1
|
Ehlers CL, Wills D, Benedict J, Amodeo LR. Use of a Fitbit-like device in rats: Sex differences, relation to EEG sleep, and use to measure the long-term effects of adolescent ethanol exposure. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1055-1066. [PMID: 37335518 PMCID: PMC10330894 DOI: 10.1111/acer.15079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/27/2023] [Accepted: 04/01/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Sleep difficulties and rhythm disturbances are some of the problems associated with adolescent binge drinking. Recently, animal models of alcohol-induced insomnia have been developed. However, studies in human subjects have recently focused not only on nighttime EEG findings but also on daytime sleepiness and disrupted activity levels as typically measured by activity tracking devices such as the "Fitbit." We sought to develop and test a Fitbit-like device (the "FitBite") in rats and use it to track rest-activity cycles following adolescent alcohol exposure. METHODS The effects of 5 weeks of adolescent ethanol vapor or control conditions were evaluated in 48 male and female Wistar rats using FitBite activity while intoxicated, and during acute (24 h post-vapor exposure) and chronic withdrawal (4 weeks post-vapor exposure). Data were analyzed using activity count and cosinor analyses. Fourteen rats were subsequently implanted with cortical electrodes, and data from the FitBite were compared with EEG data to determine how well the FitBite could identify sleep and activity cycles. RESULTS Female rats were generally more active than males, with higher circadian rhythm amplitudes and mesors (rhythm-adjusted means) across a 24-h period. There were significant correlations between EEG-estimated sleep and activity counts using the FitBite. When the rats were tested during intoxication after 4 weeks of ethanol vapor exposure, they had significantly less overall activity. Disruptions in circadian rhythm were also found with significant decreases in the circadian amplitude, mesor, and a later shift in the acrophase. At 24 h of ethanol withdrawal, rats had more episodes of activity with shorter durations during the daytime, when rats are expected to spend more of their time sleeping. This effect remained at 4 weeks following withdrawal, but circadian rhythm disruptions were no longer present. CONCLUSIONS A Fitbit-like device can be successfully used in rats to assess rest-activity cycles. Adolescent alcohol exposure produced circadian rhythm disturbances that were not observed after withdrawal. Fragmentation of ultradian rest-activity cycles during the light period was found at 24 h and 4 weeks after withdrawal and support data demonstrating the presence of sleep disturbance long after alcohol withdrawal.
Collapse
Affiliation(s)
- Cindy L. Ehlers
- Department of Neuroscience, The Scripps Research Institute, La Jolla CA 92037
| | - Derek Wills
- Department of Neuroscience, The Scripps Research Institute, La Jolla CA 92037
| | - Jessica Benedict
- Department of Neuroscience, The Scripps Research Institute, La Jolla CA 92037
| | - Leslie R. Amodeo
- Department of Psychology, California State University San Bernardino, San Bernardino CA 92407
| |
Collapse
|
2
|
Burgess HJ, Rizvydeen M, Kikyo F, Kebbeh N, Tan M, Roecklein KA, Hasler BP, King AC, Cao D. Sleep and circadian differences between light and heavy adult alcohol drinkers. Alcohol Clin Exp Res 2022; 46:1181-1191. [PMID: 35908247 PMCID: PMC9357170 DOI: 10.1111/acer.14872] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 12/01/2022]
Abstract
Background Numerous studies have reported that eveningness is associated with increased alcohol consumption. However, biological markers of circadian timing, such as dim light melatonin onset (DLMO) and circadian photoreceptor responsivity (post‐illumination pupil response, PIPR), have rarely been assessed in the context of habitual alcohol consumption. This study aimed to examine sleep, circadian timing, and photoreceptor responsivity in adult alcohol drinkers. Methods Participants (21 to 45 years) included 28 light and 50 heavy drinkers. The 8‐day study consisted of a week of adlib sleep monitored with wrist actigraphy, followed by a 9‐h laboratory session with a photoreceptor responsivity and circadian phase assessment. Results The heavy drinkers obtained on average 28 more minutes of sleep (p = 0.002) and reported more eveningness than the light drinkers (p = 0.029). There was a trend for a shorter DLMO‐midsleep interval (p = 0.059) in the heavy drinkers, reflecting a tendency for them to sleep at an earlier circadian phase. The PIPR in the heavy drinkers was significantly smaller than in the light drinkers (p = 0.032), suggesting reduced circadian photoreceptor responsivity in the heavy drinkers. A larger PIPR was significantly associated with a later DLMO in the light drinkers (r = 0.44, p = 0.019), but this relationship was absent in the heavy drinkers (r = −0.01, p = 0.94). Conclusions These results are consistent with earlier reports of more eveningness and a shorter DLMO‐midsleep interval being associated with heavier alcohol drinking. The novel finding of reduced circadian photoreceptor responsivity in heavy drinkers is consistent with prior rodent studies. Future studies should explore the impact of habitual alcohol consumption on other measures of circadian photoreceptor responsivity.
Collapse
Affiliation(s)
- Helen J Burgess
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Muneer Rizvydeen
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Fumitaka Kikyo
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Nema Kebbeh
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Tan
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kathryn A Roecklein
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brant P Hasler
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrea C King
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, USA
| | - Dingcai Cao
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
3
|
Hasler BP, Bruce S, Scharf D, Ngari W, Clark DB. Circadian misalignment and weekend alcohol use in late adolescent drinkers: preliminary evidence. Chronobiol Int 2019; 36:796-810. [PMID: 30950299 DOI: 10.1080/07420528.2019.1586720] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Alcohol use accelerates during late adolescence, predicting the development of alcohol use disorders (AUDs) and other negative outcomes. Identifying modifiable risk factors for alcohol use during this time could lead to novel prevention approaches. Burgeoning evidence suggests that sleep and circadian factors are cross-sectionally and longitudinally linked to alcohol use and problems, but more proximal relationships have been understudied. Circadian misalignment, in particular, is hypothesized to increase the risk for AUDs, but almost no published studies have included a biological measure of misalignment. In the present study, we aimed to extend existing research by assessing the relationship between adolescent circadian misalignment and alcohol use on a proximal timeframe (over two weeks) and by including three complementary measures of circadian alignment. We studied 36 healthy late (18-22 years old, 22 females) alcohol drinkers (reporting ≥1, standard drink per week over the past 30 days) over 14 days. Throughout the study, participants reported prior day's alcohol use and prior night's sleep each morning via smartphone and a secure, browser-based interface. Circadian phase was assessed via the dim light melatonin onset (DLMO) in the laboratory on two occasions (Thursday and Sunday nights) in counterbalanced order. The three measures of circadian alignment included DLMO-midsleep interval, "classic" social jet lag (weekday-weekend difference in midsleep), and "objective" social jet lag (weekday-weekend difference in DLMO). Multivariate imputation by chained equations was used to impute missing data, and Poisson regression models were used to assess associations between circadian alignment variables and weekend alcohol use. Covariates included sex, age, Thursday alcohol use, and Thursday sleep characteristics. As predicted, greater misalignment was associated with greater weekend alcohol use for two of the three alignment measures (shorter DLMO-midsleep intervals and larger weekday-weekend differences in midsleep), while larger weekday-weekend differences in DLMO were associated with less alcohol use. Notably, in contrast to expectations, the distribution of weekday-weekend differences in DLMO was nearly equally distributed between individuals advancing over the weekend and those delaying over the weekend. This unexpected finding plausibly reflects the fact that college students are not subject to the same systematically earlier weekday schedules observed in high school students and working adults. These preliminary findings support the need for larger, more definitive studies investigating the proximal relationships between circadian alignment and alcohol use among late adolescents.
Collapse
Affiliation(s)
- Brant P Hasler
- a Department of Psychiatry , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Scott Bruce
- b Department of Statistics , George Mason University , Fairfax , VA , USA
| | - Deborah Scharf
- c Department of Psychology , Lakehead University , Thunder Bay , Canada
| | - Wambui Ngari
- a Department of Psychiatry , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Duncan B Clark
- a Department of Psychiatry , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| |
Collapse
|
4
|
Capri KM, Maroni MJ, Deane HV, Pierre A, Adams AM, Goncalves FL, Meyer AS, Seggio JA. Effects of time of day and constant light on the behavioral responses and ethanol metabolism to acute alcohol administration in male Black Swiss mice. BIOL RHYTHM RES 2018. [DOI: 10.1080/09291016.2018.1543640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Kimberly M. Capri
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Marissa J. Maroni
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Hannah V. Deane
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Audeline Pierre
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Abigail M. Adams
- Department of Mathematics, Bridgewater State University, Bridgewater, MA, USA
| | - Fatiana L. Goncalves
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Andrew S. Meyer
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Joseph A. Seggio
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| |
Collapse
|
5
|
Ruby CL, Paye G, Fabi JL, Zhang J, Risinger MO, Palmer KN, Verbanes NM, D'Angelo A, Watts TM, Mabe L, Swartzwelder HS. Sex Differences in Photic Entrainment and Sensitivity to Ethanol-Induced Chronodisruption in Adult Mice After Adolescent Intermittent Ethanol Exposure. Alcohol Clin Exp Res 2018; 42:2144-2159. [PMID: 30102762 DOI: 10.1111/acer.13867] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/07/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND Evidence supports a role for the circadian system in alcohol use disorders, but the impact of adolescent alcohol exposure on circadian timing later in life is unknown. Acute ethanol (EtOH) attenuates circadian photic phase-resetting in adult, but not adolescent, rodents. However, nearly all studies have focused on males and it is unknown whether this adolescent-typical insensitivity to EtOH persists into adulthood after adolescent drinking. METHODS Circadian activity was monitored in C57BL/6J mice receiving adolescent intermittent EtOH (AIE) exposure (15% EtOH and water every other day throughout adolescence) or water alone followed by 24 days wherein EtOH was not available (washout). Mice then received a challenge dose of EtOH (1.5 g/kg, intraperitoneal) or saline 15 minutes prior to a 30-minute phase-delaying light pulse and then were released into constant darkness (DD). To control for possible phase-shifting by EtOH challenge alone, a separate group of mice underwent AIE exposure (or water-only) and washout and then received an EtOH or saline injection, but did not receive a light pulse prior to DD. RESULTS Striking sex differences in nearly all measures of circadian photic entrainment were observed during adolescence but AIE effects were subtle and few. Only EtOH-naïve adult male mice showed attenuated photic phase-shifts with EtOH challenge, while all other groups showed normal phase-resetting responses to light. AIE-exposed females showed a persistent delay in activity offset. CONCLUSIONS Adult male AIE-exposed mice retained adolescent-like insensitivity to EtOH-induced suppression of photic phase-resetting, suggesting AIE-induced "lock-in" of an adolescent behavioral phenotype. Adult AIE-exposed females showed delayed initiation of the rest phase. Our results also indicate that intermittent EtOH drinking has subtle effects on circadian activity in mice during adolescence that differ from previously reported effects on adult males. The observed sex differences in circadian activity, EtOH consumption and preference, and responses to EtOH challenge merit future mechanistic study.
Collapse
Affiliation(s)
- Christina L Ruby
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Gerneleh Paye
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Jason L Fabi
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Jiawen Zhang
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Megan O Risinger
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Kaitlyn N Palmer
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Natalie M Verbanes
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Ariana D'Angelo
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Tia M Watts
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Lauren Mabe
- Department of Biology , Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - H Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences , Duke University, Durham, North Carolina
| |
Collapse
|
6
|
Ruby CL, Verbanes NM, Palmer KN, Zisk CF, Bunion DJ, Marinos LN. Caffeine Delays Light-entrained Activity and Potentiates Circadian Photic Phase-resetting in Mice. J Biol Rhythms 2018; 33:523-534. [DOI: 10.1177/0748730418789236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Caffeine is widely used to reduce sedation and increase alertness. However, long-term caffeine use may disrupt circadian (daily, 24-h) rhythms and thereby negatively affect health. Here, we examined the effect of caffeine on photic regulation of circadian activity rhythms in mice. We found that entrainment to a standard 12-h light, 12-h dark (LD) photocycle was delayed during oral self-administration of caffeine. Both acute, high-dose caffeine and chronic, oral caffeine exposure potentiated photic phase-delays in mice, suggesting a possible mechanism by which entrainment to LD was delayed. The effect of caffeine on photic phase-resetting was mimicked by administration of adenosine A1, but not A2A, receptor antagonist in mice. Our results support the hypothesis that caffeine interferes with the ability of the circadian clock to respond normally to light.
Collapse
|
7
|
Yaw AM, Woodruff RW, Prosser RA, Glass JD. Paternal Cocaine Disrupts Offspring Circadian Clock Function in a Sex-Dependent Manner in Mice. Neuroscience 2018; 379:257-268. [DOI: 10.1016/j.neuroscience.2018.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/20/2018] [Accepted: 03/09/2018] [Indexed: 10/17/2022]
|
8
|
Gulick D, Gamsby JJ. Racing the clock: The role of circadian rhythmicity in addiction across the lifespan. Pharmacol Ther 2018; 188:124-139. [PMID: 29551440 DOI: 10.1016/j.pharmthera.2018.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although potent effects of psychoactive drugs on circadian rhythms were first described over 30 years ago, research into the reciprocal relationship between the reward system and the circadian system - and the impact of this relationship on addiction - has only become a focus in the last decade. Nonetheless, great progress has been made in that short time toward understanding how drugs of abuse impact the molecular and physiological circadian clocks, as well as how disruption of normal circadian rhythm biology may contribute to addiction and ameliorate the efficacy of treatments for addiction. In particular, data have emerged demonstrating that disrupted circadian rhythms, such as those observed in shift workers and adolescents, increase susceptibility to addiction. Furthermore, circadian rhythms and addiction impact one another longitudinally - specifically from adolescence to the elderly. In this review, the current understanding of how the circadian clock interacts with substances of abuse within the context of age-dependent changes in rhythmicity, including the potential existence of a drug-sensitive clock, the correlation between chronotype and addiction vulnerability, and the importance of rhythmicity in the mesocorticolimbic dopamine system, is discussed. The primary focus is on alcohol addiction, as the preponderance of research is in this area, with references to other addictions as warranted. The implications of clock-drug interactions for the treatment of addiction will also be reviewed, and the potential of therapeutics that reset the circadian rhythm will be highlighted.
Collapse
Affiliation(s)
- Danielle Gulick
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Joshua J Gamsby
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
9
|
Davis BT, Voigt RM, Shaikh M, Forsyth CB, Keshavarzian A. Circadian Mechanisms in Alcohol Use Disorder and Tissue Injury. Alcohol Clin Exp Res 2018; 42:668-677. [PMID: 29450896 DOI: 10.1111/acer.13612] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/06/2018] [Indexed: 12/12/2022]
Abstract
Heavy use of alcohol can lead to addictive behaviors and to eventual alcohol-related tissue damage. While increased consumption of alcohol has been attributed to various factors including level of alcohol exposure and environmental factors such as stress, data from behavioral scientists and physiological researchers are revealing roles for the circadian rhythm in mediating the development of behaviors associated with alcohol use disorder as well as the tissue damage that drives physiological disease. In this work, we compile recent work on the complex mutually influential relationship that exists between the core circadian rhythm and the pharmacodynamics of alcohol. As we do so, we highlight implications of the relationship between alcohol and common circadian mechanisms of effected organs on alcohol consumption, metabolism, toxicity, and pathology.
Collapse
Affiliation(s)
| | | | | | | | - Ali Keshavarzian
- Division of Digestive Disease and Nutrition, Section of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
10
|
Lindberg D, Andres-Beck L, Jia YF, Kang S, Choi DS. Purinergic Signaling in Neuron-Astrocyte Interactions, Circadian Rhythms, and Alcohol Use Disorder. Front Physiol 2018; 9:9. [PMID: 29467662 PMCID: PMC5808134 DOI: 10.3389/fphys.2018.00009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/05/2018] [Indexed: 12/17/2022] Open
Abstract
Alcohol use disorder (AUD) is a debilitating condition marked by cyclic patterns of craving, use, and withdrawal. These pathological behaviors are mediated by multiple neurotransmitter systems utilizing glutamate, GABA, dopamine, ATP, and adenosine. In particular, purines such as ATP and adenosine have been demonstrated to alter the phase and function of the circadian clock and are reciprocally regulated by the clock itself. Importantly, chronic ethanol intake has been demonstrated to disrupt the molecular circadian clock and is associated with altered circadian patterns of activity and sleep. Moreover, ethanol has been demonstrated to disrupt purinergic signaling, while dysfunction of the purinergic system has been implicated in conditions of drug abuse such as AUD. In this review, we summarize our current knowledge regarding circadian disruption by ethanol, focusing on the reciprocal relationship that exists between oscillatory neurotransmission and the molecular circadian clock. In particular, we offer detailed explanations and hypotheses regarding the concerted regulation of purinergic signaling and circadian oscillations by neurons and astrocytes, and review the diverse mechanisms by which purinergic dysfuction may contribute to circadian disruption or alcohol abuse. Finally, we describe the mechanisms by which ethanol may disrupt or hijack endogenous circadian rhythms to induce the maladaptive behavioral patterns associated with AUD.
Collapse
Affiliation(s)
- Daniel Lindberg
- Neurobiology of Disease, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Lindsey Andres-Beck
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Yun-Fang Jia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Seungwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Doo-Sup Choi
- Neurobiology of Disease, Mayo Clinic College of Medicine, Rochester, MN, United States.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, United States.,Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, United States
| |
Collapse
|
11
|
Lindsay JH, Prosser RA. The Mammalian Circadian Clock Exhibits Chronic Ethanol Tolerance and Withdrawal-Induced Glutamate Hypersensitivity, Accompanied by Changes in Glutamate and TrkB Receptor Proteins. Alcohol Clin Exp Res 2017; 42:315-328. [PMID: 29139560 DOI: 10.1111/acer.13554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/07/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND Alcohol tolerance and withdrawal-induced effects are criteria for alcohol use disorders listed by the DSM-V. Although tolerance and withdrawal have been studied over many decades, there is still uncertainty regarding mechanistic distinctions that characterize these different forms of ethanol (EtOH)-induced plasticity. Previously, we demonstrated that the suprachiasmatic nucleus (SCN) circadian clock develops both acute and rapid tolerance to EtOH inhibition of glutamate-induced circadian phase shifts. Here, we demonstrate that chronic EtOH tolerance and withdrawal-induced glutamate hypersensitivity occur in vitro and that rapid tolerance, chronic tolerance, and glutamate hypersensitivity have distinct cellular changes. METHODS We use single-unit extracellular electrophysiological recordings to determine whether chronic tolerance to EtOH inhibition of glutamatergic phase shifts and withdrawal-induced glutamate hypersensitivity develop in the SCN. We use Western blotting to compare phosphorylation state and total expression of N-methyl-D-aspartate (NMDA) receptor subunits and associated proteins in the SCN after mice were exposed to varying EtOH consumption paradigms. RESULTS Chronic tolerance developed after a minimum of 8 days of 4 h/d EtOH access, as indicated by a decreased sensitivity to EtOH inhibition of glutamate-induced phase shifts. We also observed an increased sensitivity to glutamate-induced phase shifts in SCN tissue following withdrawal. We demonstrated an increase in the ratio of NR2B:NR2A NMDA receptor subunit expression after 21 days, but not after 10 days of EtOH drinking. This increase persisted during EtOH withdrawal, along with an increase in NR2B Y1472 phosphorylation, mature brain-derived neurotrophic factor, and phosphorylated TrkB. CONCLUSIONS These results demonstrate that multiple tolerance forms and withdrawal-induced glutamate hypersensitivity occur in the SCN and that these different forms of EtOH-induced plasticity are accompanied by distinct changes in cellular physiology. Importantly, this study further demonstrates the power of using the SCN as a model system to investigate EtOH-induced plasticity.
Collapse
Affiliation(s)
- Jonathan H Lindsay
- Department of Biochemistry and Cellular and Molecular Biology (JHL, RAP), University of Tennessee Knoxville, NeuroNET Research Center, Knoxville, Tennessee
| | - Rebecca A Prosser
- Department of Biochemistry and Cellular and Molecular Biology (JHL, RAP), University of Tennessee Knoxville, NeuroNET Research Center, Knoxville, Tennessee
| |
Collapse
|
12
|
De Nobrega AK, Lyons LC. Drosophila: An Emergent Model for Delineating Interactions between the Circadian Clock and Drugs of Abuse. Neural Plast 2017; 2017:4723836. [PMID: 29391952 PMCID: PMC5748135 DOI: 10.1155/2017/4723836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/13/2017] [Indexed: 01/12/2023] Open
Abstract
Endogenous circadian oscillators orchestrate rhythms at the cellular, physiological, and behavioral levels across species to coordinate activity, for example, sleep/wake cycles, metabolism, and learning and memory, with predictable environmental cycles. The 21st century has seen a dramatic rise in the incidence of circadian and sleep disorders with globalization, technological advances, and the use of personal electronics. The circadian clock modulates alcohol- and drug-induced behaviors with circadian misalignment contributing to increased substance use and abuse. Invertebrate models, such as Drosophila melanogaster, have proven invaluable for the identification of genetic and molecular mechanisms underlying highly conserved processes including the circadian clock, drug tolerance, and reward systems. In this review, we highlight the contributions of Drosophila as a model system for understanding the bidirectional interactions between the circadian system and the drugs of abuse, alcohol and cocaine, and illustrate the highly conserved nature of these interactions between Drosophila and mammalian systems. Research in Drosophila provides mechanistic insights into the corresponding behaviors in higher organisms and can be used as a guide for targeted inquiries in mammals.
Collapse
Affiliation(s)
- Aliza K. De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Lisa C. Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
13
|
Albers HE, Walton JC, Gamble KL, McNeill JK, Hummer DL. The dynamics of GABA signaling: Revelations from the circadian pacemaker in the suprachiasmatic nucleus. Front Neuroendocrinol 2017; 44:35-82. [PMID: 27894927 PMCID: PMC5225159 DOI: 10.1016/j.yfrne.2016.11.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022]
Abstract
Virtually every neuron within the suprachiasmatic nucleus (SCN) communicates via GABAergic signaling. The extracellular levels of GABA within the SCN are determined by a complex interaction of synthesis and transport, as well as synaptic and non-synaptic release. The response to GABA is mediated by GABAA receptors that respond to both phasic and tonic GABA release and that can produce excitatory as well as inhibitory cellular responses. GABA also influences circadian control through the exclusively inhibitory effects of GABAB receptors. Both GABA and neuropeptide signaling occur within the SCN, although the functional consequences of the interactions of these signals are not well understood. This review considers the role of GABA in the circadian pacemaker, in the mechanisms responsible for the generation of circadian rhythms, in the ability of non-photic stimuli to reset the phase of the pacemaker, and in the ability of the day-night cycle to entrain the pacemaker.
Collapse
Affiliation(s)
- H Elliott Albers
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States.
| | - James C Walton
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - John K McNeill
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Daniel L Hummer
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Department of Psychology, Morehouse College, Atlanta, GA 30314, United States
| |
Collapse
|
14
|
Ruby CL, Palmer KN, Zhang J, Risinger MO, Butkowski MA, Swartzwelder HS. Differential Sensitivity to Ethanol-Induced Circadian Rhythm Disruption in Adolescent and Adult Mice. Alcohol Clin Exp Res 2016; 41:187-196. [PMID: 27997028 DOI: 10.1111/acer.13275] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/19/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Growing evidence supports a central role for the circadian system in alcohol use disorders, but few studies have examined this relationship during adolescence. In mammals, circadian rhythms are regulated by the suprachiasmatic nucleus, a biological clock whose timing is synchronized (reset) to the environment primarily by light (photic) input. Alcohol (ethanol [EtOH]) disrupts circadian timing in part by attenuating photic phase-resetting responses in adult rodents. However, circadian rhythms change throughout life and it is not yet known whether EtOH has similar effects on circadian regulation during adolescence. METHODS General circadian locomotor activity was monitored in male C57BL6/J mice beginning in adolescence (P27) or adulthood (P61) in a 12-hour light, 12-hour dark photocycle for ~2 weeks to establish baseline circadian activity measures. On the day of the experiment, mice received an acute injection of EtOH (1.5 g/kg, i.p.) or equal volume saline 15 minutes prior to a 30-minute light pulse at Zeitgeber Time 14 (2 hours into the dark phase) and then were released into constant darkness (DD) for ~2 weeks to assess phase-resetting responses. Control mice of each age-group received injections but no light pulse prior to DD. RESULTS While adults showed the expected decrease in photic phase-delays induced by acute EtOH, this effect was absent in adolescent mice. Adolescents also showed baseline differences in circadian rhythmicity compared to adults, including advanced photocycle entrainment, larger photic phase-delays, a shorter free-running (endogenous) circadian period, and greater circadian rhythm amplitude. CONCLUSIONS Collectively, our results indicate that adolescent mice are less sensitive to the effect of EtOH on circadian photic phase-resetting and that their daily activity rhythms are markedly different than those of adults.
Collapse
Affiliation(s)
- Christina L Ruby
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Kaitlyn N Palmer
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Jiawen Zhang
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Megan O Risinger
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - Melissa A Butkowski
- Department of Biology, Indiana University of Pennsylvania, Indiana, Pennsylvania
| | - H Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina.,Neurobiology Research Laboratory, VA Medical Center, Durham, North Carolina
| |
Collapse
|
15
|
Burgess HJ, Rizvydeen M, Fogg LF, Keshavarzian A. A single dose of alcohol does not meaningfully alter circadian phase advances and phase delays to light in humans. Am J Physiol Regul Integr Comp Physiol 2016; 310:R759-65. [PMID: 26936778 PMCID: PMC4867406 DOI: 10.1152/ajpregu.00001.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/29/2016] [Indexed: 01/13/2023]
Abstract
Central circadian timing influences mental and physical health. Research in nocturnal rodents has demonstrated that when alcohol is consumed, it reaches the central hypothalamic circadian pacemaker (suprachiasmatic nuclei) and can directly alter circadian phase shifts to light. In two separate studies, we examined, for the first time, the effects of a single dose of alcohol on circadian phase advances and phase delays to light in humans. Two 23-day within-subjects placebo-controlled counterbalanced design studies were conducted. Both studies consisted of 6 days of fixed baseline sleep to stabilize circadian timing, a 2-day laboratory session, a 6-day break, and a repeat of 6 days of fixed sleep and a 2-day laboratory session. In the phase advance study (n= 10 light drinkers, 24-45 yr), the laboratory sessions consisted of a baseline dim light phase assessment, sleep episode, alcohol (0.6 g/kg) or placebo, 2-h morning bright light pulse, and final phase assessment. In the phase-delay study (n= 14 light drinkers, 22-44 yr), the laboratory sessions consisted of a baseline phase assessment, alcohol (0.8 g/kg) or placebo, 2-h late night bright light pulse, sleep episode, and final phase assessment. In both studies, alcohol either increased or decreased the observed phase shifts to light (interaction P≥ 0.46), but the effect of alcohol vs. placebo on phase shifts to light was always on average smaller than 30 min. Thus, no meaningful effects of a single dose of alcohol vs. placebo on circadian phase shifts to light in humans were observed.
Collapse
Affiliation(s)
- Helen J Burgess
- Biological Rhythms Research Laboratory, Department of Behavioral Sciences, Rush University Medical Center, Chicago, Illinois;
| | - Muneer Rizvydeen
- Biological Rhythms Research Laboratory, Department of Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Louis F Fogg
- College of Nursing, Rush University Medical Center, Chicago, Illinois; and
| | - Ali Keshavarzian
- Department of Digestive Diseases, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
16
|
Liao J, Seggio JA, Ahmad ST. Mutations in the circadian gene period alter behavioral and biochemical responses to ethanol in Drosophila. Behav Brain Res 2016; 302:213-9. [PMID: 26802726 DOI: 10.1016/j.bbr.2016.01.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/12/2016] [Accepted: 01/17/2016] [Indexed: 11/26/2022]
Abstract
Clock genes, such as period, which maintain an organism's circadian rhythm, can have profound effects on metabolic activity, including ethanol metabolism. In turn, ethanol exposure has been shown in Drosophila and mammals to cause disruptions of the circadian rhythm. Previous studies from our labs have shown that larval ethanol exposure disrupted the free-running period and period expression of Drosophila. In addition, a recent study has shown that arrhythmic flies show no tolerance to ethanol exposure. As such, Drosophila period mutants, which have either a shorter than wild-type free-running period (perS) or a longer one (perL), may also exhibit altered responses to ethanol due to their intrinsic circadian differences. In this study, we tested the initial sensitivity and tolerance of ethanol exposure on Canton-S, perS, and perL, and then measured their Alcohol Dehydrogenase (ADH) and body ethanol levels. We showed that perL flies had slower sedation rate, longer recovery from ethanol sedation, and generated higher tolerance for sedation upon repeated ethanol exposure compared to Canton-S wild-type flies. Furthermore, perL flies had lower ADH activity and had a slower ethanol clearance compared to wild-type flies. The findings of this study suggest that period mutations influence ethanol induced behavior and ethanol metabolism in Drosophila and that flies with longer circadian periods are more sensitive to ethanol exposure.
Collapse
Affiliation(s)
- Jennifer Liao
- Department of Biology, 5720 Mayflower Hill Dr., Colby College, Waterville, ME 04901, USA.
| | - Joseph A Seggio
- Department of Biological Sciences, 24 Park Ave., Bridgewater State University, Bridgewater, MA 02325, USA.
| | - S Tariq Ahmad
- Department of Biology, 5720 Mayflower Hill Dr., Colby College, Waterville, ME 04901, USA.
| |
Collapse
|
17
|
Chronobiology of ethanol: animal models. Alcohol 2015; 49:311-9. [PMID: 25971539 DOI: 10.1016/j.alcohol.2015.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/10/2015] [Accepted: 04/10/2015] [Indexed: 01/08/2023]
Abstract
Clinical and epidemiological observations have revealed that alcohol abuse and alcoholism are associated with widespread disruptions in sleep and other circadian biological rhythms. As with other psychiatric disorders, animal models have been very useful in efforts to better understand the cause and effect relationships underlying the largely correlative human data. This review summarizes the experimental findings indicating bidirectional interactions between alcohol (ethanol) consumption and the circadian timing system, emphasizing behavioral studies conducted in the author's laboratory. Together with convergent evidence from multiple laboratories, the work summarized here establishes that ethanol intake (or administration) alters fundamental properties of the underlying circadian pacemaker. In turn, circadian disruption induced by either environmental or genetic manipulations can alter voluntary ethanol intake. These reciprocal interactions may create a vicious cycle that contributes to the downward spiral of alcohol and drug addiction. In the future, such studies may lead to the development of chronobiologically based interventions to prevent relapse and effectively mitigate some of the societal burden associated with such disorders.
Collapse
|
18
|
Nascimento NF, Carlson KN, Amaral DN, Logan RW, Seggio JA. Alcohol and lithium have opposing effects on the period and phase of the behavioral free-running activity rhythm. Alcohol 2015; 49:367-76. [PMID: 25850902 DOI: 10.1016/j.alcohol.2015.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/30/2015] [Accepted: 02/04/2015] [Indexed: 12/15/2022]
Abstract
Bipolar patients have a high prevalence of comorbid alcohol use and abuse disorders, while chronic alcohol drinking may increase the presence and severity of certain symptoms of bipolar disorder. As such, there may be many individuals that are prescribed lithium to alleviate the manic symptoms of bipolar disorder, but also drink alcohol concurrently. In addition, both alcoholics and individuals with bipolar disorder often exhibit disruptions to their sleep-wake cycles and other circadian rhythms. Interestingly, both ethanol and lithium are known to alter both the period and the phase of free-running rhythms in mammals. While lithium is known to lengthen the period, ethanol seems to shorten the period and attenuate the responses to acute light pulses. Therefore, the present study aimed to determine whether ethanol and lithium have opposing effects on the circadian pacemaker when administered together. C57BL/6J mice were provided drinking solutions containing lithium, alcohol, or both, and their free-running rhythms along with their response to photic phase shifts were investigated. Mice treated with lithium displayed period lengthening, which was almost completely negated when ethanol was added. Moreover, ethanol significantly attenuated light-induced phase delays while the addition of lithium partially restored this response. These results indicate that alcohol and lithium have opposing effects on behavioral circadian rhythms. Individuals with bipolar disorder who are prescribed lithium and who drink alcohol might be inadvertently altering their sleep and circadian cycles, which may exacerbate their symptoms.
Collapse
Affiliation(s)
- Nara F Nascimento
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Karen N Carlson
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Danielle N Amaral
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, USA
| | - Joseph A Seggio
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA.
| |
Collapse
|
19
|
Hasler BP, Soehner AM, Clark DB. Sleep and circadian contributions to adolescent alcohol use disorder. Alcohol 2015; 49:377-87. [PMID: 25442171 DOI: 10.1016/j.alcohol.2014.06.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 06/27/2014] [Accepted: 06/30/2014] [Indexed: 11/16/2022]
Abstract
Adolescence is a time of marked changes across sleep, circadian rhythms, brain function, and alcohol use. Starting at puberty, adolescents' endogenous circadian rhythms and preferred sleep times shift later, often leading to a mismatch with the schedules imposed by secondary education. This mismatch induces circadian misalignment and sleep loss, which have been associated with affect dysregulation, increased drug and alcohol use, and other risk-taking behaviors in adolescents and adults. In parallel to developmental changes in sleep, adolescent brains are undergoing structural and functional changes in the circuits subserving the pursuit and processing of rewards. These developmental changes in reward processing likely contribute to the initiation of alcohol use during adolescence. Abundant evidence indicates that sleep and circadian rhythms modulate reward function, suggesting that adolescent sleep and circadian disturbance may contribute to altered reward function, and in turn, alcohol involvement. In this review, we summarize the relevant evidence and propose that these parallel developmental changes in sleep, circadian rhythms, and neural processing of reward interact to increase risk for alcohol use disorder (AUD).
Collapse
Affiliation(s)
- Brant P Hasler
- Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, Pittsburgh, PA 15213, USA.
| | - Adriane M Soehner
- Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, Pittsburgh, PA 15213, USA
| | - Duncan B Clark
- Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
20
|
Prosser RA, Glass JD. Assessing ethanol's actions in the suprachiasmatic circadian clock using in vivo and in vitro approaches. Alcohol 2015; 49:321-339. [PMID: 25457753 DOI: 10.1016/j.alcohol.2014.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/07/2014] [Accepted: 07/09/2014] [Indexed: 12/18/2022]
Abstract
Research over the past decade has demonstrated substantial interactions between the circadian system and the processes through which alcohol affects behavior and physiology. Here we summarize the results of our collaborative efforts focused on this intersection. Using a combination of in vivo and in vitro approaches, we have shown that ethanol affects many aspects of the mammalian circadian system, both acutely as well as after chronic administration. Conversely, we have shown circadian influences on ethanol consumption. Importantly, we are beginning to delve into the cellular mechanisms associated with these effects. We are also starting to form a picture of the neuroanatomical bases for many of these actions. Finally, we put our current findings into perspective by suggesting new avenues of inquiry for our future efforts.
Collapse
|
21
|
Parekh PK, Ozburn AR, McClung CA. Circadian clock genes: effects on dopamine, reward and addiction. Alcohol 2015; 49:341-9. [PMID: 25641765 DOI: 10.1016/j.alcohol.2014.09.034] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 12/17/2022]
Abstract
Addiction is a widespread public health issue with social and economic ramifications. Substance abuse disorders are often accompanied by disruptions in circadian rhythms including sleep/wake cycles, which can exacerbate symptoms of addiction and dependence. Additionally, genetic disturbance of circadian molecular mechanisms can predispose some individuals to substance abuse disorders. In this review, we will discuss how circadian genes can regulate midbrain dopaminergic activity and subsequently, drug intake and reward. We will also suggest future directions for research on circadian genes and drugs of abuse.
Collapse
|
22
|
Rosenwasser AM, McCulley WD, Fecteau M. Circadian activity rhythms and voluntary ethanol intake in male and female ethanol-preferring rats: effects of long-term ethanol access. Alcohol 2014; 48:647-55. [PMID: 25281289 DOI: 10.1016/j.alcohol.2014.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/12/2014] [Accepted: 07/01/2014] [Indexed: 01/13/2023]
Abstract
Chronic alcohol (ethanol) intake alters fundamental properties of the circadian clock. While previous studies have reported significant alterations in free-running circadian period during chronic ethanol access, these effects are typically subtle and appear to require high levels of intake. In the present study we examined the effects of long-term voluntary ethanol intake on ethanol consumption and free-running circadian period in male and female, selectively bred ethanol-preferring P and HAD2 rats. In light of previous reports that intermittent access can result in escalated ethanol intake, an initial 2-week water-only baseline was followed by either continuous or intermittent ethanol access (i.e., alternating 15-day epochs of ethanol access and ethanol deprivation) in separate groups of rats. Thus, animals were exposed to either 135 days of continuous ethanol access or to five 15-day access periods alternating with four 15-day periods of ethanol deprivation. Animals were maintained individually in running-wheel cages under continuous darkness throughout the experiment to allow monitoring of free-running activity and drinking rhythms, and 10% (v/v) ethanol and plain water were available continuously via separate drinking tubes during ethanol access. While there were no initial sex differences in ethanol drinking, ethanol preference increased progressively in male P and HAD2 rats under both continuous and intermittent-access conditions, and eventually exceeded that seen in females. Free-running period shortened during the initial ethanol-access epoch in all groups, but the persistence of this effect showed complex dependence on sex, breeding line, and ethanol-access schedule. Finally, while females of both breeding lines displayed higher levels of locomotor activity than males, there was little evidence for modulation of activity level by ethanol access. These results are consistent with previous findings that chronic ethanol intake alters free-running circadian period, and show further that the development of chronobiological tolerance to ethanol may vary by sex and genotype.
Collapse
Affiliation(s)
- Alan M Rosenwasser
- Department of Psychology, University of Maine, Orono, ME 04469, USA; School of Biology and Ecology, University of Maine, Orono, ME 04469, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
| | | | - Matthew Fecteau
- Department of Psychology, University of Maine, Orono, ME 04469, USA
| |
Collapse
|
23
|
Intestinal CYP2E1: A mediator of alcohol-induced gut leakiness. Redox Biol 2014; 3:40-6. [PMID: 25462064 PMCID: PMC4297927 DOI: 10.1016/j.redox.2014.10.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 10/13/2014] [Accepted: 10/15/2014] [Indexed: 02/07/2023] Open
Abstract
Chronic alcohol use can result in many pathological effects including alcoholic liver disease (ALD). While alcohol is necessary for the development of ALD, only 20-30% of alcoholics develop alcoholic steatohepatitis (ASH) with progressive liver disease leading to cirrhosis and liver failure (ALD). This suggests that while chronic alcohol consumption is necessary it is not sufficient to induce clinically relevant liver damage in the absence of a secondary risk factor. Studies in rodent models and alcoholic patients show that increased intestinal permeability to microbial products like endotoxin play a critical role in promoting liver inflammation in ALD pathogenesis. Therefore identifying mechanisms of alcohol-induced intestinal permeability is important in identifying mechanisms of ALD and for designing new avenues for therapy. Cyp2e1 is a cytochrome P450 enzyme that metabolizes alcohol has been shown to be upregulated by chronic alcohol use and to be a major source of oxidative stress and liver injury in alcoholics and in animal and in vitro models of chronic alcohol use. Because Cyp2e1 is also expressed in the intestine and is upregulated by chronic alcohol use, we hypothesized it could play a role in alcohol-induced intestinal hyperpermeability. Our in vitro studies with intestinal Caco-2 cells and in mice fed alcohol showed that circadian clock proteins CLOCK and PER2 are required for alcohol-induced permeability. We also showed that alcohol increases Cyp2e1 protein and activity but not mRNA in Caco-2 cells and that an inhibitor of oxidative stress or siRNA knockdown of Cyp2e1 prevents the increase in CLOCK or PER2 proteins and prevents alcohol-induced hyperpermeability. With our collaborators we have also shown that Cyp2e1 knockout mice are resistant to alcohol-induced gut leakiness and liver inflammation. Taken together our data support a novel Cyp2e1-circadian clock protein mechanism for alcohol-induced gut leakiness that could provide new avenues for therapy of ALD.
Collapse
|
24
|
Ruby CL, Vadnie CA, Hinton DJ, Abulseoud OA, Walker DL, O'Connor KM, Noterman MF, Choi DS. Adenosinergic regulation of striatal clock gene expression and ethanol intake during constant light. Neuropsychopharmacology 2014; 39:2432-40. [PMID: 24755889 PMCID: PMC4138755 DOI: 10.1038/npp.2014.94] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/09/2014] [Accepted: 04/17/2014] [Indexed: 01/19/2023]
Abstract
Circadian rhythm and sleep disruptions occur frequently in individuals with alcohol use disorders (AUD) and present significant barriers to treatment. Recently, a variant of adenosine transporter, equilibrative nucleoside transporter 1 (ENT1), was associated with the co-occurrence of sleep problems and AUD. We have previously shown that mice lacking ENT1 (ENT1 KO) have reduced adenosine levels in the striatum and drink more alcohol compared with wild types (WT). However, it is unknown whether ENT1 deletion disrupts circadian rhythms, which may contribute to alcohol preference in ENT1 KO mice. Here we used these mice to determine whether endogenous adenosine regulates circadian genetic and behavioral rhythms and influences alcohol intake during chronodisruption. We examined circadian locomotor activity in ENT1 KO vs WT littermates and found that ENT1 KO mice were both active earlier and hyperactive compared with WT mice at night. We used real-time PCR and immunohistochemistry to estimate striatal clock gene levels and found that PER2 expression in the striatum was blunted by ENT1 deletion or A2A receptor (A2AR) antagonism. Next, we exposed ENT1 KO and WT mice to constant light (LL) and found further elevation in ethanol intake in ENT1 KO, but not in WT mice, supporting the notion that circadian dysfunction may contribute to increased alcohol intake in ENT1 KO mice. Finally, we showed that A2AR agonist administration normalized PER1 and PER2 expression and circadian locomotor activity in ENT1 KO mice. Together, our results demonstrate that adenosine signaling regulates cellular and behavioral circadian timing and influences alcohol intake during chronodisruption.
Collapse
Affiliation(s)
- Christina L Ruby
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA,Department of Biology, Indiana University of Pennsylvania, Indiana, PA, USA
| | - Chelsea A Vadnie
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - David J Hinton
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Osama A Abulseoud
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Denise L Walker
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Katheryn M O'Connor
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Maria F Noterman
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA,Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN, USA,Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA, Tel: +1 507 284 5602, Fax: +1 507 266 0824, E-mail:
| |
Collapse
|
25
|
Lindsay JH, Glass JD, Amicarelli M, Prosser RA. The mammalian circadian clock in the suprachiasmatic nucleus exhibits rapid tolerance to ethanol in vivo and in vitro. Alcohol Clin Exp Res 2014; 38:760-9. [PMID: 24512529 DOI: 10.1111/acer.12303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/16/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Ethanol (EtOH) triggers cellular adaptations that induce tolerance in many brain areas, including the suprachiasmatic nucleus (SCN), the site of the master circadian clock. EtOH inhibits light-induced phase shifts in the SCN in vivo and glutamate-induced phase shifts in vitro. The in vitro phase shifts develop acute tolerance to EtOH, occurring within minutes of initial exposure, while the in vivo phase shifts exhibit no evidence of chronic tolerance. An intermediate form, rapid tolerance, is not well studied but may predict subsequent chronic tolerance. Here, we investigated rapid tolerance in the SCN clock. METHODS Adult C57BL/6 mice were provided 15% EtOH or water for one 12-hour lights-off period. For in vitro experiments, SCN-containing brain slices were prepared in the morning and treated for 10 minutes with glutamate +/- EtOH the following night. Single-cell neuronal firing rates were recorded extracellularly during the subsequent day to determine SCN clock phase. For in vivo experiments, mice receiving EtOH 24 hours previously were exposed to a 30-minute light pulse immediately preceded by intraperitoneal saline or 2 g/kg EtOH injection. Mice were then placed in constant darkness and their phase-shifting responses measured. RESULTS In vitro, the SCN clock from EtOH-exposed mice exhibited rapid tolerance, with a 10-fold increase in EtOH needed to inhibit glutamate-induced phase shifts. Co-application of brain-derived neurotrophic factor prevented EtOH inhibition, consistent with experiments using EtOH-naïve mice. Rapid tolerance lasts 48 to 96 hours, depending on whether assessing in vitro phase advances or phase delays. Similarly, in vivo, prior EtOH consumption prevented EtOH's acute blockade of photic phase delays. Finally, immunoblot experiments showed no changes in SCN glutamate receptor subunit (NR2B) expression or phosphorylation in response to rapid tolerance induction. CONCLUSIONS The SCN circadian clock develops rapid tolerance to EtOH as assessed both in vivo and in vitro, and the tolerance lasts for several days. These data demonstrate the utility of the circadian system as a model for investigating cellular mechanisms through which EtOH acts in the brain.
Collapse
Affiliation(s)
- Jonathan H Lindsay
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | | | | | | |
Collapse
|
26
|
Adenosine and glutamate in neuroglial interaction: implications for circadian disorders and alcoholism. ADVANCES IN NEUROBIOLOGY 2014; 11:103-19. [PMID: 25236726 DOI: 10.1007/978-3-319-08894-5_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Recent studies have demonstrated that the function of glia is not restricted to the support of neuronal function. In fact, astrocytes are essential for neuronal activity in the brain and play an important role in the regulation of complex behavior. Astrocytes actively participate in synapse formation and brain information processing by releasing and uptaking glutamate, D-serine, adenosine 5'-triphosphate (ATP), and adenosine. In the central nervous system, adenosine-mediated neuronal activity modulates the actions of other neurotransmitter systems. Adenosinergic fine-tuning of the glutamate system in particular has been shown to regulate circadian rhythmicity and sleep, as well as alcohol-related behavior and drinking. Adenosine gates both photic (light-induced) glutamatergic and nonphotic (alerting) input to the circadian clock located in the suprachiasmatic nucleus of the hypothalamus. Astrocytic, SNARE-mediated ATP release provides the extracellular adenosine that drives homeostatic sleep. Acute ethanol increases extracellular adenosine, which mediates the ataxic and hypnotic/sedative effects of alcohol, while chronic ethanol leads to downregulated adenosine signaling that underlies insomnia, a major predictor of relapse. Studies using mice lacking the equilibrative nucleoside transporter 1 have illuminated how adenosine functions through neuroglial interactions involving glutamate uptake transporter GLT-1 [referred to as excitatory amino acid transporter 2 (EAAT2) in human] and possibly water channel aquaporin 4 to regulate ethanol sensitivity, reward-related motivational processes, and alcohol intake.
Collapse
|
27
|
Damaggio AS, Gorman MR. Circadian phase determines effects of repeated ethanol vapor exposure and withdrawal on body temperature and activity rhythms of male mice. Alcohol Clin Exp Res 2013; 38:879-88. [PMID: 24256465 DOI: 10.1111/acer.12297] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 08/30/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Physiological responses to acute ethanol (EtOH) injection depend critically on the timing of their administration. Whether daily timing modulates effects of longer intoxication intervals characteristic of alcohol-dependent humans remains unknown. The present work examines time-of-day effects during EtOH exposure and withdrawal measured by locomotor activity (ActLoc ) and body temperature (Tb ) across multiple rounds of EtOH exposure/withdrawal. METHODS Two groups of C57BL/6J mice (n = 8 per group), implanted with radio-telemeters, were entrained to opposite light-dark periods (14:10 LD cycle) so that their rest/activity cycles were 12 hours apart. Under a 2-hour skeleton photoperiod animals were simultaneously exposed to 3 daily cycles of EtOH vapor inhalation (14 hours EtOH on) and withdrawal (10 hours EtOH off). During this time, air-only control groups (n = 4 per group) matched for entrainment were handled in a comparable manner. After the third cycle of EtOH vapor, the animals were left undisturbed for 11 days to recover. The 14-day protocol was repeated 3 additional times. RESULTS During intoxication, mice exposed to EtOH in the subjective night exhibited greater hypothermia and more overall disruptions in the Tb and ActLoc rhythms. Acute withdrawal induced hypothermia during the subjective night and hyperthermia during the subjective day. Animals in both phases demonstrated significant disruptions in ActLoc during withdrawal. ActLoc had little effect on Tb during EtOH exposure, but it significantly influenced Tb during acute withdrawal. CONCLUSIONS The physiological responses of both EtOH exposure and withdrawal differ as a function of time of day. These findings suggest that controlling for the circadian phase of exposure and/or withdrawal may mitigate the severity of symptomatic withdrawal.
Collapse
Affiliation(s)
- Amanda S Damaggio
- Center for Chronobiology and Department of Psychology, University of California, San Diego, La Jolla, California
| | | |
Collapse
|
28
|
The role of clock in ethanol-related behaviors. Neuropsychopharmacology 2013; 38:2393-400. [PMID: 23722243 PMCID: PMC3799058 DOI: 10.1038/npp.2013.138] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 02/08/2023]
Abstract
Mice with a mutation in the Clock gene (ClockΔ19) exhibit increased preference for stimulant rewards and sucrose. They also have an increase in dopaminergic activity in the ventral tegmental area (VTA) and a general increase in glutamatergic tone that might underlie these behaviors. However, it is unclear if their phenotype would extend to a very different class of drug (ethanol), and if so, whether these systems might be involved in their response. Continuous access voluntary ethanol intake was evaluated in ClockΔ19 mutants and wild-type (WT) mice. We found that ClockΔ19 mice exhibited significantly increased ethanol intake in a two-bottle choice paradigm. Interestingly, this effect was more robust in female mice. Moreover, chronic ethanol experience resulted in a long-lasting decrease in VTA Clock expression. To determine the importance of VTA Clock expression in ethanol intake, we knocked down Clock expression in the VTA of WT mice via RNA interference. We found that reducing Clock expression in the VTA resulted in significantly increased ethanol intake similar to the ClockΔ19 mice. Interestingly, we also discovered that ClockΔ19 mice exhibit significantly augmented responses to the sedative effects of ethanol and ketamine, but not pentobarbital. However, their drinking behavior was not affected by acamprosate, an FDA-approved drug for the treatment of alcoholism, suggesting that their increased glutamatergic tone might underlie the increased sensitivity to the sedative/hypnotic properties of ethanol but not the rewarding properties of ethanol. Taken together, we have identified a significant role for Clock in the VTA as a negative regulator of ethanol intake and implicate the VTA dopamine system in this response.
Collapse
|
29
|
Forsyth CB, Voigt RM, Shaikh M, Tang Y, Cederbaum AI, Turek FW, Keshavarzian A. Role for intestinal CYP2E1 in alcohol-induced circadian gene-mediated intestinal hyperpermeability. Am J Physiol Gastrointest Liver Physiol 2013; 305:G185-95. [PMID: 23660503 PMCID: PMC3725682 DOI: 10.1152/ajpgi.00354.2012] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have shown that alcohol increases Caco-2 intestinal epithelial cell monolayer permeability in vitro by inducing the expression of redox-sensitive circadian clock proteins CLOCK and PER2 and that these proteins are necessary for alcohol-induced hyperpermeability. We hypothesized that alcohol metabolism by intestinal Cytochrome P450 isoform 2E1 (CYP2E1) could alter circadian gene expression (Clock and Per2), resulting in alcohol-induced hyperpermeability. In vitro Caco-2 intestinal epithelial cells were exposed to alcohol, and CYP2E1 protein, activity, and mRNA were measured. CYP2E1 expression was knocked down via siRNA and alcohol-induced hyperpermeability, and CLOCK and PER2 protein expression were measured. Caco-2 cells were also treated with alcohol or H₂O₂ with or without N-acetylcysteine (NAC) anti-oxidant, and CLOCK and PER2 proteins were measured at 4 or 2 h. In vivo Cyp2e1 protein and mRNA were also measured in colon tissue from alcohol-fed mice. Alcohol increased CYP2E1 protein by 93% and enzyme activity by 69% in intestinal cells in vitro. Alcohol feeding also increased mouse colonic Cyp2e1 protein by 73%. mRNA levels of Cyp2e1 were not changed by alcohol in vitro or in mouse intestine. siRNA knockdown of CYP2E1 in Caco-2 cells prevented alcohol-induced hyperpermeability and induction of CLOCK and PER2 proteins. Alcohol-induced and H₂O₂-induced increases in intestinal cell CLOCK and PER2 were significantly inhibited by treatment with NAC. We concluded that our data support a novel role for intestinal CYP2E1 in alcohol-induced intestinal hyperpermeability via a mechanism involving CYP2E1-dependent induction of oxidative stress and upregulation of circadian clock proteins CLOCK and PER2.
Collapse
Affiliation(s)
- Christopher B. Forsyth
- Departments of 1Internal Medicine, Division of Digestive Diseases and Nutrition, ,2Biochemistry,
| | - Robin M. Voigt
- Departments of 1Internal Medicine, Division of Digestive Diseases and Nutrition,
| | - Maliha Shaikh
- Departments of 1Internal Medicine, Division of Digestive Diseases and Nutrition,
| | - Yueming Tang
- Departments of 1Internal Medicine, Division of Digestive Diseases and Nutrition,
| | - Arthur I. Cederbaum
- 3Mount Sinai School of Medicine, Department of Pharmacology and System Therapeutics, New York, New York;
| | - Fred W. Turek
- 8Northwestern University Feinberg School of Medicine, Chicago; ,4Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, Illinois;
| | - Ali Keshavarzian
- Departments of 1Internal Medicine, Division of Digestive Diseases and Nutrition, ,5Pharmacology, and ,6Molecular Biophysics and Physiology, Rush University Medical Center, Chicago; ,7Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
30
|
McCulley WD, Ascheid S, Crabbe JC, Rosenwasser AM. Selective breeding for ethanol-related traits alters circadian phenotype. Alcohol 2013; 47:187-94. [PMID: 23414725 DOI: 10.1016/j.alcohol.2013.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/10/2013] [Accepted: 01/12/2013] [Indexed: 10/27/2022]
Abstract
Previous studies in mice and rats have shown that selective breeding for high and low ethanol preference results in divergence of circadian phenotype in the selected lines. These results indicate that some alleles influencing ethanol preference also contribute to circadian rhythm regulation. Selective breeding has also been used to produce lines of mice differing in a number of other ethanol-related traits, while studies of phenotypic and genetic correlation indicate that diverse ethanol-related traits are influenced by both shared and unshared genetics. In the present study, we examined several features of circadian activity rhythms in a mouse line selected for binge-like drinking and in mouse lines selected for high and low severity of ethanol withdrawal convulsions. Specifically, Experiment 1 compared High Drinking in the Dark (HDID-1) mice to their genetically heterogeneous progenitor line (HS/Npt), and Experiment 2 compared Withdrawal Seizure-Prone (WSP-2) and Withdrawal Seizure-Resistant (WSR-2) mice. Both line pairs displayed differences in their daily activity patterns under light-dark conditions. In addition, HDID-1 mice showed shorter free-running periods in constant light and less coherent activity rhythms across lighting conditions relative to HS/Npt controls, while WSP-2 mice showed longer free-running periods in constant darkness relative to WSR-2 mice. These results strengthen the evidence for genetic linkages between responsiveness to ethanol and circadian regulation, and extend this evidence to include ethanol-related phenotypes other than preference drinking. However, the present results also indicate that the nature of genetic correlations between and within phenotypic domains is highly complex.
Collapse
|
31
|
Hasler BP, Clark DB. Circadian misalignment, reward-related brain function, and adolescent alcohol involvement. Alcohol Clin Exp Res 2013; 37:558-65. [PMID: 23360461 DOI: 10.1111/acer.12003] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 08/14/2012] [Indexed: 11/26/2022]
Abstract
BACKGROUND Developmental changes in sleep and circadian rhythms that occur during adolescence may contribute to reward-related brain dysfunction, and consequently increase the risk of alcohol use disorders (AUDs). METHODS This review (i) describes marked changes in circadian rhythms, reward-related behavior and brain function, and alcohol involvement that occur during adolescence, (ii) offers evidence that these parallel developmental changes are associated, and (iii) posits a conceptual model by which misalignment between sleep-wake timing and endogenous circadian timing may increase the risk of adolescent AUDs by altering reward-related brain function. RESULTS The timing of sleep shifts later throughout adolescence, in part due to developmental changes in endogenous circadian rhythms, which tend to become more delayed. This tendency for delayed sleep and circadian rhythms is at odds with early school start times during secondary education, leading to misalignment between many adolescents' sleep-wake schedules and their internal circadian timing. Circadian misalignment is associated with increased alcohol use and other risk-taking behaviors, as well as sleep loss and sleep disturbance. Growing evidence indicates that circadian rhythms modulate the reward system, suggesting that circadian misalignment may impact adolescent alcohol involvement by altering reward-related brain function. Neurocognitive function is also subject to sleep and circadian influence, and thus circadian misalignment may also impair inhibitory control and other cognitive processes relevant to alcohol use. Specifically, circadian misalignment may further exacerbate the cortical-subcortical imbalance within the reward circuit, an imbalance thought to explain increased risk-taking and sensation-seeking during adolescence. Adolescent alcohol use is highly contextualized, however, and thus studies testing this model will also need to consider factors that may influence both circadian misalignment and alcohol use. CONCLUSIONS This review highlights growing evidence supporting a path by which circadian misalignment may disrupt reward mechanisms, which may in turn accelerate the transition from alcohol use to AUDs in vulnerable adolescents.
Collapse
Affiliation(s)
- Brant P Hasler
- Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O’Hara Street, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
32
|
Rosenwasser AM, Fixaris MC. Chronobiology of alcohol: studies in C57BL/6J and DBA/2J inbred mice. Physiol Behav 2013; 110-111:140-7. [PMID: 23313401 DOI: 10.1016/j.physbeh.2013.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 11/02/2012] [Accepted: 01/06/2013] [Indexed: 12/18/2022]
Abstract
Human alcoholics display dramatic disruptions of circadian rhythms that may contribute to the maintenance of excessive drinking, thus creating a vicious cycle. While clinical studies cannot establish direct causal mechanisms, recent animal experiments have revealed bidirectional interactions between circadian rhythms and ethanol intake, suggesting that the chronobiological disruptions seen in human alcoholics are mediated in part by alterations in circadian pacemaker function. The present study was designed to further explore these interactions using C57BL/6J (B6) and DBA/2J (D2) inbred mice, two widely employed strains differing in both circadian and alcohol-related phenotypes. Mice were maintained in running-wheel cages with or without free-choice access to ethanol and exposed to a variety of lighting regimens, including standard light-dark cycles, constant darkness, constant light, and a "shift-lag" schedule consisting of repeated light-dark phase shifts. Relative to the standard light-dark cycle, B6 mice showed reduced ethanol intake in both constant darkness and constant light, while D2 mice showed reduced ethanol intake only in constant darkness. In contrast, shift-lag lighting failed to affect ethanol intake in either strain. Access to ethanol altered daily activity patterns in both B6 and D2 mice, and increased activity levels in D2 mice, but had no effects on other circadian parameters. Thus, the overall pattern of results was broadly similar in both strains, and consistent with previous observations that chronic ethanol intake alters circadian activity patterns while environmental perturbation of circadian rhythms modulates voluntary ethanol intake. These results suggest that circadian-based interventions may prove useful in the management of alcohol use disorders.
Collapse
|
33
|
Brager AJ, Hammer SB. Impact of wheel running on chronic ethanol intake in aged Syrian hamsters. Physiol Behav 2012; 107:418-23. [PMID: 23022151 DOI: 10.1016/j.physbeh.2012.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 06/14/2012] [Accepted: 09/18/2012] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Alcohol dependence in aging populations is seen as a public health concern, most recently because of the significant proportion of heavy drinking among "Baby Boomers." Basic animal research on the effects of aging on physiological and behavioral regulation of ethanol (EtOH) intake is sparse, since most of this research is limited to younger models of alcoholism. Here, EtOH drinking and preference were measured in groups of aged Syrian hamsters. Further, because voluntary exercise (wheel-running) is a rewarding substitute for EtOH in young adult hamsters, the potential for such reward substitution was also assessed. METHODS Aged (24 month-old) male hamsters were subjected to a three-stage regimen of free-choice EtOH (20% v/v) or water and unlocked or locked running wheels to investigate the modulatory effects of voluntary wheel running on EtOH intake and preference. Levels of fluid intake and activity were recorded daily across 60 days of experimentation. RESULTS Prior to wheel running, levels of EtOH intake were significantly less than levels of water intake, resulting in a low preference for EtOH (30%). Hamsters with access to an unlocked running wheel had decreased EtOH intake and preference compared with hamsters with access to a locked running wheel. These group differences in EtOH intake and preference were sustained for up to 10 days after running wheels were re-locked. DISCUSSION These results extend upon those of our previous work in young adult hamsters, indicating that aging dampens EtOH intake and preference. Voluntary wheel running further limited EtOH intake, suggesting that exercise could offer a practical approach for managing late-life alcoholism.
Collapse
Affiliation(s)
- Allison J Brager
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| | | |
Collapse
|
34
|
Perreau-Lenz S, Vengeliene V, Noori HR, Merlo-Pich EV, Corsi MA, Corti C, Spanagel R. Inhibition of the casein-kinase-1-ε/δ/ prevents relapse-like alcohol drinking. Neuropsychopharmacology 2012; 37:2121-31. [PMID: 22549116 PMCID: PMC3398717 DOI: 10.1038/npp.2012.62] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
During the past decade, it has been shown that circadian clock genes have more than a simple circadian time-keeping role. Clock genes also modulate motivational processes and have been implicated in the development of psychiatric disorders such as drug addiction. Recent studies indicate that casein-kinase 1ε/δ (CK1ε/δ)--one of the components of the circadian molecular clockwork-might be involved in the etiology of addictive behavior. The present study was initiated to study the specific role of CK1ε/δ in alcohol relapse-like drinking using the 'Alcohol Deprivation Effect' model. The effect of CK1ε/δ inhibition was tested on alcohol consumption in long-term alcohol-drinking rats upon re-exposure to alcohol after deprivation using a four-bottle free-choice paradigm with water, 5%, 10%, and 20% ethanol solutions, as well as on saccharin preference in alcohol-naive rats. The inhibition of CK1ε/δ with systemic PF-670462 (0, 10, and 30 mg/kg) injections dose-dependently decreased, and at a higher dosage prevented the alcohol deprivation effect, as compared with vehicle-treated rats. The impact of the treatment was further characterized using nonlinear regression analyses on the daily profiles of drinking and locomotor activity. We reveal that CK1ε/δ inhibition blunted the high daytime alcohol intake typically observed upon alcohol re-exposure, and induced a phase shift of locomotor activity toward daytime. Only the highest dose of PF-670462 shifted the saccharin intake daily rhythm toward daytime during treatment, and decreased saccharin preference after treatment. Our data suggest that CK1 inhibitors may be candidates for drug treatment development for alcoholism.
Collapse
Affiliation(s)
- Stéphanie Perreau-Lenz
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany.
| | - Valentina Vengeliene
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Hamid R Noori
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | | | | | - Corrado Corti
- Aptuit, Medicine Research Centre, Verona, Italy,Aptuit, Medicine Research Centre, via Alessandro Fleming 4, 37129 Verona, Italy, Tel: +39 045 8219 576, Fax: +39 045 8218 047, E-mail:
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| |
Collapse
|
35
|
Seggio JA, Possidente B, Ahmad ST. Larval ethanol exposure alters adult circadian free-running locomotor activity rhythm in Drosophila melanogaster. Chronobiol Int 2012; 29:75-81. [PMID: 22217104 DOI: 10.3109/07420528.2011.635236] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Alcohol consumption causes disruptions in a variety of daily rhythms, including the sleep-wake cycle. Few studies have explored the effect of alcohol exposure only during developmental stages preceding maturation of the adult circadian clock, and none have examined the effects of alcohol on clock function in Drosophila. This study investigates developmental and behavioral correlates between larval ethanol exposure and the adult circadian clock in Drosophila melanogaster, a well-established model for studying circadian rhythms and effects of ethanol exposure. We reared Drosophila larvae on 0%, 10%, or 20% ethanol-supplemented food and assessed effects upon eclosion and the free-running period of the circadian rhythm of locomotor activity. We observed a dose-dependent effect of ethanol on period, with higher doses resulting in shorter periods. We also identified the third larval instar stage as a critical time for the developmental effects of 10% ethanol on circadian period. These results demonstrate that developmental ethanol exposure causes sustainable shortening of the adult free-running period in Drosophila melanogaster, even after adult exposure to ethanol is terminated, and suggests that the third instar is a sensitive time for this effect.
Collapse
Affiliation(s)
- Joseph A Seggio
- Department of Biology, Bridgewater State University, Bridgewater, Massachusetts 02325, USA.
| | | | | |
Collapse
|
36
|
Kubo Y, Tahara Y, Hirao A, Shibata S. 2,2,2-Tribromoethanol phase-shifts the circadian rhythm of the liver clock in Per2::Luciferase knockin mice: lack of dependence on anesthetic activity. J Pharmacol Exp Ther 2012; 340:698-705. [PMID: 22171092 DOI: 10.1124/jpet.111.188615] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Comprehensive gene expression profiling in mice in response to the inhalation of sevoflurane has revealed that circadian clock gene expression is affected strongly in the liver, heart, lung, and kidney, in this order, but moderately in the spleen and slightly in the brain. Therefore, we examined whether the administration of general anesthetics at different times of the day induces phase shifts of the liver clock in Per2::Luciferase knockin mice. One to 4 days of intraperitoneal injection of 2,2,2-tribromoethanol (240 mg/kg, anesthetic time 60 min) or 2,2,2-trichloroethanol (240 mg/kg, 60 min), common anesthetics in veterinary surgery, caused phase delays when injected during the daytime and phase advances when injected during the nighttime. Inhalation administration of isoflurane for 30 or 60 min during the daytime did not induce a phase delay. Injection of propofol (300 mg/kg, 17 min) during the daytime induced an insignificant phase delay of the Per2 bioluminescence rhythm. Injection of 2,2,2-tribromoethanol did not induce a phase shift in the suprachiasmatic nucleus, the main oscillator, or in behavioral locomotor rhythms, suggesting that 2,2,2-tribromoethanol induced phase shifts of the liver clock independent of the main suprachiasmatic clock. The expression of clock genes, such as Bmal1 and Clock, in mouse liver was decreased strongly 1 and 4 h after a single injection of 2,2,2-tribromoethanol. These results demonstrate that 2,2,2-tribromoethanol or 2,2,2-trichloroethanol produce phase shifts of the peripheral clock, independent of anesthetic activity. These anesthetics may cause circadian rhythm disorders in peripheral organs when administered as general anesthetics several times during the day.
Collapse
Affiliation(s)
- Yuji Kubo
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | | | | | | |
Collapse
|
37
|
Conroy DA, Hairston IS, Arnedt JT, Hoffmann RF, Armitage R, Brower KJ. Dim light melatonin onset in alcohol-dependent men and women compared with healthy controls. Chronobiol Int 2012; 29:35-42. [PMID: 22217099 PMCID: PMC4258345 DOI: 10.3109/07420528.2011.636852] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sleep disturbances in alcohol-dependent (AD) individuals may persist despite abstinence from alcohol and can influence the course of the disorder. Although the mechanisms of sleep disturbances of AD are not well understood and some evidence suggests dysregulation of circadian rhythms, dim light melatonin onset (DLMO) has not previously been assessed in AD versus healthy control (HC) individuals in a sample that varied by sex and race. The authors assessed 52 AD participants (mean ± SD age: 36.0 ± 11.0 yrs of age, 10 women) who were 3-12 wks since their last drink (abstinence: 57.9 ± 19.3 d) and 19 age- and sex-matched HCs (34.4 ± 10.6 yrs, 5 women). Following a 23:00-06:00 h at-home sleep schedule for at least 5 d and screening/baseline nights in the sleep laboratory, participants underwent a 3-h extension of wakefulness (02:00 h bedtime) during which salivary melatonin samples were collected every 30 min beginning at 19:30 h. The time of DLMO was the primary measure of circadian physiology and was assessed with two commonly used methodologies. There was a slower rate of rise and lower maximal amplitude of the melatonin rhythm in the AD group. DLMO varied by the method used to derive it. Using 3 pg/mL as threshold, no significant differences were found between the AD and HC groups. Using 2 standard deviations above the mean of the first three samples, the DLMO in AD occurred significantly later, 21:02 ± 00:41 h, than in HC, 20:44 ± 00:21 h (t = -2.4, p = .02). Although melatonin in the AD group appears to have a slower rate of rise, using well-established criteria to assess the salivary DLMO did not reveal differences between AD and HC participants. Only when capturing melatonin when it is already rising was DLMO found to be significantly delayed by a mean 18 min in AD participants. Future circadian analyses on alcoholics should account for these methodological caveats.
Collapse
Affiliation(s)
- Deirdre A Conroy
- Addiction Research Center, Department of Psychiatry, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Brager AJ, Prosser RA, Glass JD. Circadian and acamprosate modulation of elevated ethanol drinking in mPer2 clock gene mutant mice. Chronobiol Int 2012; 28:664-72. [PMID: 21929298 DOI: 10.3109/07420528.2011.601968] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The PER2 clock gene modulates ethanol consumption, such that mutant mice not expressing functional mPer2 have altered circadian behavior that promotes higher ethanol intake and preference. Experiments were undertaken to characterize circadian-related behavioral effects of mPer2 deletion on ethanol intake and to explore how acamprosate (used to reduce alcohol dependence) alters diurnal patterns of ethanol intake. Male mPer2 mutant and WT (wild-type) mice were entrained to a 12:12 h light-dark (12L:12D) photocycle, and their locomotor and drinking activities were recorded. Circadian locomotor measurements confirmed that mPer2 mutants had an advanced onset of nocturnal activity of about 2 h relative to WTs, and an increased duration of nocturnal activity (p < .01). Also, mPer2 mutants preferred and consumed more ethanol and had more daily ethanol drinking episodes vs. WTs. Measurements of systemic ethanol using subcutaneous microdialysis confirmed the advanced rise in ethanol intake in the mPer2 mutants, with 24-h averages being ∼60 vs. ∼25 mM for WTs (p < .01). A 6-day regimen of single intraperitoneal (i.p.) acamprosate injections (300 mg/kg) at zeitgeber time (ZT) 10 did not alter the earlier onset of nocturnal ethanol drinking in the mPer2 mutants, but reduced the overall amplitude of drinking and preference (both p < .01). Acamprosate also reduced these parameters in WTs. These results suggest that elevated ethanol intake in mPer2 mutants may be a partial consequence of an earlier nighttime activity onset and increase in nocturnal drinking activity. The suppressive action of acamprosate on ethanol intake is not due to an altered diurnal pattern of drinking, but rather a decrease in the number of daily drinking bouts and amount of drinking per bout.
Collapse
Affiliation(s)
- Allison J Brager
- Department of Biological Sciences, Kent State University, Kent, Ohio 44242, USA
| | | | | |
Collapse
|
39
|
Sarkar DK. Circadian genes, the stress axis, and alcoholism. Alcohol Res 2012; 34:362-6. [PMID: 23134053 PMCID: PMC3860413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The body's internal system to control the daily rhythm of the body's functions (i.e., the circadian system), the body's stress response, and the body's neurobiology are highly interconnected. Thus, the rhythm of the circadian system impacts alcohol use patterns; at the same time, alcohol drinking also can alter circadian functions. The sensitivity of the circadian system to alcohol may result from alcohol's effects on the expression of several of the clock genes that regulate circadian function. The stress response system involves the hypothalamus and pituitary gland in the brain and the adrenal glands, as well as the hormones they secrete, including corticotrophin-releasing hormone, adrenocorticotrophic hormone, and glucocorticoids. It is controlled by brain-signaling molecules, including endogenous opioids such as β-endorphin. Alcohol consumption influences the activity of this system and vice versa. Finally, interactions exist between the circadian system, the hypothalamic-pituitary-adrenal axis, and alcohol consumption. Thus, it seems that certain clock genes may control functions of the stress response system and that these interactions are affected by alcohol.
Collapse
|
40
|
Singh P, Suar D, Leiter MP. Antecedents, Work-Related Consequences, and Buffers of Job Burnout Among Indian Software Developers. JOURNAL OF LEADERSHIP & ORGANIZATIONAL STUDIES 2011. [DOI: 10.1177/1548051811429572] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study examines the antecedents, consequences, and buffers of job burnout among software developers using job demands resources theory. Data were collected from 372 software developers in India using questionnaires. Results reveal that software developers experiencing more role ambiguity, role conflict, schedule pressure, irregular shifts, group noncooperation, psychological contract violation, and work–family conflict are at a greater risk of job burnout. The most important antecedent of job burnout was found to be work–family conflict. Job burnout increased job performance but decreased organizational commitment and interpersonal relationships. Subjective well-being and practicing yoga and meditation were inversely related to burnout-linked job performance. Subjective well-being, social support, and practicing yoga and meditation were also found to decrease the adverse association of job burnout with organizational commitment and interpersonal relationships. In the context of work-related consequences, job burnout had the biggest adverse association with organizational commitment, and practicing yoga and meditation was found to be the most influential buffer to counter the adverse consequences of job burnout.
Collapse
Affiliation(s)
- Pankaj Singh
- T. A. Pai Management Institute, Manipal, Karnataka, India
| | - Damodar Suar
- Indian Institute of Technology, Kharagpur, West Bengal, India
| | | |
Collapse
|
41
|
Brager A, Prosser RA, Glass JD. Acamprosate-responsive brain sites for suppression of ethanol intake and preference. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1032-43. [PMID: 21697518 DOI: 10.1152/ajpregu.00179.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acamprosate suppresses alcohol intake and craving in recovering alcoholics; however, the central sites of its action are unclear. To approach this question, brain regions responsive to acamprosate were mapped using acamprosate microimplants targeted to brain reward and circadian areas implicated in alcohol dependence. mPer2 mutant mice with nonfunctional mPer2, a circadian clock gene that gates endogenous timekeeping, were included, owing to their high levels of ethanol intake and preference. Male wild-type (WT) and mPer2 mutant mice received free-choice (15%) ethanol/water for 3 wk. The ethanol was withdrawn for 3 wk and then reintroduced to facilitate relapse. Four days before ethanol reintroduction, mice received bilateral blank or acamprosate-containing microimplants releasing ∼50 ng/day into reward [ventral tegmental (VTA), peduculopontine tegmentum (PPT), and nucleus accumbens (NA)] and circadian [intergeniculate leaflet (IGL) and suprachiasmatic nucleus (SCN)] areas. The hippocampus was also targeted. Circadian locomotor activity was measured throughout. Ethanol intake and preference were greater in mPer2 mutants than in wild-type (WT) mice (27 g·kg(-1)·day(-1) vs. 13 g·kg(-1)·day(-1) and 70% vs. 50%, respectively; both, P < 0.05). In WTs, acamprosate in all areas, except hippocampus, suppressed ethanol intake and preference (by 40-60%) during ethanol reintroduction. In mPer2 mutants, acamprosate in the VTA, PPT, and SCN suppressed ethanol intake and preference by 20-30%. These data are evidence that acamprosate's suppression of ethanol intake and preference are manifest through actions within major reward and circadian sites.
Collapse
Affiliation(s)
- Allison Brager
- Department of Biological Sciences, Kent State University, Kent, Ohio 44242, USA
| | | | | |
Collapse
|
42
|
Hasler BP, Smith LJ, Cousins JC, Bootzin RR. Circadian rhythms, sleep, and substance abuse. Sleep Med Rev 2011; 16:67-81. [PMID: 21620743 DOI: 10.1016/j.smrv.2011.03.004] [Citation(s) in RCA: 182] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 03/15/2011] [Accepted: 03/17/2011] [Indexed: 12/31/2022]
Abstract
Substance abuse is linked to numerous mental and physical health problems, including disturbed sleep. The association between substance use and sleep appears to be bidirectional, in that substance use may directly cause sleep disturbances, and difficulty sleeping may be a risk factor for relapse to substance use. Growing evidence similarly links substance use to disturbances in circadian rhythms, although many gaps in knowledge persist, particularly regarding whether circadian disturbance leads to substance abuse or dependence. Given the integral role circadian rhythms play in regulating sleep, circadian mechanisms may account in part for sleep-substance abuse interactions. Furthermore, a burgeoning research base supports a role for the circadian system in regulating reward processing, indicating that circadian mechanisms may be directly linked to substance abuse independently of sleep pathways. More work in this area is needed, particularly in elucidating how sleep and circadian disturbance may contribute to initiation of, and/or relapse to, substance use. Sleep and circadian-based interventions could play a critical role in the prevention and treatment of substance use disorders.
Collapse
Affiliation(s)
- Brant P Hasler
- University of Pittsburgh School of Medicine, Department of Psychiatry, 3811 O'Hara Street, Pittsburgh, PA 15213, USA.
| | | | | | | |
Collapse
|
43
|
Brager AJ, Ruby CL, Prosser RA, Glass JD. Acute ethanol disrupts photic and serotonergic circadian clock phase-resetting in the mouse. Alcohol Clin Exp Res 2011; 35:1467-74. [PMID: 21463340 DOI: 10.1111/j.1530-0277.2011.01483.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Alcohol dependence is associated with impaired circadian rhythms and sleep. Ethanol administration disrupts circadian clock phase-resetting, suggesting a mode for the disruptive effect of alcohol dependence on the circadian timing system. In this study, we extend previous work in C57BL/6J mice to: (i) characterize the suprachiasmatic nucleus (SCN) pharmacokinetics of acute systemic ethanol administration, (ii) explore the effects of acute ethanol on photic and nonphotic phase-resetting, and (iii) determine if the SCN is a direct target for photic effects. METHODS First, microdialysis was used to characterize the pharmacokinetics of acute intraperitoneal (i.p.) injections of 3 doses of ethanol (0.5, 1.0, and 2.0 g/kg) in the mouse SCN circadian clock. Second, the effects of acute i.p. ethanol administration on photic phase delays and serotonergic ([+]8-OH-DPAT-induced) phase advances of the circadian activity rhythm were assessed. Third, the effects of reverse-microdialysis ethanol perfusion of the SCN on photic phase-resetting were characterized. RESULTS Peak ethanol levels from the 3 doses of ethanol in the SCN occurred within 20 to 40 minutes postinjection with half-lives for clearance ranging from 0.6 to 1.8 hours. Systemic ethanol treatment dose-dependently attenuated photic and serotonergic phase-resetting. This treatment also did not affect basal SCN neuronal activity as assessed by Fos expression. Intra-SCN perfusion with ethanol markedly reduced photic phase delays. CONCLUSIONS These results confirm that acute ethanol attenuates photic phase-delay shifts and serotonergic phase-advance shifts in the mouse. This dual effect could disrupt photic and nonphotic entrainment mechanisms governing circadian clock timing. It is also significant that the SCN clock is a direct target for disruptive effects of ethanol on photic shifting. Such actions by ethanol could underlie the disruptive effects of alcohol abuse on behavioral, physiological, and endocrine rhythms associated with alcoholism.
Collapse
Affiliation(s)
- Allison J Brager
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | | | | | | |
Collapse
|
44
|
Agapito M, Mian N, Boyadjieva NI, Sarkar DK. Period 2 gene deletion abolishes beta-endorphin neuronal response to ethanol. Alcohol Clin Exp Res 2010; 34:1613-8. [PMID: 20586752 DOI: 10.1111/j.1530-0277.2010.01246.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Ethanol exposure during early life has been shown to permanently alter the circadian expression of clock regulatory genes and the beta-endorphin precursor proopiomelanocortin (POMC) gene in the hypothalamus. Ethanol also alters the stress- and immune-regulatory functions of beta-endorphin neurons in laboratory rodents. Our aim was to determine whether the circadian clock regulatory Per2 gene modulates the action of ethanol on beta-endorphin neurons in mice. METHODS Per2 mutant (mPer2(Brdml)) and wild type (C57BL/6J) mice were used to determine the effect of Per2 mutation on ethanol-regulated beta-endorphin neuronal activity during neonatal period using an in vitro mediobasal hypothalamic (MBH) cell culture model and an in vivo milk formula feeding animal model. The beta-endorphin neuronal activity following acute and chronic ethanol treatments was evaluated by measuring the peptide released from cultured cells or peptide levels in the MBH tissues, using enzyme-linked immunosorbent assay (ELISA). RESULTS Per2 mutant mice showed a higher basal level of beta-endorphin release from cultured MBH cells and a moderate increase in the peptide content in the MBH in comparison with control mice. However, unlike wild type mice, Per2 mutant mice showed no stimulatory or inhibitory beta-endorphin-secretory responses to acute and chronic ethanol challenges in vitro. Furthermore, Per2 mutant mice, but not wild type mice, failed to show the stimulatory and inhibitory responses of MBH beta-endorphin levels to acute and chronic ethanol challenges in vivo. CONCLUSIONS These results suggest for the first time that the Per2 gene may be critically involved in regulating beta-endorphin neuronal function. Furthermore, the data revealed an involvement of the Per2 gene in regulating beta-endorphin neuronal responses to ethanol.
Collapse
Affiliation(s)
- Maria Agapito
- Endocrine Program, Department of Animal Sciences, Graduate Program of Neuroscience, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901, USA
| | | | | | | |
Collapse
|
45
|
Hammer SB, Ruby CL, Brager AJ, Prosser RA, Glass JD. Environmental modulation of alcohol intake in hamsters: effects of wheel running and constant light exposure. Alcohol Clin Exp Res 2010; 34:1651-8. [PMID: 20569242 DOI: 10.1111/j.1530-0277.2010.01251.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Alcohol abuse leads to marked disruptions of circadian rhythms, and these disturbances in themselves can increase the drive to drink. Circadian clock timing is regulated by light, as well as by nonphotic influences such as food, social interactions, and wheel running. We previously reported that alcohol markedly disrupts photic and nonphotic modes of circadian rhythm regulation in Syrian hamsters. As an extension of this work, we characterize the hedonic interrelationship between wheel running and ethanol (EtOH) intake and the effects of environmental circadian disruption (long-term exposure to constant light [LL]) on the drive to drink. METHODS First, we tested the effect of wheel running on chronic free-choice consumption of a 20% (v/v) EtOH solution and water. Second, the effect of this alcohol drinking on wheel running in alcohol-naive animals was investigated. Third, we assessed the influence of LL, known to suppress locomotor activity and cause circadian rhythm disruption, on EtOH consumption and wheel-running behavior. RESULTS Inhibitory effects of wheel running on EtOH intake and vice versa were observed. Exposure to LL, while not affecting EtOH intake, induced rhythm splitting in 75% of the animals. Notably, the splitting phenotype was associated with lower levels of EtOH consumption and preference prior to, and throughout, the period of LL exposure. CONCLUSIONS These results are evidence that exercise may offer an efficacious clinical approach to reducing EtOH intake. Also, predisposition for light-induced (or other) forms of circadian disruption may modulate the drive to drink.
Collapse
Affiliation(s)
- Steven B Hammer
- Department of Biological Sciences, Kent State University, Kent, Ohio 44242, USA
| | | | | | | | | |
Collapse
|
46
|
Brager AJ, Ruby CL, Prosser RA, Glass JD. Chronic ethanol disrupts circadian photic entrainment and daily locomotor activity in the mouse. Alcohol Clin Exp Res 2010; 34:1266-73. [PMID: 20477766 DOI: 10.1111/j.1530-0277.2010.01204.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Chronic ethanol abuse is associated with disrupted circadian rhythms and sleep. Ethanol administration impairs circadian clock phase-resetting, suggesting a mode for the disruptive effect of alcohol abuse on circadian timing. Here, we extend previous studies to explore the effects of chronic forced ethanol on photic phase-resetting, photic entrainment, and daily locomotor activity patterns in C57BL/6J mice. METHODS First, microdialysis was used to characterize the circadian patterns of ethanol uptake in the suprachiasmatic (SCN) circadian clock and correlate this with systemic ethanol levels and episodic drinking of 10 or 15% ethanol. Second, the effects of chronic forced ethanol drinking and withdrawal on photic phase-delays of the circadian activity rhythm were assessed. Third, the effects of chronic ethanol drinking on entrainment to a weak photic zeitgeber (1 minute of 25 lux intensity light per day) were assessed. This method was used to minimize any masking actions of light that could mask ethanol effects on clock entrainment. RESULTS Peak ethanol levels in the SCN and periphery occurred during the dark phase and coincided with the time when light normally induces phase-delays in mice. These delays were dose-dependently inhibited by chronic ethanol and its withdrawal. Chronic ethanol did not impede re-entrainment to a shifted light cycle but affected entrainment under the weak photic zeitgeber and disrupted the daily pattern of locomotor activity. CONCLUSIONS These results confirm that chronic ethanol consumption and withdrawal markedly impair circadian clock photic phase-resetting. Ethanol also disturbs the temporal structure of nighttime locomotor activity and photic entrainment. Collectively, these results suggest a direct action of ethanol on the SCN clock.
Collapse
Affiliation(s)
- Allison J Brager
- Department of Biological Sciences, Kent State University, Ohio, USA
| | | | | | | |
Collapse
|
47
|
Rosenwasser AM, Clark JW, Fixaris MC, Belanger GV, Foster JA. Effects of repeated light-dark phase shifts on voluntary ethanol and water intake in male and female Fischer and Lewis rats. Alcohol 2010; 44:229-37. [PMID: 20488643 DOI: 10.1016/j.alcohol.2010.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 03/08/2010] [Accepted: 03/30/2010] [Indexed: 11/27/2022]
Abstract
Several lines of evidence implicate reciprocal interactions between excessive alcohol (ethanol) intake and dysregulation of circadian biological rhythms. Thus, chronic alcohol intake leads to widespread circadian disruption in both humans and experimental animals, while in turn, chronobiological disruption has been hypothesized to promote or sustain excessive alcohol intake. Nevertheless, the effects of circadian disruption on voluntary ethanol intake have not been investigated extensively, and prior studies have reported both increased and decreased ethanol intake in rats maintained under "shift-lag" lighting regimens mimicking those experienced by shift workers and transmeridian travelers. In the present study, male and female inbred Fischer and Lewis rats were housed in running wheel cages with continuous free-choice access to both water and 10% (vol/vol) ethanol solution and exposed to repeated 6-h phase advances of the daily light-dark (LD) cycle, whereas controls were kept under standard LD 12:12 conditions. Shift-lag lighting reduced overall ethanol and water intake, and reduced ethanol preference in Fischer rats. Although contrary to the hypothesis that circadian disruption would increase voluntary ethanol intake, these results are consistent with our previous report of reduced ethanol intake in selectively bred high-alcohol-drinking (HAD1) rats housed under a similar lighting regimen. We conclude that chronic circadian disruption is a form of chronobiological stressor that, like other stressors, can either increase or decrease ethanol intake, depending on a variety of poorly understood variables.
Collapse
|