that a mismatch between contemporary lifestyle and environmental demands leads to disrupted circadian rhythms that impair optimal physiological and behavioral function, which can increase the vulnerability to develop substance use disorder and related problems. The circadian system plays an important role in regulating the sleep-wake cycle and reward processing, both of which directly affect substance abuse. Distorted substance use can have a reciprocal effect on the circadian system by influencing circadian clock gene expression. Considering the detrimental health consequences and profound societal impact of substance use disorder, it is crucial to comprehend its complex association with circadian rhythms, which can pave the way for the generation of novel chronotherapeutic treatment approaches. In this narrative review, we have explored the potential contributions of disrupted circadian rhythms and sleep on use and relapse of different substances of abuse. The involvement of circadian clock genes with drug reward pathways is discussed, along with the potential research areas that can be explored to minimize disordered substance use by improving circadian hygiene.
Collapse
Affiliation(s)
- Pallavi Sharma
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA;
| | | |
Collapse
|
7
|
Tahara Y, Ding J, Ito A, Shibata S. Sweetened caffeine drinking revealed behavioral rhythm independent of the central circadian clock in male mice. NPJ Sci Food 2024; 8:51. [PMID: 39160163 PMCID: PMC11333706 DOI: 10.1038/s41538-024-00295-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Caffeine consumption is associated with the evening chronotype, and caffeine administration in mice results in prolonged period of the circadian rhythm in locomotor activity. However, as caffeine is bitter, sweetened caffeine is preferred by humans and mice; yet, its impact on the circadian clock has not been explored. In this study, mice were provided with freely available sweetened caffeine to investigate its effects on behavioral rhythms and peripheral clocks. Mice that freely consumed sweetened caffeine shifted from nocturnal to diurnal activity rhythms. In addition to the light-dark entrained behavioral rhythm component, some animals exhibited free-running period longer than 24-h. Intraperitoneal administration of caffeine at the beginning of the light phase also acutely induced diurnal behavior. The behavioral rhythms with long period (26-30 h) due to sweetened caffeine were observed even in mice housed under constant light or with a lesioned central circadian clock located in the suprachiasmatic nucleus of the hypothalamus; however, the rhythmicity was unstable. PER2::LUCIFERASE rhythms in peripheral tissues, such as the kidney, as measured via in vivo whole-body imaging during caffeine consumption, showed reduced amplitude and desynchronized phases among individuals. These results indicate that consumption of sweetened caffeine induces diurnal and long-period behavioral rhythms irrespective of the central clock, causing desynchronization of the clock in the body.
Collapse
Affiliation(s)
- Yu Tahara
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-0037, Japan.
- School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-0056, Japan.
| | - Jingwei Ding
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-0037, Japan
| | - Akito Ito
- School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-0056, Japan
| | - Shigenobu Shibata
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-0037, Japan
- School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-0056, Japan
| |
Collapse
|
8
|
Berbegal-Sáez P, Gallego-Landin I, Macía J, Alegre-Zurano L, Castro-Zavala A, Welz PS, Benitah SA, Valverde O. Lack of Bmal1 leads to changes in rhythmicity and impairs motivation towards natural stimuli. Open Biol 2024; 14:240051. [PMID: 39045857 PMCID: PMC11267724 DOI: 10.1098/rsob.240051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 07/25/2024] Open
Abstract
Maintaining proper circadian rhythms is essential for coordinating biological functions in mammals. This study investigates the effects of daily arrhythmicity using Bmal1-knockout (KO) mice as a model, aiming to understand behavioural and motivational implications. By employing a new mathematical analysis based on entropy divergence, we identified disrupted intricate activity patterns in mice derived by the complete absence of BMAL1 and quantified the difference regarding the activity oscillation's complexity. Changes in locomotor activity coincided with disturbances in circadian gene expression patterns. Additionally, we found a dysregulated gene expression profile particularly in brain nuclei like the ventral striatum, impacting genes related to reward and motivation. Further investigation revealed that arrhythmic mice exhibited heightened motivation for food and water rewards, indicating a link between circadian disruptions and the reward system. This research sheds light on how circadian clock alterations impact the gene expression regulating the reward system and how this, in turn, can lead to altered seeking behaviour and motivation for natural rewards. In summary, the present study contributes to our understanding of how reward processing is under the regulation of circadian clock machinery.
Collapse
Affiliation(s)
- Paula Berbegal-Sáez
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
| | - Ines Gallego-Landin
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
| | - Javier Macía
- Department of Medicine and Life Sciences (MELIS), Synthetic Biology for Biomedical Applications, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laia Alegre-Zurano
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
| | - Adriana Castro-Zavala
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
| | - Patrick-Simon Welz
- Program in Cancer Research, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Salvador A. Benitah
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelon08028, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Olga Valverde
- Department of Medicine and Life Sciences (MELIS), Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra, Barcelona, Spain
- Neuroscience Research Program, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|
9
|
Thoré ESJ, Aulsebrook AE, Brand JA, Almeida RA, Brodin T, Bertram MG. Time is of the essence: The importance of considering biological rhythms in an increasingly polluted world. PLoS Biol 2024; 22:e3002478. [PMID: 38289905 PMCID: PMC10826942 DOI: 10.1371/journal.pbio.3002478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Biological rhythms have a crucial role in shaping the biology and ecology of organisms. Light pollution is known to disrupt these rhythms, and evidence is emerging that chemical pollutants can cause similar disruption. Conversely, biological rhythms can influence the effects and toxicity of chemicals. Thus, by drawing insights from the extensive study of biological rhythms in biomedical and light pollution research, we can greatly improve our understanding of chemical pollution. This Essay advocates for the integration of biological rhythmicity into chemical pollution research to gain a more comprehensive understanding of how chemical pollutants affect wildlife and ecosystems. Despite historical barriers, recent experimental and technological advancements now facilitate the integration of biological rhythms into ecotoxicology, offering unprecedented, high-resolution data across spatiotemporal scales. Recognizing the importance of biological rhythms will be essential for understanding, predicting, and mitigating the complex ecological repercussions of chemical pollution.
Collapse
Affiliation(s)
- Eli S. J. Thoré
- Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden
- TRANSfarm—Science, Engineering, & Technology Group, KU Leuven, Lovenjoel, Belgium
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Anne E. Aulsebrook
- Department of Ornithology, Max Planck Institute for Biological Intelligence, Seewiesen, Germany
| | - Jack A. Brand
- Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden
- Institute of Zoology, Zoological Society of London, London, United Kingdom
| | - Rafaela A. Almeida
- Laboratory of Aquatic Ecology, Evolution, and Conservation, Department of Biology, KU Leuven, Leuven, Belgium
| | - Tomas Brodin
- Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Michael G. Bertram
- Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden
- Department of Zoology, Stockholm University, Stockholm, Sweden
- School of Biological Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
10
|
Chirokoff V, Dupuy M, Abdallah M, Fatseas M, Serre F, Auriacombe M, Misdrahi D, Berthoz S, Swendsen J, Sullivan EV, Chanraud S. Craving dynamics and related cerebral substrates predict timing of use in alcohol, tobacco, and cannabis use disorders. ADDICTION NEUROSCIENCE 2023; 9:100138. [PMID: 38389954 PMCID: PMC10883348 DOI: 10.1016/j.addicn.2023.100138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Background Patients treated for Substance Use Disorders exhibit highly fluctuating patterns of craving that could reveal novel prognostic markers of use. Accordingly, we 1) measured fluctuations within intensively repeated measures of craving and 2) linked fluctuations of craving to connectivity indices within resting-state (rs) brain regions to assess their relation to use among patients undergoing treatment for Alcohol, Tobacco and Cannabis Use Disorders. Method Participants -64 individuals with SUD for tobacco, alcohol, or cannabis and 35 healthy controls-completed a week of Ecological Momentary Assessment (EMA) during which they reported craving intensity and substance use five times daily. Before EMA, a subsample of 50 patients, and 34 healthy controls also completed resting-state (rs)-MRI acquisitions. Craving temporal dynamics within each day were characterized using Standard Deviation (SD), Auto-Correlation Factor (ACF), and Mean Successive Square Difference (MSSD). Absolute Difference (AD) in craving between assessments was a prospective prediction measure. Results Within-day, higher MSSD predicted greater substance use while controlling for mean craving. Prospectively higher AD predicted later increased substance use independently of previous use or craving level. Moreover, MSSD was linked to strength in five functional neural connections, most involving frontotemporal systems. Cerebello-thalamic and thalamo-frontal connectivity were also linked to substance use and distinguished the SUD from the controls. Conclusion To the best of our knowledge, this is the first study to indicate that instability in craving may be a trigger for use in several SUD types, beyond the known effect of craving intensity.
Collapse
Affiliation(s)
- Valentine Chirokoff
- University of Bordeaux, CNRS-UMR 5287 – Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), Bordeaux, France
- EPHE, PSL Research University, Paris, France
| | - Maud Dupuy
- University of Bordeaux, CNRS-UMR 5287 – Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), Bordeaux, France
| | - Majd Abdallah
- Bordeaux University, CNRS, Bordeaux Bioinformatics Center, IBGC UMR 5095, Bordeaux, France
- Groupe d’Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, Univeristy of Bordeaux, Bordeaux, France
| | - Melina Fatseas
- University of Bordeaux, CNRS-UMR 5287 – Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), Bordeaux, France
- CH Charles Perrens, Bordeaux, France
- CHU Bordeaux, Bordeaux, France
| | - Fuschia Serre
- University of Bordeaux, CNRS UMR 6033– Sleep, Addiction and Neuropsychiatry (SANPSY), Bordeaux, France
| | - Marc Auriacombe
- CH Charles Perrens, Bordeaux, France
- University of Bordeaux, CNRS UMR 6033– Sleep, Addiction and Neuropsychiatry (SANPSY), Bordeaux, France
| | - David Misdrahi
- University of Bordeaux, CNRS-UMR 5287 – Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), Bordeaux, France
- CH Charles Perrens, Bordeaux, France
| | - Sylvie Berthoz
- University of Bordeaux, CNRS-UMR 5287 – Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), Bordeaux, France
- Institut Mutualiste Montsouris, Department of Psychiatry for Adolescents and Young Adults, Paris, France
| | - Joel Swendsen
- University of Bordeaux, CNRS-UMR 5287 – Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), Bordeaux, France
- EPHE, PSL Research University, Paris, France
| | - Edith V. Sullivan
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Sandra Chanraud
- University of Bordeaux, CNRS-UMR 5287 – Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (INCIA), Bordeaux, France
- EPHE, PSL Research University, Paris, France
| |
Collapse
|
11
|
Cuenoud B, Huang Z, Hartweg M, Widmaier M, Lim S, Wenz D, Xin L. Effect of circadian rhythm on NAD and other metabolites in human brain. Front Physiol 2023; 14:1285776. [PMID: 38028810 PMCID: PMC10665902 DOI: 10.3389/fphys.2023.1285776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Nicotinamide Adenine Dinucleotide (NAD) plays a central role in the master circadian clock of the brain (the suprachiasmatic nuclei, SCN) as demonstrated in many model organisms. NAD acts as an enzyme co-factor and substrate and its modulation was found to be tightly regulated to the periodicity of the cycles. However, in human brain, the effect of the circadian rhythm (CR) on the metabolism of the SCN and other brain regions is poorly understood. We conducted a magnetic resonance spectroscopy (MRS) study at a high magnetic field, measuring the occipital brain NAD levels and other metabolites in two different morning and afternoon diurnal states in 25 healthy participants. Salivary cortisol levels were determined to confirm that the experiment was done in two chronologically different physiological conditions, and a behavioral test of risk-taking propensity was administered. Overall, we found that the CR did not significantly affect NAD levels in the occipital brain region. The other brain metabolites measured, including lactate, were not significantly affected by the CR either, except for taurine. The CR did impact risk-taking behavior and salivary cortisol level, confirming that the participants were in two circadian different behavioral and physiological states in the morning and in the afternoon. Measurement of the CR effect on NAD and taurine levels in other brain regions might provide stronger effects.
Collapse
Affiliation(s)
- Bernard Cuenoud
- Research and Clinical Development, Nestlé Health Science, Epalinges, Switzerland
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Zhiwei Huang
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mickael Hartweg
- Clinical Research Unit, Nestlé Research and Development, Lausanne, Switzerland
| | - Mark Widmaier
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - SongI. Lim
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Daniel Wenz
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lijing Xin
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
12
|
Ramasamy T, Doke M, McLaughlin JP, Samikkannu T. Circadian disruption and psychostimulants dysregulates plasma acute-phase proteins and circulating cell-free mitochondrial DNA. Brain Behav Immun Health 2023; 31:100659. [PMID: 37455861 PMCID: PMC10344797 DOI: 10.1016/j.bbih.2023.100659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
Background Previous studies have indicated a close link between the inflammatory response, exacerbated by circadian disruption and psychostimulants such as cocaine and methamphetamine (METH). Indicators of this inflammation include cortisol and acute-phase proteins (APPs) like C-reactive protein (CRP), complement C3 (C3), and serum amyloid A (SAA). The connection between these inflammation markers and circulating mitochondrial DNA (mtDNA) has been gaining attention. However, the specific influence of cocaine and METH on APP, cortisol, and mtDNA levels in mice with disturbed circadian rhythm has yet to be explored, which is the main aim of this research. Methods In our study, we employed 10-12-week-old male C57BL/6J mice, which underwent an imposed 6-h phase advance every six days for a total of eight cycles. This process led to the formation of mice with disrupted circadian rhythm and sleep disorders (CRSD). We administered 11 dosages of cocaine and METH 15 mg/kg and 20 mg/kg, respectively to these CRSD mice over the course of 22 days. Quantitative assessments of CRP, C3, SAA, cortisol, and cell-free circulating mtDNA were conducted using enzyme-linked immunosorbent assay (ELISA), Western Blot, and quantitative real-time polymerase chain reaction (qRT-PCR) techniques. Results The experiment revealed that disruption in circadian rhythm alone or cocaine or METH on their own increased CRP, C3, SAA, and cortisol levels in comparison with the control group. CRSD mice, exposed to cocaine and METH, showed a significant rise in CRP, C3, and SAA, while those without exposure remained stable. We also found a reduction in circulating cell-free mtDNA in all CRSD mice, regardless of cocaine and METH exposure. Conclusions The findings of our study affirm that the levels of CRP, C3, SAA, and cortisol, which reflect inflammation, are enhanced by circadian disruption, cocaine, and METH, and these levels show a strong correlation with the content of circulating cell-free mtDNA. Furthermore, it also shows the potential link between the disruption of the circadian clock and the inflammatory response triggered by cocaine and METH.
Collapse
Affiliation(s)
- Tamizhselvi Ramasamy
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, 77843, USA
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, Tamilnadu, India
| | - Mayur Doke
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, 77843, USA
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Thangavel Samikkannu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
13
|
Foo JC, Skorodumov I, Spanagel R, Meinhardt MW. Sex- and age-specific effects on the development of addiction and compulsive-like drinking in rats. Biol Sex Differ 2023; 14:44. [PMID: 37420305 PMCID: PMC10327342 DOI: 10.1186/s13293-023-00529-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/27/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND Biological factors are known to influence disease trajectories and treatment effectiveness in alcohol addiction and preclinical and clinical evidence suggests that sex is an important factor influencing disease dynamics in alcohol dependence. Another critical factor is age at first intoxicating drink, which has been identified as a risk factor for later alcohol binging. Preclinical research allows prospective monitoring of rodents throughout the lifespan, providing very detailed information that cannot be acquired in humans. Lifetime monitoring in rodents can be conducted under highly controlled conditions, during which one can systematically introduce multiple biological and environmental factors that impact behaviors of interest. METHODS Here, we used the alcohol deprivation effect (ADE) rat model of alcohol addiction in a computerized drinkometer system, acquiring high-resolution data to study changes over the course of addictive behavior as well as compulsive-like drinking in cohorts of adolescent vs. adult as well as male vs. female rats. RESULTS Female rats drank more alcohol than male rats during the whole experiment, drinking much more weak alcohol (5%) and similar amounts of stronger alcohol solutions (10%, 20%); female rats also consumed more alcohol than male rats during quinine taste adulteration. Increased consumption in females compared to males was driven by larger access sizes of alcohol. Differences in circadian patterns of movement were observed between groups. Early age of onset of drinking (postnatal day 40) in male rats had surprisingly little impact on the development of drinking behavior and compulsivity (quinine taste adulteration) when compared to rats that started drinking during early adulthood (postnatal day 72). CONCLUSIONS Our results suggest that there are sex-specific drinking patterns, not only in terms of total amount consumed, but specifically in terms of solution preference and access size. These findings provide a better understanding of sex and age factors involved in the development of drinking behavior, and can inform the preclinical development of models of addiction, drug development and exploration of options for new treatments.
Collapse
Affiliation(s)
- Jerome C. Foo
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Ivan Skorodumov
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Rainer Spanagel
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Marcus W. Meinhardt
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
- Department of Molecular Neuroimaging, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
14
|
Ketchesin KD, Becker-Krail DD, Xue X, Wilson RS, Lam TT, Williams KR, Nairn AC, Tseng GC, Logan RW. Differential Effects of Cocaine and Morphine on the Diurnal Regulation of the Mouse Nucleus Accumbens Proteome. J Proteome Res 2023. [PMID: 37311105 PMCID: PMC10392613 DOI: 10.1021/acs.jproteome.3c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Substance use disorders are associated with disruptions in sleep and circadian rhythms that persist during abstinence and may contribute to relapse risk. Repeated use of substances such as psychostimulants and opioids may lead to significant alterations in molecular rhythms in the nucleus accumbens (NAc), a brain region central to reward and motivation. Previous studies have identified rhythm alterations in the transcriptome of the NAc and other brain regions following the administration of psychostimulants or opioids. However, little is known about the impact of substance use on the diurnal rhythms of the proteome in the NAc. We used liquid chromatography coupled to tandem mass spectrometry-based quantitative proteomics, along with a data-independent acquisition analysis pipeline, to investigate the effects of cocaine or morphine administration on diurnal rhythms of proteome in the mouse NAc. Overall, our data reveal cocaine and morphine differentially alter diurnal rhythms of the proteome in the NAc, with largely independent differentially expressed proteins dependent on time-of-day. Pathways enriched from cocaine altered protein rhythms were primarily associated with glucocorticoid signaling and metabolism, whereas morphine was associated with neuroinflammation. Collectively, these findings are the first to characterize the diurnal regulation of the NAc proteome and demonstrate a novel relationship between the phase-dependent regulation of protein expression and the differential effects of cocaine and morphine on the NAc proteome. The proteomics data in this study are available via ProteomeXchange with identifier PXD042043.
Collapse
Affiliation(s)
- Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Darius D Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Rashaun S Wilson
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- W.M. Keck Biotechnology Mass Spectrometry (MS) & Proteomics Resource Laboratory, Yale University School of Medicine, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - TuKiet T Lam
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- W.M. Keck Biotechnology Mass Spectrometry (MS) & Proteomics Resource Laboratory, Yale University School of Medicine, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Kenneth R Williams
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Angus C Nairn
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- Department of Psychiatry, Yale University School of Medicine, Connecticut Mental Health Center, New Haven, Connecticut 06511, United States
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01655, United States
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
15
|
Ketchesin KD, Becker-Krail DD, Xue X, Wilson RS, Lam TT, Williams KR, Nairn AC, Tseng GC, Logan RW. Differential Effects of Cocaine and Morphine on the Diurnal Regulation of the Mouse Nucleus Accumbens Proteome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530696. [PMID: 36909659 PMCID: PMC10002738 DOI: 10.1101/2023.03.01.530696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Substance use disorders (SUDs) are associated with disruptions in sleep and circadian rhythms that persist during abstinence and may contribute to relapse risk. Repeated use of substances such as psychostimulants and opioids may lead to significant alterations in molecular rhythms in the nucleus accumbens (NAc), a brain region central to reward and motivation. Previous studies have identified rhythm alterations in the transcriptome of the NAc and other brain regions following the administration of psychostimulants or opioids. However, little is known about the impact of substance use on the diurnal rhythms of the proteome in the NAc. We used liquid chromatography coupled to tandem mass spectrometry-based (LC-MS/MS) quantitative proteomics, along with a data-independent acquisition (DIA) analysis pipeline, to investigate the effects of cocaine or morphine administration on diurnal rhythms of proteome in the mouse NAc. Overall, our data reveals cocaine and morphine differentially alters diurnal rhythms of the proteome in the NAc, with largely independent differentially expressed proteins dependent on time-of-day. Pathways enriched from cocaine altered protein rhythms were primarily associated with glucocorticoid signaling and metabolism, whereas morphine was associated with neuroinflammation. Collectively, these findings are the first to characterize the diurnal regulation of the NAc proteome and demonstrate a novel relationship between phase-dependent regulation of protein expression and the differential effects of cocaine and morphine on the NAc proteome.
Collapse
|
16
|
de Zavalia N, Ferraro S, Amir S. Sexually dimorphic role of circadian clock genes in alcohol drinking behavior. Psychopharmacology (Berl) 2023; 240:431-440. [PMID: 36184679 DOI: 10.1007/s00213-022-06247-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/17/2022] [Indexed: 11/25/2022]
Abstract
Sex differences in alcohol use and abuse are pervasive and carry important implications for the prevention and treatment of alcohol use disorder (AUD), yet insight into underlying sexually dimorphic mechanisms is limited. Growing experimental and clinical evidence points to an important influence of circadian rhythms and circadian clock genes in the control of alcohol drinking behavior and AUD. Sex differences in the expression of circadian rhythms and in the molecular circadian clock that drive these rhythms have been reported in humans and animals. While studying the role of striatal circadian clock gene expression in the control of affective and goal-directed behaviors, we uncovered a novel sexually dimorphic function of the clock genes Bmal1 and Per2 in the control of voluntary alcohol consumption in mice, which may contribute to sex differences in alcohol drinking behavior. In this mini review, we briefly discuss relevant literature on AUD, circadian rhythms and clock genes, and on sex differences in these domains, and describe our own findings on clock genes as sexually dimorphic regulators of alcohol drinking behavior in mice.
Collapse
Affiliation(s)
- Nuria de Zavalia
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Sarah Ferraro
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Shimon Amir
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada.
| |
Collapse
|
17
|
Association of chronotype, social jetlag, sleep duration and depressive symptoms in Chinese college students. J Affect Disord 2023; 320:735-741. [PMID: 36270445 DOI: 10.1016/j.jad.2022.10.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 09/30/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES To describe the prevalence of depressive symptoms among Chinese college students and examine the relationship between sleep and circadian rhythm disruption (SCRD) indicators (chronotype, social jetlag, sleep duration) and depressive symptoms. METHODS From April to May 2019, the College Student Behavior and Health Cohort Study was conducted among Chinese college students from 2 universities in Anhui and Jiangxi provinces. The current study used data from the third follow-up study. Questionnaire content includes socio-demographic and lifestyle information. Social jetlag and sleep duration were calculated by answering the question about sleep time. Chronotype was assessed by the Morning and Evening Questionnaire (MEQ-5). Depressive symptoms were evaluated by the Patient Health Questionnaire 9 (PHQ-9). A Chi-square test was used to examine the proportion depressive symptoms. Multinomial logistic regression model were used to explore the associations of circadian rhythm indicators with depressive symptoms. RESULTS The prevalence of mild depression, moderate and above depression was 18.8 % and 6.9 %. Evening types (E-types) and short sleep duration were significantly associated with depression. Stratified analysis indicated that E-types and social jetlag≥2 h were associated with mild depression (OR = 5.67, 95 % CI: 1.83-17.51), as well as stratified analysis indicated that E-types and sleep duration<8 h were associated with mild depression (OR = 5.10, 95 % CI: 1.88-13.87). CONCLUSIONS The findings suggest that depressive symptoms are more severe when multiple SCRD indicators are out of whack. The intervention programs of depressive symptoms should consider the context of the multidimensional aspects of Chinese college student sleep.
Collapse
|
18
|
Gamble MC, Chuan B, Gallego-Martin T, Shelton MA, Puig S, O'Donnell CP, Logan RW. A role for the circadian transcription factor NPAS2 in the progressive loss of non-rapid eye movement sleep and increased arousal during fentanyl withdrawal in male mice. Psychopharmacology (Berl) 2022; 239:3185-3200. [PMID: 35915264 PMCID: PMC10925970 DOI: 10.1007/s00213-022-06200-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/22/2022] [Indexed: 10/16/2022]
Abstract
RATIONALE Synthetic opioids like fentanyl are contributing to the rise in rates of opioid use disorder and drug overdose deaths. Sleep dysfunction and circadian rhythm disruption may worsen during opioid withdrawal and persist during abstinence. Severe and persistent sleep and circadian alterations are putative factors in opioid craving and relapse. However, very little is known about the impact of fentanyl on sleep architecture and sleep-wake cycles, particularly opioid withdrawal. Further, circadian rhythms regulate sleep-wake cycles, and the circadian transcription factor, neuronal PAS domain 2 (NPAS2) is involved in the modulation of sleep architecture and drug reward. Here, we investigate the role of NPAS2 in fentanyl-induced sleep alterations. OBJECTIVES To determine the effect of fentanyl administration and withdrawal on sleep architecture, and the role of NPAS2 as a factor in fentanyl-induced sleep changes. METHODS Electroencephalography (EEG) and electromyography (EMG) was used to measure non-rapid eye movement sleep (NREMS) and rapid eye movement sleep (REMS) at baseline and following acute and chronic fentanyl administration in wild-type and NPAS2-deficient male mice. RESULTS Acute and chronic administration of fentanyl led to increased wake and arousal in both wild-type and NPAS2-deficient mice, an effect that was more pronounced in NPAS2-deficient mice. Chronic fentanyl administration led to decreased NREMS, which persisted during withdrawal, progressively decreasing from day 1 to 4 of withdrawal. The impact of fentanyl on NREMS and arousal was more pronounced in NPAS2-deficient mice. CONCLUSIONS Chronic fentanyl disrupts NREMS, leading to a progressive loss of NREMS during subsequent days of withdrawal. Loss of NPAS2 exacerbates the impact of fentanyl on sleep and wake, revealing a potential role for the circadian transcription factor in opioid-induced sleep changes.
Collapse
Affiliation(s)
- Mackenzie C Gamble
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
- Molecular and Translational Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Byron Chuan
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Teresa Gallego-Martin
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Micah A Shelton
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Stephanie Puig
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Christopher P O'Donnell
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, 02118, USA.
| |
Collapse
|
19
|
Sleep Disorder and Cocaine Abuse Impact Purine and Pyrimidine Nucleotide Metabolic Signatures. Metabolites 2022; 12:metabo12090869. [PMID: 36144274 PMCID: PMC9502494 DOI: 10.3390/metabo12090869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
Disturbances in the circadian rhythm alter the normal sleep-wake cycle, which increases vulnerability to drug abuse. Drug abuse can disrupt several homeostatic processes regulated by the circadian rhythm and influence addiction paradigms, including cravings for cocaine. The relationship between circadian rhythm and cocaine abuse is complex and bidirectional, and disruption impacts both brain function and metabolic profiles. Therefore, elucidating the impact of circadian rhythm changes and cocaine abuse on the human metabolome may provide new insights into identifying potential biomarkers. We examine the effect of cocaine administration with and without circadian rhythm sleep disruption (CRSD) on metabolite levels and compare these to healthy controls in an in vivo study. A metabolomics analysis is performed on the control, CRSD, cocaine, and CRSD with cocaine groups. Plasma metabolite concentrations are analyzed using a liquid chromatography electrochemical array platform. We identify 242 known metabolites compared to the control; 26 in the CRSD with cocaine group, 4 in the CRSD group, and 22 in the cocaine group are significantly differentially expressed. Intriguingly, in the CRSD with cocaine treatment group, the expression levels of uridine monophosphate (p < 0.008), adenosine 5′-diphosphate (p < 0.044), and inosine (p < 0.019) are significantly altered compared with those in the cocaine group. In summary, alterations in purine and pyrimidine metabolism provide clues regarding changes in the energy profile and metabolic pathways associated with chronic exposure to cocaine and CRSD.
Collapse
|
20
|
Foo JC, Meinhardt MW, Skorodumov I, Spanagel R. Alcohol solution strength preference predicts compulsive-like drinking behavior in rats. Alcohol Clin Exp Res 2022; 46:1710-1719. [PMID: 35871774 DOI: 10.1111/acer.14910] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/09/2022] [Accepted: 07/11/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Understanding compulsive drinking behavior is key to improving outcomes in the treatment of addiction. In the present study, we investigated compulsive-like drinking in alcohol-addicted rats using the alcohol deprivation effect (ADE) model of relapse behavior, which involves repeated deprivation and reintroduction phases; the latter approximate relapse. METHODS High-resolution longitudinal drinking and locomotor data were measured while rats (n = 30) underwent a four-bottle (water, 5%, 10%, 20% alcohol v/v) free-choice ADE paradigm. Alcohol bottles were adulterated with the bitter compound quinine during a reintroduction phase to test for compulsive behavior. We characterized how drinking and locomotor behavior during ADE + quinine differed from a regular ADE and how, at the individual level, behavioral parameters extracted from the regular ADE related to compulsive-like drinking. Associations of drinking with locomotor activity were also examined. RESULTS In the ADE with quinine, we observed reduced consumption of alcohol and a shift to preference for stronger alcohol. Quinine acted by decreasing both the access size and frequency of drinking of 5% alcohol while increasing the frequency of consumption of 20% alcohol. Preference for higher alcohol concentrations prior to the quinine challenge was associated with greater compulsive-like drinking behavior; higher baseline consumption of 20% alcohol correlated with more drinking of quinine-adulterated solutions while high frequency and amount of 5% alcohol consumption at baseline were correlated with being more strongly affected by quinine. Associations between locomotor activity and drinking behavior were observed at the hourly level. These associations reflected changing preferences across experimental phases. CONCLUSION Drinking patterns, and specifically solution preference, may offer insights into the presentation of compulsive-like drinking. The findings provide a preclinical basis for observations from epidemiological studies that link higher risk and burden of alcohol-related disease to stronger alcohol concentrations and encourage further translational studies to better understand the underlying mechanisms.
Collapse
Affiliation(s)
- Jerome C Foo
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marcus W Meinhardt
- Institute for Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ivan Skorodumov
- Institute for Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rainer Spanagel
- Institute for Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
21
|
Zou H, Zhou H, Yan R, Yao Z, Lu Q. Chronotype, circadian rhythm, and psychiatric disorders: Recent evidence and potential mechanisms. Front Neurosci 2022; 16:811771. [PMID: 36033630 PMCID: PMC9399511 DOI: 10.3389/fnins.2022.811771] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 06/21/2022] [Indexed: 12/27/2022] Open
Abstract
The circadian rhythm is crucial for physiological and behavioral functions. Chronotype, which represents individual preferences for activity and performance, is associated with human health issues, particularly psychiatric disorders. This narrative review, which focuses on the relationship between chronotype and mental disorders, provides an insight into the potential mechanism. Recent evidence indicates that (1) the evening chronotype is a risk factor for depressive disorders and substance use disorders, whereas the morning chronotype is a protective factor. (2) Evening chronotype individuals with bipolar disorder tend to have more severe symptoms and comorbidities. (3) The evening chronotype is only related to anxiety symptoms. (4) The relationship between chronotype and schizophrenia remains unclear, despite increasing evidence on their link. (5) The evening chronotype is significantly associated with eating disorders, with the majority of studies have focused on binge eating disorders. Furthermore, the underlying mechanisms or influence factors are described in detail, including clock genes, brain characteristics, neuroendocrinology, the light/dark cycle, social factors, psychological factors, and sleep disorders. These findings provide the latest evidence on chronotypes and psychiatric disorders and serve as a valuable reference for researchers.
Collapse
Affiliation(s)
- Haowen Zou
- Nanjing Brain Hospital, Medical School, Nanjing University, Nanjing, China
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hongliang Zhou
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Yan
- Nanjing Brain Hospital, Medical School, Nanjing University, Nanjing, China
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Zhijian Yao
- Nanjing Brain Hospital, Medical School, Nanjing University, Nanjing, China
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - Qing Lu
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
- Child Development and Learning Science, Key Laboratory of Ministry of Education, Nanjing, China
| |
Collapse
|
22
|
Becker-Krail DD, Ketchesin KD, Burns JN, Zong W, Hildebrand MA, DePoy LM, Vadnie CA, Tseng GC, Logan RW, Huang YH, McClung CA. Astrocyte Molecular Clock Function in the Nucleus Accumbens Is Important for Reward-Related Behavior. Biol Psychiatry 2022; 92:68-80. [PMID: 35461698 PMCID: PMC9232937 DOI: 10.1016/j.biopsych.2022.02.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Substance use disorders are associated with disruptions in circadian rhythms. Both human and animal work have shown the integral role for circadian clocks in the modulation of reward behaviors. Astrocytes have emerged as key regulators of circadian rhythmicity. However, no studies to date have identified the role of circadian astrocyte function in the nucleus accumbens (NAc), a hub for reward regulation, or determined the importance of these rhythms for reward-related behavior. METHODS Using astrocyte-specific RNA sequencing across time of day, we first characterized diurnal variation of the NAc astrocyte transcriptome. We then investigated the functional significance of this circadian regulation through viral-mediated disruption of molecular clock function in NAc astrocytes, followed by assessment of reward-related behaviors, metabolic-related molecular assays, and whole-cell electrophysiology in the NAc. RESULTS Strikingly, approximately 43% of the astrocyte transcriptome has a diurnal rhythm, and key metabolic pathways were enriched among the top rhythmic genes. Moreover, mice with a viral-mediated loss of molecular clock function in NAc astrocytes show a significant increase in locomotor response to novelty, exploratory drive, operant food self-administration, and motivation. At the molecular level, these animals also show disrupted metabolic gene expression, along with significant downregulation of both lactate and glutathione levels in the NAc. Loss of NAc astrocyte clock function also significantly altered glutamatergic signaling onto neighboring medium spiny neurons, alongside upregulated glutamate-related gene expression. CONCLUSIONS Taken together, these findings demonstrate a novel role for astrocyte circadian molecular clock function in the regulation of the NAc and reward-related behaviors.
Collapse
Affiliation(s)
- Darius D Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer N Burns
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wei Zong
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mariah A Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lauren M DePoy
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chelsea A Vadnie
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Yanhua H Huang
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
23
|
Becker-Krail DD, Walker WH, Nelson RJ. The Ventral Tegmental Area and Nucleus Accumbens as Circadian Oscillators: Implications for Drug Abuse and Substance Use Disorders. Front Physiol 2022; 13:886704. [PMID: 35574492 PMCID: PMC9094703 DOI: 10.3389/fphys.2022.886704] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
Circadian rhythms convergently evolved to allow for optimal synchronization of individuals’ physiological and behavioral processes with the Earth’s 24-h periodic cycling of environmental light and temperature. Whereas the suprachiasmatic nucleus (SCN) is considered the primary pacemaker of the mammalian circadian system, many extra-SCN oscillatory brain regions have been identified to not only exhibit sustainable rhythms in circadian molecular clock function, but also rhythms in overall region activity/function and mediated behaviors. In this review, we present the most recent evidence for the ventral tegmental area (VTA) and nucleus accumbens (NAc) to serve as extra-SCN oscillators and highlight studies that illustrate the functional significance of the VTA’s and NAc’s inherent circadian properties as they relate to reward-processing, drug abuse, and vulnerability to develop substance use disorders (SUDs).
Collapse
Affiliation(s)
- Darius D Becker-Krail
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
24
|
López-Gil JF, Cavero-Redondo I, Tárraga López PJ, Jiménez-López E, González AD, Sequí-Domínguez I, Mesas AE. Anxiety-Induced Sleep Disturbance and Associated Lifestyle Behaviors According to Sex in Argentine Adolescents. Front Behav Neurosci 2022; 16:860241. [PMID: 35548694 PMCID: PMC9084278 DOI: 10.3389/fnbeh.2022.860241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/15/2022] [Indexed: 12/01/2022] Open
Abstract
Purpose The aim of the current study was twofold: first, to determine the prevalence of anxiety-induced sleep disturbances among Argentine adolescents according to sex, and second, to identify the association between these sleep disturbances and lifestyle behaviors in this population. Methods This is a cross-sectional study with data from the Global School-based Student Health Survey (GSHS) in Argentina (2018). A total of 32,393 adolescents (aged 12–17 years; 53.4% girls) were included in the final analysis. Anxiety-induced sleep disturbances were assessed with the question “During the past 12 months, how often have you been so worried about something that you could not sleep at night?” Results The prevalence of anxiety-induced sleep disturbances was higher in girls (17.4%) than in boys (7.9%) (p < 0.001). In boys, results indicated that those who used marijuana (cannabis) (odds ratio [OR] = 1.46, 95% confidence interval [CI] 1.08–1.98), used amphetamine or methamphetamine (OR = 2.19, 95% CI 1.28–3.77), walked or biked to or from school (OR = 1.53, 95% CI 1.19–1.96), and spent 3 h or more in sedentary behaviors (OR = 1.35, 95% CI 1.05–1.74) were more likely to report anxiety-induced sleep disturbances. In girls, those who ate from a fast-food restaurant (OR = 1.24, 95% CI 1.05–1.47), consumed alcoholic beverages (OR = 1.45, 95% CI 1.19–1.75), smoked cigarettes (OR = 2.09, 95%CI 1.05–4.14), consumed any tobacco product (OR = 1.47, 95% CI 1.19–1.82), used amphetamine or methamphetamine (OR = 2.08, 95% CI 1.33–3.26), and those who spent 3 h or more in sedentary behaviors (OR = 1.32, 95% CI 1.11–1.57) were more likely to report frequent anxiety-induced sleep disturbances. Conclusion In conclusion, considerable sex differences were observed with respect to the prevalence of anxiety-related sleep disturbances and associated lifestyle aspects.
Collapse
Affiliation(s)
| | - Iván Cavero-Redondo
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Pedro J. Tárraga López
- Department of Medical Sciences, Faculty of Medicine, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Estela Jiménez-López
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Department of Psychiatry, Hospital Virgen de La Luz, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Cuenca, Spain
- *Correspondence: Estela Jiménez-López,
| | - Alberto Durán González
- Postgraduate Program in Public Health, Universidade Estadual de Londrina, Londrina, Brazil
| | | | - Arthur Eumann Mesas
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Postgraduate Program in Public Health, Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
25
|
Cervantes M, Lewis RG, Della-Fazia MA, Borrelli E, Sassone-Corsi P. Dopamine D2 receptor signaling in the brain modulates circadian liver metabolomic profiles. Proc Natl Acad Sci U S A 2022; 119:e2117113119. [PMID: 35271395 PMCID: PMC8931347 DOI: 10.1073/pnas.2117113119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
SignificanceWe analyzed the liver metabolome of mice deficient in the expression of the dopamine D2 receptor (D2R) in striatal medium spiny neurons (iMSN-D2RKO) and found profound changes in the liver circadian metabolome compared to control mice. Additionally, we show activation of dopaminergic circuits by acute cocaine administration in iMSN-D2RKO mice reprograms the circadian liver metabolome in response to cocaine. D2R signaling in MSNs is key for striatal output and essential for regulating the first response to the cellular and rewarding effects of cocaine. Our results suggest changes in dopamine signaling in specific striatal neurons evoke major changes in liver physiology. Dysregulation of liver metabolism could contribute to an altered allostatic state and therefore be involved in continued use of drugs.
Collapse
Affiliation(s)
- Marlene Cervantes
- INSERM U1233, Center for Epigenetics and Metabolism, University of California, Irvine, CA 92697
- Department of Biological Chemistry, University of California, Irvine, CA 92697
| | - Robert G. Lewis
- INSERM U1233, Center for Epigenetics and Metabolism, University of California, Irvine, CA 92697
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697
| | | | - Emiliana Borrelli
- INSERM U1233, Center for Epigenetics and Metabolism, University of California, Irvine, CA 92697
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697
| | - Paolo Sassone-Corsi
- INSERM U1233, Center for Epigenetics and Metabolism, University of California, Irvine, CA 92697
- Department of Biological Chemistry, University of California, Irvine, CA 92697
| |
Collapse
|
26
|
Balcı Ö, Çalışkan M. Investigation of the relationship between chronotype, learning style and academic achievement of university students during distance education in the pandemic period. Chronobiol Int 2022; 39:858-871. [PMID: 35176946 DOI: 10.1080/07420528.2022.2041658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The purpose of this study was to investigate the relationship between chronotype, learning style, and academic achievement. The study sample consisted of 1884 volunteer undergraduate students from 58 different universities across Turkey. The data were collected online during distance education because of the Covid-19 pandemic. Turkish version of Morningness-Eveningness Questionnaire (MEQ) and Big16 Learning Modality Inventory were used for data collection. The correlation between chronotype and cumulative grade point average (CGPA) scores was analyzed using Spearman's rho, and differences among the chronotypes were analyzed with Kruskal-Wallis test. Chi-square tests with pairwise z-test were used to analyze the relationships between the participants' learning style preferences and chronotypes. Also, ANCOVA analyses were performed to evaluate the interactions of gender × chronotype and gender × learning style on CGPA scores. The results revealed that participants' preference for visual and auditory styles differed by chronotype. Visual learning style was more dominant among morning (M) types, while auditory style was more dominant among evening (E) types. However, the most preferred learning style was visual, followed by auditory and kinesthetic styles for all chronotypes. No relationship was found between chronotype and academic achievement. The CGPA scores of the participants with kinesthetic learning style were lower than the participants with auditory and visual learning styles. The female participants had higher CGPA score than males. However, the CGPA scores did not differ for both male and female participants with different chronotype and learning style preferences. From the results, we suggest that academic achievement can be enhanced by consideration of students' learning style preferences for all chronotypes.
Collapse
Affiliation(s)
- Özgül Balcı
- School of Foreign Languages, Necmettin Erbakan University, Konya, Turkey
| | - Muhittin Çalışkan
- Ahmet Keleşoğlu Faculty of Education, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
27
|
Becker-Krail DD, Parekh PK, Ketchesin KD, Yamaguchi S, Yoshino J, Hildebrand MA, Dunham B, Ganapathiraiu MK, Logan RW, McClung CA. Circadian transcription factor NPAS2 and the NAD + -dependent deacetylase SIRT1 interact in the mouse nucleus accumbens and regulate reward. Eur J Neurosci 2022; 55:675-693. [PMID: 35001440 PMCID: PMC9355311 DOI: 10.1111/ejn.15596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 12/14/2021] [Accepted: 01/06/2022] [Indexed: 02/03/2023]
Abstract
Substance use disorders are associated with disruptions to both circadian rhythms and cellular metabolic state. At the molecular level, the circadian molecular clock and cellular metabolic state may be interconnected through interactions with the nicotinamide adenine dinucleotide (NAD+ )-dependent deacetylase, sirtuin 1 (SIRT1). In the nucleus accumbens (NAc), a region important for reward, both SIRT1 and the circadian transcription factor neuronal PAS domain protein 2 (NPAS2) are highly enriched, and both are regulated by the metabolic cofactor NAD+ . Substances of abuse, like cocaine, greatly disrupt cellular metabolism and promote oxidative stress; however, their effects on NAD+ in the brain remain unclear. Interestingly, cocaine also induces NAc expression of both NPAS2 and SIRT1, and both have independently been shown to regulate cocaine reward in mice. However, whether NPAS2 and SIRT1 interact in the NAc and/or whether together they regulate reward is unknown. Here, we demonstrate diurnal expression of Npas2, Sirt1 and NAD+ in the NAc, which is altered by cocaine-induced upregulation. Additionally, co-immunoprecipitation reveals NPAS2 and SIRT1 interact in the NAc, and cross-analysis of NPAS2 and SIRT1 chromatin immunoprecipitation sequencing reveals several reward-relevant and metabolic-related pathways enriched among shared gene targets. Notably, NAc-specific Npas2 knock-down or a functional Npas2 mutation in mice attenuates SIRT1-mediated increases in cocaine preference. Together, our data reveal an interaction between NPAS2 and SIRT1 in the NAc, which may serve to integrate cocaine's effects on circadian and metabolic factors, leading to regulation of drug reward.
Collapse
Affiliation(s)
- Darius D. Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Puja K. Parekh
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Shintaro Yamaguchi
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Yoshino
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mariah A. Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Brandon Dunham
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madhavi K. Ganapathiraiu
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA,Correspondence: Colleen A. McClung,
| |
Collapse
|
28
|
Mech AM, Merteroglu M, Sealy IM, Teh MT, White RJ, Havelange W, Brennan CH, Busch-Nentwich EM. Behavioral and Gene Regulatory Responses to Developmental Drug Exposures in Zebrafish. Front Psychiatry 2022; 12:795175. [PMID: 35082702 PMCID: PMC8785235 DOI: 10.3389/fpsyt.2021.795175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/29/2021] [Indexed: 01/22/2023] Open
Abstract
Developmental consequences of prenatal drug exposure have been reported in many human cohorts and animal studies. The long-lasting impact on the offspring-including motor and cognitive impairments, cranial and cardiac anomalies and increased prevalence of ADHD-is a socioeconomic burden worldwide. Identifying the molecular changes leading to developmental consequences could help ameliorate the deficits and limit the impact. In this study, we have used zebrafish, a well-established behavioral and genetic model with conserved drug response and reward pathways, to identify changes in behavior and cellular pathways in response to developmental exposure to amphetamine, nicotine or oxycodone. In the presence of the drug, exposed animals showed altered behavior, consistent with effects seen in mammalian systems, including impaired locomotion and altered habituation to acoustic startle. Differences in responses seen following acute and chronic exposure suggest adaptation to the presence of the drug. Transcriptomic analysis of exposed larvae revealed differential expression of numerous genes and alterations in many pathways, including those related to cell death, immunity and circadian rhythm regulation. Differential expression of circadian rhythm genes did not correlate with behavioral changes in the larvae, however, two of the circadian genes, arntl2 and per2, were also differentially expressed at later stages of development, suggesting a long-lasting impact of developmental exposures on circadian gene expression. The immediate-early genes, egr1, egr4, fosab, and junbb, which are associated with synaptic plasticity, were downregulated by all three drugs and in situ hybridization showed that the expression for all four genes was reduced across all neuroanatomical regions, including brain regions implicated in reward processing, addiction and other psychiatric conditions. We anticipate that these early changes in gene expression in response to drug exposure are likely to contribute to the consequences of prenatal exposure and their discovery might pave the way to therapeutic intervention to ameliorate the long-lasting deficits.
Collapse
Affiliation(s)
- Aleksandra M. Mech
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, United Kingdom
| | - Munise Merteroglu
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Ian M. Sealy
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Muy-Teck Teh
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, England, United Kingdom
| | - Richard J. White
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - William Havelange
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, United Kingdom
| | - Caroline H. Brennan
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, United Kingdom
| | - Elisabeth M. Busch-Nentwich
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, United Kingdom
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
29
|
Meinhardt MW, Giannone F, Hirth N, Bartsch D, Spampinato SM, Kelsch W, Spanagel R, Sommer WH, Hansson AC. Disrupted circadian expression of beta-arrestin 2 affects reward-related µ-opioid receptor function in alcohol dependence. J Neurochem 2021; 160:454-468. [PMID: 34919270 DOI: 10.1111/jnc.15559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
There is increasing evidence for a daily rhythm of μ-opioid receptor (MOR) efficacy and the development of alcohol dependence. Previous studies show that beta-Arrestin 2 (bArr2) has an impact on alcohol intake, at least partially mediated via modulation of MOR signaling, which in turn mediates the alcohol rewarding effects. Considering the interplay of circadian rhythms on MOR and alcohol dependence, we aimed to investigate bArr2 in alcohol dependence at different time-points of the day/light cycle on the level of bArr2 mRNA (in situ hybridization), MOR availability (receptor autoradiography) and MOR signaling (Damgo-stimulated G-protein coupling) in the nucleus accumbens of alcohol-dependent and non-dependent Wistar rats. Using a microarray data set we found that bArr2, but not bArr1, shows a diurnal transcription pattern in the accumbens of naïve rats with higher expression levels during the active cycle. In three-week abstinent rats, bArr2 is upregulated in the accumbens at the beginning of the active cycle (ZT15), whereas no differences were found at the beginning of the inactive cycle (ZT3), compared to controls. This effect was accompanied with a specific downregulation of MOR binding in the active cycle. Additionally, we detect a higher receptor coupling during the inactive cycle compared to the active cycle in alcohol-dependent animals. Together, we report a daily rhythmicity for bArr2 expression linked to an inverse pattern of MOR, suggesting an involvement for bArr2 on circadian regulation of G-protein coupled receptors in alcohol dependence. The presented data may have implications for the development of novel bArr2-related treatment targets for alcoholism.
Collapse
Affiliation(s)
- Marcus W Meinhardt
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany.,Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Francesco Giannone
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Nathalie Hirth
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Dusan Bartsch
- Department of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Santi M Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Wolfgang Kelsch
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Wolfgang H Sommer
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Anita C Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| |
Collapse
|
30
|
Association between nicotine dependency with occupational injury in Korean men. Ann Occup Environ Med 2021; 33:e14. [PMID: 34754475 PMCID: PMC8203833 DOI: 10.35371/aoem.2021.33.e14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/09/2021] [Indexed: 01/23/2023] Open
Abstract
Background The relationship between smoking status or second-hand smoking and occupational injuries has been the subject of considerable study, but few have studied the relationship between nicotine dependence and occupational injuries. The objective of this study was to investigate the relationship between nicotine dependence and occupational injury among employees at a range of Korean companies. Methods Initially, the personal and occupational characteristics and nicotine dependences of workers were measured, and 12 months later a survey was used to determine whether subjects had experienced any occupational injury. This study was conducted in several workplaces on 6,893 male workers in manufacturing and service industries that received health screening at Inha University Hospital in Incheon. Results The adjusted odds ratios (ORs) of occupational injury in the low, moderate, and high nicotine dependence groups were 1.38 (95% confidence interval [CI]: 1.04-1.84), 1.52 (95% CI: 1.10-2.10), and 1.71 (95% CI: 0.92-3.19), respectively. For smokers only, adjusted ORs tended to increase linearly (p for trend < 0.05). When only smokers were included, analysis of continuous FTND (Fagerstrom Test of Nicotine Dependence) scores showed that adjusted OR increased by 1.10 (95% CI: 1.03-1.19) per FTND point. After stratifying the data by working type and working hours per week, the non-shift work group maintained this relationship (OR: 1.13, 95% CI: 1.04-1.24) and OR was higher in the group that works more than 60 hours per week with FTND score as a continuous variable (OR: 1.24, 95% CI: 1.07-1.44). Conclusions The study shows nicotine dependency might affect occupational injury. From a short-term perspective, addressing worker's nicotine dependence by giving an adequate break time or smoking area might reduce work-related injuries.
Collapse
|
31
|
Bailey LS, Bagley JR, Dodd R, Olson A, Bolduc M, Philip VM, Reinholdt LG, Sukoff Rizzo SJ, Tarantino L, Gagnon L, Chesler EJ, Jentsch JD. Heritable variation in locomotion, reward sensitivity and impulsive behaviors in a genetically diverse inbred mouse panel. GENES, BRAIN, AND BEHAVIOR 2021; 20:e12773. [PMID: 34672075 PMCID: PMC9044817 DOI: 10.1111/gbb.12773] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022]
Abstract
Drugs of abuse, including alcohol and stimulants like cocaine, produce effects that are subject to individual variability, and genetic variation accounts for at least a portion of those differences. Notably, research in both animal models and human subjects point toward reward sensitivity and impulsivity as being trait characteristics that predict relatively greater positive subjective responses to stimulant drugs. Here we describe use of the eight collaborative cross (CC) founder strains and 38 (reversal learning) or 10 (all other tests) CC strains to examine the heritability of reward sensitivity and impulsivity traits, as well as genetic correlations between these measures and existing addiction-related phenotypes. Strains were all tested for activity in an open field and reward sensitivity (intake of chocolate BOOST®). Mice were then divided into two counterbalanced groups and underwent reversal learning (impulsive action and waiting impulsivity) or delay discounting (impulsive choice). CC and founder mice show significant heritability for impulsive action, impulsive choice, waiting impulsivity, locomotor activity, and reward sensitivity, with each impulsive phenotype determined to be non-correlating, independent traits. This research was conducted within the broader, inter-laboratory effort of the Center for Systems Neurogenetics of Addiction (CSNA) to characterize CC and DO mice for multiple, cocaine abuse related traits. These data will facilitate the discovery of genetic correlations between predictive traits, which will then guide discovery of genes and genetic variants that contribute to addictive behaviors.
Collapse
Affiliation(s)
- Lauren S Bailey
- State University of New York - Binghamton University, Binghamton, New York, USA
| | - Jared R Bagley
- State University of New York - Binghamton University, Binghamton, New York, USA
| | - Rainy Dodd
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | | | | | | | | | - Stacey J Sukoff Rizzo
- The Jackson Laboratory, Bar Harbor, Maine, USA
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lisa Tarantino
- The Jackson Laboratory, Bar Harbor, Maine, USA
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | - James David Jentsch
- State University of New York - Binghamton University, Binghamton, New York, USA
- The Jackson Laboratory, Bar Harbor, Maine, USA
| |
Collapse
|
32
|
Peng LU, Bai G, Pang Y. Roles of NPAS2 in circadian rhythm and disease. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1257-1265. [PMID: 34415290 DOI: 10.1093/abbs/gmab105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Indexed: 11/14/2022] Open
Abstract
NPAS2, a circadian rhythm gene encoding the neuronal PAS domain protein 2 (NPAS2), has received widespread attention because of its complex functions in cells and diverse roles in disease progression, especially tumorigenesis. NPAS2 binds with DNA at E-box sequences and forms heterodimers with another circadian protein, brain and muscle ARNT-like protein 1 (BMAL1). Nucleotide variations of the NPAS2 gene have been shown to influence the overall survival and risk of death of cancer patients, and differential expression of NPAS2 has been linked to patient outcomes in breast cancer, lung cancer, non-Hodgkin's lymphoma, and other diseases. Here, we review the latest advances in our understanding of NPAS2 with the aim of drawing attention to its potential clinical applications and prospects.
Collapse
Affiliation(s)
- L u Peng
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Gaigai Bai
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yingxin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
33
|
Stebbins HE, Jacobs ME, Hatton KT, Kaila EN, Rhoades MM. Spontaneous eye blink rate mediates the relationship between sleepiness and impulsivity to negative stimuli. Biol Psychol 2021; 165:108191. [PMID: 34530069 DOI: 10.1016/j.biopsycho.2021.108191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
Previous studies have demonstrated that sleep deprivation results in a negativity bias, especially in the context of impaired response inhibition. In the present study we investigated spontaneous eye blink rate (EBR), a correlate of dopamine function, as a mediator of the relationship between subjective sleepiness and impulsivity toward negative stimuli on a Go/NoGo task. Participants rated their sleepiness on a number of measures including the Epworth Sleepiness Scale (ESS), the Karolinska Sleepiness Scale (KSS) and subscales of the Chronic Sleep Reduction Questionnaire (CSRQ). The findings revealed that EBR mediated the relationship between sleepiness as measured by the Karolinska Sleepiness Scale (KSS) and commission errors on negatively valanced stimuli. These findings suggest that reduced inhibition in responding to negative stimuli can be found as a function of subjective sleepiness and that changes in dopamine function may be one contributing factor explaining this relationship.
Collapse
Affiliation(s)
- Hilary E Stebbins
- Department of Psychological Science, University of Mary Washington, USA.
| | - Megan E Jacobs
- Department of Psychological Science, University of Mary Washington, USA
| | | | - Erin N Kaila
- Department of Psychological Science, University of Mary Washington, USA
| | - Mollie M Rhoades
- Department of Psychological Science, University of Mary Washington, USA
| |
Collapse
|
34
|
Saad L, Zwiller J, Kalsbeek A, Anglard P. Epigenetic Regulation of Circadian Clocks and Its Involvement in Drug Addiction. Genes (Basel) 2021; 12:1263. [PMID: 34440437 PMCID: PMC8394526 DOI: 10.3390/genes12081263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 12/19/2022] Open
Abstract
Based on studies describing an increased prevalence of addictive behaviours in several rare sleep disorders and shift workers, a relationship between circadian rhythms and addiction has been hinted for more than a decade. Although circadian rhythm alterations and molecular mechanisms associated with neuropsychiatric conditions are an area of active investigation, success is limited so far, and further investigations are required. Thus, even though compelling evidence connects the circadian clock to addictive behaviour and vice-versa, yet the functional mechanism behind this interaction remains largely unknown. At the molecular level, multiple mechanisms have been proposed to link the circadian timing system to addiction. The molecular mechanism of the circadian clock consists of a transcriptional/translational feedback system, with several regulatory loops, that are also intricately regulated at the epigenetic level. Interestingly, the epigenetic landscape shows profound changes in the addictive brain, with significant alterations in histone modification, DNA methylation, and small regulatory RNAs. The combination of these two observations raises the possibility that epigenetic regulation is a common plot linking the circadian clocks with addiction, though very little evidence has been reported to date. This review provides an elaborate overview of the circadian system and its involvement in addiction, and we hypothesise a possible connection at the epigenetic level that could further link them. Therefore, we think this review may further improve our understanding of the etiology or/and pathology of psychiatric disorders related to drug addiction.
Collapse
Affiliation(s)
- Lamis Saad
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands;
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Andries Kalsbeek
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands;
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Patrick Anglard
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- Institut National de la Santé et de la Recherche Médicale (INSERM), 75013 Paris, France
| |
Collapse
|
35
|
Zhang R, Manza P, Tomasi D, Kim SW, Shokri-Kojori E, Demiral SB, Kroll DS, Feldman DE, McPherson KL, Biesecker CL, Wang GJ, Volkow ND. Dopamine D1 and D2 receptors are distinctly associated with rest-activity rhythms and drug reward. J Clin Invest 2021; 131:e149722. [PMID: 34264865 DOI: 10.1172/jci149722] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/14/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Certain components of rest-activity rhythms such as greater eveningness (delayed phase), physical inactivity (blunted amplitude) and shift work (irregularity) are associated with increased risk for drug use. Dopaminergic (DA) signaling has been hypothesized to mediate the associations, though clinical evidence is lacking. METHODS We examined associations between rhythm components and striatal D1 (D1R) and D2/3 receptor (D2/3R) availability in 32 healthy adults (12 female, age: 42.40±12.22) and its relationship to drug reward. Rest-activity rhythms were assessed by one-week actigraphy combined with self-reports. [11C]NNC112 and [11C]raclopride Positron Emission Tomography (PET) scans were conducted to measure D1R and D2/3R availability, respectively. Additionally, self-reported drug-rewarding effects of 60 mg oral methylphenidate were assessed. RESULTS We found that delayed rhythm was associated with higher D1R availability in caudate, which was not attributable to sleep loss or 'social jet lag', whereas physical inactivity was associated with higher D2/3R availability in nucleus accumbens (NAc). Delayed rest-activity rhythm, higher caudate D1R and NAc D2/3R availability were associated with greater sensitivity to the rewarding effects of methylphenidate. CONCLUSION These findings reveal specific components of rest-activity rhythms associated with striatal D1R, D2/3R availability and drug-rewarding effects. Personalized interventions that target rest-activity rhythms may help prevent and treat substance use disorders. TRIAL REGISTRATION ClinicalTrials.gov: NCT03190954FUNDING. This work was accomplished with support from the National Institute on Alcohol Abuse and Alcoholism (ZIAAA000550).
Collapse
Affiliation(s)
- Rui Zhang
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Peter Manza
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Dardo Tomasi
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Sung Won Kim
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Ehsan Shokri-Kojori
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Sukru B Demiral
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Danielle S Kroll
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Dana E Feldman
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Katherine L McPherson
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Catherine L Biesecker
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, United States of America
| | - Nora D Volkow
- National Institute on Drug Abuse, NIH, Bethesda, United States of America
| |
Collapse
|
36
|
Buck JM, O'Neill HC, Stitzel JA. The Intergenerational Transmission of Developmental Nicotine Exposure-Induced Neurodevelopmental Disorder-Like Phenotypes is Modulated by the Chrna5 D397N Polymorphism in Adolescent Mice. Behav Genet 2021; 51:665-684. [PMID: 34159514 DOI: 10.1007/s10519-021-10071-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/07/2021] [Indexed: 12/01/2022]
Abstract
Maternal tobacco smoking during pregnancy constitutes developmental nicotine exposure (DNE) and is associated with nicotine dependence and neurodevelopmental disorders in both children and grandchildren as well as animal models thereof. Genetic variants such as the CHRNA5 single nucleotide polymorphism (SNP) rs16969968, which leads to an aspartic acid to asparagine substitution at amino acid position 398 (D398N) in the alpha-5 nicotinic acetylcholine receptor subunit, can also confer risk for nicotine dependence and neurodevelopmental disorders in the absence of DNE. However, the degrees to which, the consequences of maternal smoking on offspring outcomes are influenced by genetic variants and interactions therewith are not well understood. Addressing this void in the literature, the present study utilizes a DNE mouse model engineered to possess the equivalent of the human D398N SNP in CHRNA5 (D397N SNP in mice) to assess how the N397 risk allele impacts the induction and intergenerational transmission of a range of neurodevelopmental disorder-related behavioral phenotypes in first- and second-generation DNE offspring. Results reveal that offspring possessing the N397 variant in the absence of DNE as well as DNE offspring and grand offspring possessing theD397 variant exhibit analogous neurodevelopmental disorder-like phenotypes including hyperactivity, risk-taking behaviors, aberrant rhythmicity of activity, and enhanced nicotine consumption. DNE amplified these behavioral anomalies in first-generation N397 progeny, but the severity of DNE-evoked behavioral perturbations did not significantly differ between first-generation D397 and N397 DNE mice for any measure. Remarkably, the behavioral profiles of second-generation N397 DNE progeny closely resembled DNE-naive D397 mice, suggesting that the N397 variant may protect against the intergenerational transmission of DNE-induced neurodevelopmental disorder-like behaviors.
Collapse
Affiliation(s)
- Jordan M Buck
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, USA
| | - Heidi C O'Neill
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA.
- Department of Integrative Physiology, University of Colorado, Boulder, USA.
| |
Collapse
|
37
|
Han Q, Bagi Z, Rudic RD. Review: Circadian clocks and rhythms in the vascular tree. Curr Opin Pharmacol 2021; 59:52-60. [PMID: 34111736 DOI: 10.1016/j.coph.2021.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/17/2022]
Abstract
The progression of vascular disease is influenced by many factors including aging, gender, diet, hypertension, and poor sleep. The intrinsic vascular circadian clock and the timing it imparts on the vasculature both conditions and is conditioned by all these variables. Circadian rhythms and their molecular components are rhythmically cycling in each endothelial cell, smooth muscle cell, in each artery, arteriole, vein, venule, and capillary. New research continues to tackle how circadian clocks act in the vasculature, describing influences in experimental and human disease, identifying potential target genes, compensatory molecules, that ultimately reveal a complexity that is vascular-bed-specific, cell-type-specific, and even single-cell-specific. Though we are yet to achieve a complete understanding, here we survey recent observations that are shedding more light on the nature of the interaction between circadian rhythms and the vascular system with implications for blood vessel disease.
Collapse
Affiliation(s)
- Qimei Han
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Raducu Daniel Rudic
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
38
|
Partington HS, Nutter JM, Eells JB. Nurr1 deficiency shortens free running period, enhances photoentrainment to phase advance, and disrupts circadian cycling of the dopamine neuron phenotype. Behav Brain Res 2021; 411:113347. [PMID: 33991560 DOI: 10.1016/j.bbr.2021.113347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023]
Abstract
Neurological and neuropsychiatric disorders, including addiction, schizophrenia, and Parkinson's disease (PD), involve dysfunction in midbrain dopamine (DA) neurotransmission with severity of disease symptoms and progression associated with disrupted circadian rhythms. The nuclear transcription factor Nurr1, essential for DA neuron (DAN) development, survival, and maintenance, is also known to interact with circadian rhythm regulating clock proteins. In the Nurr1-null heterozygous (+/-) mice, a Nurr1 deficient model which reproduces some of the alterations in DA function found in schizophrenia and PD, we measured, using wheel-running activity, the free running period (tau) and photoperiod entrainment. Because Nurr1 has a role in regulating the DA phenotype, we also measured the circadian fluctuations in the number of DANs using tyrosine hydroxylase (TH) immunofluorescence. In Nurr1 +/- mice, tau was significantly shorter and entrainment to a 6 h earlier shift in the dark cycle was accelerated. The Nurr1 wild-type (+/+) mice cycled DAN numbers across time, with a significantly greater number (∼2-fold increase) of DANs at zeitgeber time (ZT) 0 than ZT12. The +/- mice, however, did not cycle the DA phenotype, as no differences in DAN numbers were observed between ZT0 and ZT12. Additionally, the +/- mice had significantly fewer DANs at ZT0 but not at ZT12 as compared to +/+ mice. Based these data, circadian rhythms and fluctuations in the DA phenotype requires normal Nurr1 function. A better understanding is needed of the mechanisms regulating the DA phenotype and subsequent neurotransmission across the circadian cycle and how this is altered in circadian rhythm and DA neurotransmission-associated disorders.
Collapse
Affiliation(s)
- Heath S Partington
- East Carolina University, Department of Anatomy and Cell Biology, Brody School of Medicine, Greenville, NC, USA
| | - Jennifer Makenzie Nutter
- East Carolina University, Department of Anatomy and Cell Biology, Brody School of Medicine, Greenville, NC, USA
| | - Jeffrey B Eells
- East Carolina University, Department of Anatomy and Cell Biology, Brody School of Medicine, Greenville, NC, USA.
| |
Collapse
|
39
|
Roy K, Maji D, Deb I. Increase of Cry 1 expression is a common phenomenon of the disturbed circadian clock in ischemic stroke and opioid addiction. Biochem Biophys Res Commun 2021; 558:8-13. [PMID: 33894675 DOI: 10.1016/j.bbrc.2021.04.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/14/2021] [Indexed: 10/21/2022]
Abstract
Increasing evidences suggest the involvement of disrupted circadian clock in various pathologies including stroke and substance abuse. Here we took an attempt to do a comparative study on the regulation of circadian clock gene expression under two pathological circumstances - Opioid addiction and Ischemic stroke in the same cell line model (human neuroblastoma SH-SY5Y cells). To mimic in vivo ischemic stroke condition cells were placed in a hypoxia chamber and incubated for 10 h in balanced salt solution lacking glucose, aerated with an anaerobic gas mixture (95% N2 and 5% C02). For opioid addiction cells were treated with morphine sulphate at 10 μM dose for 48 h. We found that although circadian clock gets disturbed in both states, pattern of alteration of clock gene expressions were different and change was more severe in ischemic stroke than addiction. Interestingly, increase in expression of Cry1 showed as a common factor to both the diseases. This paper also emphasizes the interconnection between the severities of neuronal injury induced by ischemic stroke or opioid abuse to circadian system. Finally, this study will further enrich our knowledge towards the pattern of circadian rhythm disturbances under different pathological states.
Collapse
Affiliation(s)
- Kaninika Roy
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Daytee Maji
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Ishani Deb
- Department of Biochemistry, University of Calcutta, Kolkata, India.
| |
Collapse
|
40
|
Roy K, Maji D, Deb I. Oxygen glucose deprivation impairs circadian clock genes expressions in Neuro 2A neuroblastoma cells unlike C6 glioma. BIOL RHYTHM RES 2021. [DOI: 10.1080/09291016.2021.1911551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Kaninika Roy
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Daytee Maji
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Ishani Deb
- Department of Biochemistry, University of Calcutta, Kolkata, India
| |
Collapse
|
41
|
Bakhti-Suroosh A, Towers EB, Lynch WJ. A buprenorphine-validated rat model of opioid use disorder optimized to study sex differences in vulnerability to relapse. Psychopharmacology (Berl) 2021; 238:1029-1046. [PMID: 33404740 PMCID: PMC7786148 DOI: 10.1007/s00213-020-05750-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022]
Abstract
RATIONALE Opioid use disorder (OUD) is a major epidemic in the USA. Despite evidence indicating that OUD may be particularly severe for women, preclinical models have yet to establish sex as a major factor in OUD. OBJECTIVES Here, we examined sex differences in vulnerability to relapse following intermittent access fentanyl self-administration and protracted abstinence and used buprenorphine, the FDA-approved treatment for OUD, to test the validity of our model. METHODS Following acquisition of fentanyl self-administration under one of two training conditions, male and female rats were given extended, 24-h/day access to fentanyl (0.25 μg/kg/infusion, 10 days) using an intermittent access procedure. Vulnerability to relapse was assessed using an extinction/cue-induced reinstatement procedure following 14 days of abstinence; buprenorphine (0 or 3 mg/kg/day) was administered throughout abstinence. RESULTS Levels of drug-seeking were high following extended-access fentanyl self-administration and abstinence; buprenorphine markedly decreased drug-seeking supporting the validity of our relapse model. Females self-administered more fentanyl and responded at higher levels during subsequent extinction testing. Buprenorphine was effective in both sexes and eliminated sex and estrous phase differences in drug-seeking. Interestingly, the inclusion of a time-out during training had a major impact on later fentanyl self-administration in females, but not males, indicating that the initial exposure conditions can persistently impact vulnerability in females. CONCLUSIONS These findings demonstrate the utility of this rat model for determining sex and hormonal influences on the development and treatment of OUD.
Collapse
Affiliation(s)
- Anousheh Bakhti-Suroosh
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, P.O. Box 801402, Charlottesville, VA, 22904, USA
| | - Eleanor Blair Towers
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, P.O. Box 801402, Charlottesville, VA, 22904, USA
| | - Wendy J Lynch
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, P.O. Box 801402, Charlottesville, VA, 22904, USA.
| |
Collapse
|
42
|
Sharma R, Puckett H, Kemerling M, Parikh M, Sahota P, Thakkar M. Antisense-Induced Downregulation of Clock Genes in the Shell Region of the Nucleus Accumbens Reduces Binge Drinking in Mice. Alcohol Clin Exp Res 2021; 45:530-542. [PMID: 33606281 PMCID: PMC8535763 DOI: 10.1111/acer.14549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/22/2022]
Abstract
INTRODUCTIONS Binge drinking is a deadly pattern of alcohol consumption. Evidence suggests that genetic variation in clock genes is strongly associated with alcohol misuse; however, the neuroanatomical basis for such a relationship is unknown. The shell region of the nucleus accumbens (NAcSh) is well known to play a role in binge drinking. Hence, we examined whether clock genes in the NAcSh regulate binge drinking. METHODS To address this question, 2 experiments were performed on male C57BL/6J mice. In the first experiment, mice exposed to alcohol or sucrose under the 4-day drinking-in-the-dark (DID) paradigm were euthanized at 2 different time points on day 4 [7 hours after light (pre-binge drinking) or dark (post-binge drinking) onset]. The brains were processed for RT-PCR to examine the expression of circadian clock genes (Clock, Per1, and Per2) in the NAcSh and suprachiasmatic nucleus (SCN). In the second experiment, mice were exposed to alcohol, sucrose, or water as described above. On day 4, 1 hour prior to the onset of alcohol exposure, mice were bilaterally infused with either a mixture of circadian clock gene antisense oligodeoxynucleotides (AS-ODNs; antisense group) or nonsense/random ODNs (R-ODNs; control group) through surgically implanted cannulas above the NAcSh. Alcohol/sucrose/water consumption was measured for 4 hours. Blood alcohol concentration was measured to confirm binge drinking. Microinfusion sites were histologically verified using cresyl violet staining. RESULTS As compared to sucrose, mice euthanized post-binge drinking (not pre-binge drinking) on day 4 displayed a greater expression of circadian genes in the NAcSh but not in the SCN. Knockdown of clock genes in the NAcSh caused a significantly lower volume of alcohol to be consumed on day 4 than in the control treatment. No differences were found in sucrose or water consumption. CONCLUSIONS Our results suggest that clock genes in the NAcSh play a crucial role in binge drinking.
Collapse
Affiliation(s)
- Rishi Sharma
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Hunter Puckett
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Micaela Kemerling
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Meet Parikh
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Pradeep Sahota
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Mahesh Thakkar
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
43
|
Diurnal changes in perineuronal nets and parvalbumin neurons in the rat medial prefrontal cortex. Brain Struct Funct 2021; 226:1135-1153. [PMID: 33585984 DOI: 10.1007/s00429-021-02229-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/21/2021] [Indexed: 01/08/2023]
Abstract
Perineuronal nets (PNNs) surrounding fast-spiking, parvalbumin (PV) interneurons provide excitatory:inhibitory balance, which is impaired in several disorders associated with altered diurnal rhythms, yet few studies have examined diurnal rhythms of PNNs or PV cells. We measured the intensity and number of PV cells and PNNs labeled with Wisteria floribunda agglutinin (WFA) and also the oxidative stress marker 8-oxo-deoxyguanosine (8-oxo-dG) in rat prelimbic medial prefrontal cortex (mPFC) at Zeitgeber times (ZT) ZT0 (lights-on, inactive phase), ZT6 (mid-inactive phase), ZT12 (lights-off, active phase), and ZT18 (mid-active phase). Relative to ZT0, the intensities of PNN and PV labeling were increased in the dark (active) phase compared with the light (inactive) phase. The intensity of 8-oxo-dG was decreased from ZT0 at all times (ZT6,12,18). We also measured GAD 65/67 and vGLUT1 puncta apposed to PV cells with and without PNNs. There were more excitatory puncta on PV cells with PNNs at ZT18 vs. ZT6, but no changes in PV cells without PNNs and no changes in inhibitory puncta. Whole-cell slice recordings in fast-spiking (PV) cells with PNNs showed an increased ratio of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor:N-methyl-D-aspartate receptor (AMPA: NMDA) at ZT18 vs. ZT6. The number of PV cells and PV/PNN cells containing orthodenticle homeobox 2 (OTX2), which maintains PNNs, showed a strong trend toward an increase from ZT6 to ZT18. Diurnal fluctuations in PNNs and PV cells are expected to alter cortical excitatory:inhibitory balance and provide new insights into treatments for diseases impacted by disturbances in sleep and circadian rhythms.
Collapse
|
44
|
Circadian-Dependent and Sex-Dependent Increases in Intravenous Cocaine Self-Administration in Npas2 Mutant Mice. J Neurosci 2021; 41:1046-1058. [PMID: 33268545 DOI: 10.1523/jneurosci.1830-20.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 11/21/2022] Open
Abstract
Substance use disorder (SUD) is associated with disruptions in circadian rhythms. The circadian transcription factor neuronal PAS domain protein 2 (NPAS2) is enriched in reward-related brain regions and regulates reward, but its role in SU is unclear. To examine the role of NPAS2 in drug taking, we measured intravenous cocaine self-administration (acquisition, dose-response, progressive ratio, extinction, cue-induced reinstatement) in wild-type (WT) and Npas2 mutant mice at different times of day. In the light (inactive) phase, cocaine self-administration, reinforcement, motivation and extinction responding were increased in all Npas2 mutants. Sex differences emerged during the dark (active) phase with Npas2 mutation increasing self-administration, extinction responding, and reinstatement only in females as well as reinforcement and motivation in males and females. To determine whether circulating hormones are driving these sex differences, we ovariectomized WT and Npas2 mutant females and confirmed that unlike sham controls, ovariectomized mutant mice showed no increase in self-administration. To identify whether striatal brain regions are activated in Npas2 mutant females, we measured cocaine-induced ΔFosB expression. Relative to WT, ΔFosB expression was increased in D1+ neurons in the nucleus accumbens (NAc) core and dorsolateral (DLS) striatum in Npas2 mutant females after dark phase self-administration. We also identified potential target genes that may underlie the behavioral responses to cocaine in Npas2 mutant females. These results suggest NPAS2 regulates reward and activity in specific striatal regions in a sex and time of day (TOD)-specific manner. Striatal activation could be augmented by circulating sex hormones, leading to an increased effect of Npas2 mutation in females.SIGNIFICANCE STATEMENT Circadian disruptions are a common symptom of substance use disorders (SUDs) and chronic exposure to drugs of abuse alters circadian rhythms, which may contribute to subsequent SU. Diurnal rhythms are commonly found in behavioral responses to drugs of abuse with drug sensitivity and motivation peaking during the dark (active) phase in nocturnal rodents. Emerging evidence links disrupted circadian genes to SU vulnerability and drug-induced alterations to these genes may augment drug-seeking. The circadian transcription factor neuronal PAS domain protein 2 (NPAS2) is enriched in reward-related brain regions and regulates reward, but its role in SU is unclear. To examine the role of NPAS2 in drug taking, we measured intravenous cocaine self-administration in wild-type (WT) and Npas2 mutant mice at different times of day.
Collapse
|
45
|
Tamura EK, Oliveira-Silva KS, Ferreira-Moraes FA, Marinho EAV, Guerrero-Vargas NN. Circadian rhythms and substance use disorders: A bidirectional relationship. Pharmacol Biochem Behav 2021; 201:173105. [PMID: 33444601 DOI: 10.1016/j.pbb.2021.173105] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 01/23/2023]
Abstract
The circadian system organizes circadian rhythms (biological cycles that occur around 24 h) that couple environmental cues (zeitgebers) with internal functions of the organism. The misalignment between circadian rhythms and external cues is known as chronodisruption and contributes to the development of mental, metabolic and other disorders, including cancer, cardiovascular diseases and addictive disorders. Drug addiction represents a global public health concern and affects the health and well-being of individuals, families and communities. In this manuscript, we reviewed evidence indicating a bidirectional relationship between the circadian system and the development of addictive disorders. We provide information on the interaction between the circadian system and drug addiction for each drug or drug class (alcohol, cannabis, hallucinogens, psychostimulants and opioids). We also describe evidence showing that drug use follows a circadian pattern, which changes with the progression of addiction. Furthermore, clock gene expression is also altered during the development of drug addiction in many brain areas related to drug reward, drug seeking and relapse. The regulation of the glutamatergic and dopaminergic neurocircuitry by clock genes is postulated to be the main circadian mechanism underlying the escalation of drug addiction. The bidirectional interaction between the circadian system and drug addiction seems to be mediated by the effects caused by each drug or class of drugs of abuse. These studies provide new insights on the development of successful strategies aimed at restoring/stabilizing circadian rhythms to reduce the risk for addiction development and relapse.
Collapse
Affiliation(s)
- Eduardo K Tamura
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil.
| | - Kallyane S Oliveira-Silva
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Felipe A Ferreira-Moraes
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Eduardo A V Marinho
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Natalí N Guerrero-Vargas
- Department of Anatomy, Faculty of Medicine, Universidad Nacional Autonóma de México, Av Universidad 3000, Ciudad Universitaria, México City 04510, Mexico
| |
Collapse
|
46
|
Q P, KC W, CL E. Common genetic substrates of alcohol and substance use disorder severity revealed by pleiotropy detection against GWAS catalog in two populations. Addict Biol 2021; 26:e12877. [PMID: 32027075 PMCID: PMC7415504 DOI: 10.1111/adb.12877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/15/2019] [Accepted: 01/11/2020] [Indexed: 12/01/2022]
Abstract
Alcohol and other substance use disorders (AUD and SUD) are complex diseases that are postulated to have a polygenic inheritance and are often comorbid with other disorders. The comorbidities may arise partially through genetic pleiotropy. Identification of specific gene variants accounting for large parts of the variance in these disorders has yet to be accomplished. We describe a flexible strategy that takes a variant-trait association database and determines if a subset of disease/straits are potentially pleiotropic with the disorder under study. We demonstrate its usage in a study of use disorders in two independent cohorts: alcohol, stimulants, cannabis (CUD), and multi-substance use disorders (MSUD) in American Indians (AI) and AUD and CUD in Mexican Americans (MA). Using a machine learning method with variants in GWAS catalog, we identified 229 to 246 pleiotropic variants for AI and 153 to 160 for MA for each SUD. Inflammation was the most enriched for MSUD and AUD in AIs. Neurological disorder was the most significantly enriched for CUD in both cohorts, and for AUD and stimulants in AIs. Of the select pleiotropic genes shared among substances-cohorts, multiple biological pathways implicated in SUD and other psychiatric disorders were enriched, including neurotrophic factors, immune responses, extracellular matrix, and circadian regulation. Shared pleiotropic genes were significantly up-regulated in brain regions playing important roles in SUD, down-regulated in esophagus mucosa, and differentially regulated in adrenal gland. This study fills a gap for pleiotropy detection in understudied admixed populations and identifies pleiotropic variants that may be potential targets of interest for SUD.
Collapse
Affiliation(s)
- Peng Q
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - Wilhelmsen KC
- Department of Genetics and Neurology, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Ehlers CL
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037 USA
| |
Collapse
|
47
|
Hashemzadeh I, Marquez-Arrico JE, Hashemzadeh K, Navarro JF, Adan A. Circadian Functioning and Quality of Life in Substance Use Disorder Patients With and Without Comorbid Major Depressive Disorder. Front Psychiatry 2021; 12:750500. [PMID: 34777054 PMCID: PMC8586202 DOI: 10.3389/fpsyt.2021.750500] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 01/04/2023] Open
Abstract
Aim: Although a relationship between circadian disruption and development of several psychiatric disorders, such as major depressive disorder (MDD) and substance use disorder (SUD), has been observed, knowledge on this area is scarce yet. Therefore, this study aims to analyze the circadian functioning and quality of life (QOL) in SUD patients with and without comorbid MDD, two highly prevalent clinical entities with difficult therapeutic management. Methods: One hundred sixty-three male patients under treatment, 81 with SUD and 82 with SUD comorbid major depressive disorder (SUD + MDD), were evaluated. For the circadian functioning assessment, we calculated Social Jet Lag (SJL) and used the reduced Morningness-Eveningness Questionnaire (rMEQ) and the Pittsburgh Sleep Quality Index (PSQI). QOL was measured using the shortened version of the World Health Organization's Quality of Life Questionnaire (WHOQOL-BREF). We collected sociodemographic and clinical variables to evaluate their possible influence on the circadian functioning. Intergroup differences among the variables were examined by different analyses of covariance (ANCOVA and MANCOVA). The possible relationships of quantitative clinical variables with rMEQ, PSQI, and WHOQOL-BREF were explored using bivariate correlation analysis. Results: Lower SJL appears in the SUD + MDD group compared with SUD. The intermediate-type was more prevalent in the SUD group, while a higher percentage of morning-type patients was found in the SUD + MDD. Sleep quality (including latency and daytime dysfunction) was worse for SUD + MDD patients than for SUD even after controlling age and age of SUD onset variables. Last, QOL was poorer in patients with SUD + MDD and, for them, psychological health had a negative relationship with SJL and severity of depression. Conclusions: Our data support and extend previous findings indicating that SUD + MDD is associated with worse clinical characteristics, more sleep problems, and poorer QOL than SUD patients. These results underline the importance of a precise assessment of these measurements in future studies conducted in SUD patients with/without MDD comorbidity that could be considered from a therapeutic point of view.
Collapse
Affiliation(s)
- Iman Hashemzadeh
- Department of Clinical Psychology and Psychobiology, School of Psychology, University of Barcelona, Barcelona, Spain
| | - Julia E Marquez-Arrico
- Department of Clinical Psychology and Psychobiology, School of Psychology, University of Barcelona, Barcelona, Spain
| | - Kosar Hashemzadeh
- Department of Psychology, Fasa Branch, Islamic Azad University, Fasa, Iran
| | | | - Ana Adan
- Department of Clinical Psychology and Psychobiology, School of Psychology, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
48
|
Siemann JK, Grueter BA, McMahon DG. Rhythms, Reward, and Blues: Consequences of Circadian Photoperiod on Affective and Reward Circuit Function. Neuroscience 2020; 457:220-234. [PMID: 33385488 DOI: 10.1016/j.neuroscience.2020.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/01/2023]
Abstract
Circadian disruptions, along with altered affective and reward states, are commonly associated with psychiatric disorders. In addition to genetics, the enduring influence of environmental factors in programming neural networks is of increased interest in assessing the underpinnings of mental health. The duration of daylight or photoperiod is known to impact both the serotonin and dopamine systems, which are implicated in mood and reward-based disorders. This review first examines the effects of circadian disruption and photoperiod in the serotonin system in both human and preclinical studies. We next highlight how brain regions crucial for the serotoninergic system (i.e., dorsal raphe nucleus; DRN), and dopaminergic (i.e., nucleus accumbens; NAc and ventral tegmental area; VTA) system are intertwined in overlapping circuitry, and play influential roles in the pathology of mood and reward-based disorders. We then focus on human and animal studies that demonstrate the impact of circadian factors on the dopaminergic system. Lastly, we discuss how environmental factors such as circadian photoperiod can impact the neural circuits that are responsible for regulating affective and reward states, offering novel insights into the biological mechanisms underlying the pathophysiology, systems, and therapeutic treatments necessary for mood and reward-based disorders.
Collapse
Affiliation(s)
- Justin K Siemann
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA; Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Brad A Grueter
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA; Department of Anesthesiology, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37235, USA; Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA; Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
49
|
Brami-Cherrier K, Lewis RG, Cervantes M, Liu Y, Tognini P, Baldi P, Sassone-Corsi P, Borrelli E. Cocaine-mediated circadian reprogramming in the striatum through dopamine D2R and PPARγ activation. Nat Commun 2020; 11:4448. [PMID: 32895370 PMCID: PMC7477550 DOI: 10.1038/s41467-020-18200-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Substance abuse disorders are linked to alteration of circadian rhythms, although the molecular and neuronal pathways implicated have not been fully elucidated. Addictive drugs, such as cocaine, induce a rapid increase of dopamine levels in the brain. Here, we show that acute administration of cocaine triggers reprogramming in circadian gene expression in the striatum, an area involved in psychomotor and rewarding effects of drugs. This process involves the activation of peroxisome protein activator receptor gamma (PPARγ), a nuclear receptor involved in inflammatory responses. PPARγ reprogramming is altered in mice with cell-specific ablation of the dopamine D2 receptor (D2R) in the striatal medium spiny neurons (MSNs) (iMSN-D2RKO). Administration of a specific PPARγ agonist in iMSN-D2RKO mice elicits substantial rescue of cocaine-dependent control of circadian genes. These findings have potential implications for development of strategies to treat substance abuse disorders. Drugs of abuse have been shown to perturb circadian rhythms. Here, the authors show in mice that cocaine exposure modulates circadian gene expression in the striatum through a previously unappreciated pathway that involves dopamine D2 receptors and the nuclear receptor PPARγ.
Collapse
Affiliation(s)
- Karen Brami-Cherrier
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA
| | - Robert G Lewis
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA
| | - Marlene Cervantes
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA
| | - Yu Liu
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California Irvine, Irvine, CA, 92697, USA
| | - Paola Tognini
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California Irvine, Irvine, CA, 92697, USA
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA.
| | - Emiliana Borrelli
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
50
|
Vetrova MV, Rybakova KV, Goncharov OV, Kuchmenko DN, Genina IN, Semenova NV, Makarov IV, Zubova EY, Neznanov NG, Krupitsky EM. [Characteristics of sleep disturbances related to substance use disorders]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:153-159. [PMID: 32621482 DOI: 10.17116/jnevro2020120051153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Sleep disturbances are frequently observed in patients with substance use disorders during active use as well as in withdrawal period and in remission. However, there is limited information about the association between sleep disturbances and substance use disorders. This review summarizes results of the studies on specific characteristics of sleep disturbances related to alcohol, opioids and psychostimulants (cocaine) use. Data on objective and subjective measurements of sleep characteristics at different stages of the course of an addiction disorder (active use, withdrawal, remission) are presented.
Collapse
Affiliation(s)
- M V Vetrova
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia.,Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
| | - K V Rybakova
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia
| | - O V Goncharov
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia
| | - D N Kuchmenko
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia
| | - I N Genina
- State addiction hospital, St. Petersburg, Russia
| | - N V Semenova
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia
| | - I V Makarov
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia
| | - E Yu Zubova
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia
| | - N G Neznanov
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia
| | - E M Krupitsky
- Bekhterev National Medical Research Center of Psychiatry and Neurology, St. Petersburg, Russia.,Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia
| |
Collapse
|