1
|
Fu Y, Ye T, Chen M, Lai B, Zheng P. Protocol to study the role of medial entorhinal cortex-basolateral amygdala circuit in context-induced retrieval of morphine withdrawal memory in mice. STAR Protoc 2025; 6:103542. [PMID: 39921862 PMCID: PMC11851280 DOI: 10.1016/j.xpro.2024.103542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/11/2024] [Accepted: 12/09/2024] [Indexed: 02/10/2025] Open
Abstract
The medial entorhinal cortex (MEC) is crucial for context memory, yet its role in context-induced retrieval of morphine withdrawal memory remains to be investigated. Here, we present a protocol to evaluate the importance of projection neurons from the MEC to the basolateral amygdala (BLA) (MEC-BLA neurons) in mice during context-induced retrieval of morphine withdrawal memory using a conditioned place aversion (CPA) model. We describe steps for surgical procedure and behavioral experiments. Then, we detail procedures of immunofluorescence staining and image analysis. For complete details on the use and execution of this protocol, please refer to Fu et al.1.
Collapse
Affiliation(s)
- Yali Fu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Department of Neurology of Zhongshan Hospital, Fudan University, Shanghai 200032, China; Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China
| | - Ting Ye
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Department of Neurology of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ming Chen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Department of Neurology of Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Bin Lai
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Department of Neurology of Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Ping Zheng
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Department of Neurology of Zhongshan Hospital, Fudan University, Shanghai 200032, China; Medical College of China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
2
|
Gulledge M, Carlezon WA, McHugh RK, Kinard EA, Prerau MJ, Chartoff EH. Spontaneous oxycodone withdrawal disrupts sleep, diurnal, and electrophysiological dynamics in rats. PLoS One 2025; 20:e0312794. [PMID: 39823427 PMCID: PMC11741586 DOI: 10.1371/journal.pone.0312794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 10/13/2024] [Indexed: 01/19/2025] Open
Abstract
Opioid dependence is defined by an aversive withdrawal syndrome upon drug cessation that can motivate continued drug-taking, development of opioid use disorder, and precipitate relapse. An understudied but common opioid withdrawal symptom is disrupted sleep, reported as both insomnia and daytime sleepiness. Despite the prevalence and severity of sleep disturbances during opioid withdrawal, there is a gap in our understanding of their interactions. The goal of this study was to establish an in-depth, temporal signature of spontaneous oxycodone withdrawal effects on the diurnal composition of discrete sleep stages and the dynamic spectral properties of the electroencephalogram (EEG) signal in male rats. We continuously recorded EEG and electromyography (EMG) signals for 8 d of spontaneous withdrawal after a 14-d escalating-dose oxycodone regimen (0.5-8.0 mg/kg, 2×d; SC). During withdrawal, there was a profound loss (peaking on days 2-3) and gradual return of diurnal structure in sleep, body temperature, and locomotor activity, as well as decreased sleep and wake bout durations dependent on lights on/off. Withdrawal was associated with significant alterations in the slope of the aperiodic 1/f component of the EEG power spectrum, an established biomarker of arousal level. Early in withdrawal, NREM exhibited an acute flattening and return to baseline of both low (1-4 Hz) and high (15-50 Hz) frequency components of the 1/f spectrum. These findings suggest temporally dependent withdrawal effects on sleep, reflecting the complex way in which the allostatic forces of opioid withdrawal impinge upon sleep and diurnal processes. These foundational data based on continuous tracking of vigilance state, sleep stage composition, and spectral EEG properties provide a detailed construct with which to form and test hypotheses on the mechanisms of opioid-sleep interactions.
Collapse
Affiliation(s)
- Michael Gulledge
- Dept. of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts, United States of America
- Graduate Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, United States of America
| | - William A. Carlezon
- Dept. of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts, United States of America
| | - R. Kathryn McHugh
- Dept. of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts, United States of America
| | - Elizabeth A. Kinard
- Dept. of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts, United States of America
| | - Michael J. Prerau
- Division of Sleep Medicine, Dept. of Medicine, Harvard Medical School, Brigham & Women’s Hospital, Boston, Massachusetts, United States of America
| | - Elena H. Chartoff
- Dept. of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts, United States of America
| |
Collapse
|
3
|
Samanta S, Bagchi D, Gold MS, Badgaiyan RD, Barh D, Blum K. A Complex Relationship Among the Circadian Rhythm, Reward Circuit and Substance Use Disorder (SUD). Psychol Res Behav Manag 2024; 17:3485-3501. [PMID: 39411118 PMCID: PMC11479634 DOI: 10.2147/prbm.s473310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
The human brain not only controls the various physiological functions but is also the prime regulator of circadian rhythms, rewards, and behaviors. Environmental factors, professional stress, and social disintegration are regarded as the initial causative factors of addiction behavior. Shift work, artificial light exposure at night, and chronic and acute jet lag influence circadian rhythm dysfunction. The result is impaired neurotransmitter release, dysfunction of neural circuits, endocrine disturbance, and metabolic disorder, leading to advancement in substance use disorder. There is a bidirectional relationship between chronodisruption and addiction behavior. Circadian rhythm dysfunction, neuroadaptation in the reward circuits, and alteration in clock gene expression in the mesolimbic areas influence substance use disorder (SUD), and chronotherapy has potential benefits in the treatment strategies. This review explores the relationship among the circadian rhythm dysfunction, reward circuit, and SUD. The impact of chronotherapy on SUD has also been discussed.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore, West Bengal, 721101, India
| | - Debasis Bagchi
- Department of Biology, College of Arts and Sciences, Adelphi University, Garden City, NY, USA and Department of Psychology, Gordon F. Derner School of Psychology, Adelphi University, Garden City, NY, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Southern University, Houston, TX, 77004, USA
| | - Mark S Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Rajendra D Badgaiyan
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Debmalya Barh
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, BeloHorizonte, 31270-901, Brazil
- Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, 721172, West Bengal, India
| | - Kenneth Blum
- Division of Addiction Research & Education, Center for Sports, Exercise, and Mental Health, Western University Health Sciences, Pomona, CA, 91766, USA
- Institute of Psychology, Eotvos Loránd University, Budapest, 1053, Hungary
- Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Center, Dayton, OH, 45435, USA
- Department of Psychiatry, University of Vermont, Burlington, VT, 05405, USA
- Division of Nutrigenomics, The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX, 78701, USA
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| |
Collapse
|
4
|
Peng F, Lu J, Su K, Liu X, Luo H, He B, Wang C, Zhang X, An F, Lv D, Luo Y, Su Q, Jiang T, Deng Z, He B, Xu L, Guo T, Xiang J, Gu C, Wang L, Xu G, Xu Y, Li M, Kelley KW, Cui B, Liu Q. Oncogenic fatty acid oxidation senses circadian disruption in sleep-deficiency-enhanced tumorigenesis. Cell Metab 2024; 36:1598-1618.e11. [PMID: 38772364 DOI: 10.1016/j.cmet.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Circadian disruption predicts poor cancer prognosis, yet how circadian disruption is sensed in sleep-deficiency (SD)-enhanced tumorigenesis remains obscure. Here, we show fatty acid oxidation (FAO) as a circadian sensor relaying from clock disruption to oncogenic metabolic signal in SD-enhanced lung tumorigenesis. Both unbiased transcriptomic and metabolomic analyses reveal that FAO senses SD-induced circadian disruption, as illustrated by continuously increased palmitoyl-coenzyme A (PA-CoA) catalyzed by long-chain fatty acyl-CoA synthetase 1 (ACSL1). Mechanistically, SD-dysregulated CLOCK hypertransactivates ACSL1 to produce PA-CoA, which facilitates CLOCK-Cys194 S-palmitoylation in a ZDHHC5-dependent manner. This positive transcription-palmitoylation feedback loop prevents ubiquitin-proteasomal degradation of CLOCK, causing FAO-sensed circadian disruption to maintain SD-enhanced cancer stemness. Intriguingly, timed β-endorphin resets rhythmic Clock and Acsl1 expression to alleviate SD-enhanced tumorigenesis. Sleep quality and serum β-endorphin are negatively associated with both cancer development and CLOCK/ACSL1 expression in patients with cancer, suggesting dawn-supplemented β-endorphin as a potential chronotherapeutic strategy for SD-related cancer.
Collapse
Affiliation(s)
- Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Jinxin Lu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Keyu Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xinyu Liu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Huandong Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Bin He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Fan An
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Dekang Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Yuanyuan Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Qitong Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Tonghui Jiang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Ziqian Deng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Bin He
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lingzhi Xu
- Department of Oncology, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jin Xiang
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ling Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Guowang Xu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Ying Xu
- Cambridge-Soochow University Genomic Resource Center, Soochow University, Suzhou, Jiangsu, China
| | - Mindian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Keith W Kelley
- Department of Pathology, College of Medicine and Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Du K, Shi Q, Zhou X, Zhang L, Su H, Zhang C, Wei Z, Liu T, Wang L, Wang X, Cong B, Yun K. Melatonin attenuates fentanyl - induced behavioral sensitization and circadian rhythm disorders in mice. Physiol Behav 2024; 279:114523. [PMID: 38492912 DOI: 10.1016/j.physbeh.2024.114523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/01/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Melatonin is a neurohormone synthesized by the pineal gland to regulate the circadian rhythms and has proven to be effective in treating drug addiction and dependence. However, the effects of melatonin to modulate the drug-seeking behavior of fentanyl and its underlying molecular mechanism is elusive. This study was designed to investigate the effects of melatonin on fentanyl - induced behavioral sensitization and circadian rhythm disorders in mice. The accompanying changes in the expression of Brain and Muscle Arnt-Like (BMAL1), tyrosine hydroxylase (TH), and monoamine oxidase A (MAO-A) in relevant brain regions including the suprachiasmatic nucleus (SCN), nucleus accumbens (NAc), prefrontal cortex (PFC), and hippocampus (Hip) were investigated by western blot assays to dissect the mechanism by which melatonin modulates fentanyl - induced behavioral sensitization and circadian rhythm disorders. The present study suggest that fentanyl (0.05, 0.1 and 0.2 mg/kg) could induce behavioral sensitization and melatonin (30.0 mg/kg) could attenuate the behavioral sensitization and circadian rhythm disorders in mice. Fentanyl treatment reduced the expression of BMAL1 and MAO-A and increased that of TH in relevant brain regions. Furthermore, melatonin treatment could reverse the expression levels of BMAL1, MAO-A, and TH. In conclusion, our study demonstrate for the first time that melatonin has therapeutic potential for fentanyl addiction.
Collapse
Affiliation(s)
- Kaili Du
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China; School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China; School of Basic Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Qianwen Shi
- School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China; Shanxi Key Laboratory of Forensic Medicine, Shanxi, 030600, China
| | - Xiuya Zhou
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China; School of Basic Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Lifei Zhang
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China; School of Basic Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Hongliang Su
- School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China; Shanxi Key Laboratory of Forensic Medicine, Shanxi, 030600, China
| | - Chao Zhang
- School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China; Shanxi Key Laboratory of Forensic Medicine, Shanxi, 030600, China
| | - Zhiwen Wei
- School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China; Shanxi Key Laboratory of Forensic Medicine, Shanxi, 030600, China
| | - Ting Liu
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China; School of Basic Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China; School of Basic Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaohui Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, 030001, China; School of Basic Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Bin Cong
- School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China; Shanxi Key Laboratory of Forensic Medicine, Shanxi, 030600, China; School of Forensic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Keming Yun
- School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China; Shanxi Key Laboratory of Forensic Medicine, Shanxi, 030600, China.
| |
Collapse
|
6
|
Ketchesin KD, Becker-Krail DD, Xue X, Wilson RS, Lam TT, Williams KR, Nairn AC, Tseng GC, Logan RW. Differential Effects of Cocaine and Morphine on the Diurnal Regulation of the Mouse Nucleus Accumbens Proteome. J Proteome Res 2023. [PMID: 37311105 PMCID: PMC10392613 DOI: 10.1021/acs.jproteome.3c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Substance use disorders are associated with disruptions in sleep and circadian rhythms that persist during abstinence and may contribute to relapse risk. Repeated use of substances such as psychostimulants and opioids may lead to significant alterations in molecular rhythms in the nucleus accumbens (NAc), a brain region central to reward and motivation. Previous studies have identified rhythm alterations in the transcriptome of the NAc and other brain regions following the administration of psychostimulants or opioids. However, little is known about the impact of substance use on the diurnal rhythms of the proteome in the NAc. We used liquid chromatography coupled to tandem mass spectrometry-based quantitative proteomics, along with a data-independent acquisition analysis pipeline, to investigate the effects of cocaine or morphine administration on diurnal rhythms of proteome in the mouse NAc. Overall, our data reveal cocaine and morphine differentially alter diurnal rhythms of the proteome in the NAc, with largely independent differentially expressed proteins dependent on time-of-day. Pathways enriched from cocaine altered protein rhythms were primarily associated with glucocorticoid signaling and metabolism, whereas morphine was associated with neuroinflammation. Collectively, these findings are the first to characterize the diurnal regulation of the NAc proteome and demonstrate a novel relationship between the phase-dependent regulation of protein expression and the differential effects of cocaine and morphine on the NAc proteome. The proteomics data in this study are available via ProteomeXchange with identifier PXD042043.
Collapse
Affiliation(s)
- Kyle D Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Darius D Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Rashaun S Wilson
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- W.M. Keck Biotechnology Mass Spectrometry (MS) & Proteomics Resource Laboratory, Yale University School of Medicine, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - TuKiet T Lam
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- W.M. Keck Biotechnology Mass Spectrometry (MS) & Proteomics Resource Laboratory, Yale University School of Medicine, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Kenneth R Williams
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Angus C Nairn
- Yale/NIDA Neuroproteomics Center, 300 George Street, New Haven, Connecticut 06511, United States
- Department of Psychiatry, Yale University School of Medicine, Connecticut Mental Health Center, New Haven, Connecticut 06511, United States
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01655, United States
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
7
|
Ketchesin KD, Becker-Krail DD, Xue X, Wilson RS, Lam TT, Williams KR, Nairn AC, Tseng GC, Logan RW. Differential Effects of Cocaine and Morphine on the Diurnal Regulation of the Mouse Nucleus Accumbens Proteome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530696. [PMID: 36909659 PMCID: PMC10002738 DOI: 10.1101/2023.03.01.530696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Substance use disorders (SUDs) are associated with disruptions in sleep and circadian rhythms that persist during abstinence and may contribute to relapse risk. Repeated use of substances such as psychostimulants and opioids may lead to significant alterations in molecular rhythms in the nucleus accumbens (NAc), a brain region central to reward and motivation. Previous studies have identified rhythm alterations in the transcriptome of the NAc and other brain regions following the administration of psychostimulants or opioids. However, little is known about the impact of substance use on the diurnal rhythms of the proteome in the NAc. We used liquid chromatography coupled to tandem mass spectrometry-based (LC-MS/MS) quantitative proteomics, along with a data-independent acquisition (DIA) analysis pipeline, to investigate the effects of cocaine or morphine administration on diurnal rhythms of proteome in the mouse NAc. Overall, our data reveals cocaine and morphine differentially alters diurnal rhythms of the proteome in the NAc, with largely independent differentially expressed proteins dependent on time-of-day. Pathways enriched from cocaine altered protein rhythms were primarily associated with glucocorticoid signaling and metabolism, whereas morphine was associated with neuroinflammation. Collectively, these findings are the first to characterize the diurnal regulation of the NAc proteome and demonstrate a novel relationship between phase-dependent regulation of protein expression and the differential effects of cocaine and morphine on the NAc proteome.
Collapse
|
8
|
Xue X, Zong W, Glausier JR, Kim SM, Shelton MA, Phan BN, Srinivasan C, Pfenning AR, Tseng GC, Lewis DA, Seney ML, Logan RW. Molecular rhythm alterations in prefrontal cortex and nucleus accumbens associated with opioid use disorder. Transl Psychiatry 2022; 12:123. [PMID: 35347109 PMCID: PMC8960783 DOI: 10.1038/s41398-022-01894-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 11/21/2022] Open
Abstract
Severe and persistent disruptions to sleep and circadian rhythms are common in people with opioid use disorder (OUD). Preclinical evidence suggests altered molecular rhythms in the brain modulate opioid reward and relapse. However, whether molecular rhythms are disrupted in the brains of people with OUD remained an open question, critical to understanding the role of circadian rhythms in opioid addiction. Using subjects' times of death as a marker of time of day, we investigated transcriptional rhythms in the brains of subjects with OUD compared to unaffected comparison subjects. We discovered rhythmic transcripts in both the dorsolateral prefrontal cortex (DLPFC) and nucleus accumbens (NAc), key brain areas involved in OUD, that were largely distinct between OUD and unaffected subjects. Fewer rhythmic transcripts were identified in DLPFC of subjects with OUD compared to unaffected subjects, whereas in the NAc, nearly double the number of rhythmic transcripts was identified in subjects with OUD. In NAc of subjects with OUD, rhythmic transcripts peaked either in the evening or near sunrise, and were associated with an opioid, dopamine, and GABAergic neurotransmission. Associations with altered neurotransmission in NAc were further supported by co-expression network analysis which identified OUD-specific modules enriched for transcripts involved in dopamine, GABA, and glutamatergic synaptic functions. Additionally, rhythmic transcripts in DLPFC and NAc of subjects with OUD were enriched for genomic loci associated with sleep-related GWAS traits, including sleep duration and insomnia. Collectively, our findings connect transcriptional rhythm changes in opioidergic, dopaminergic, GABAergic signaling in the human brain to sleep-related traits in opioid addiction.
Collapse
Affiliation(s)
- Xiangning Xue
- grid.21925.3d0000 0004 1936 9000Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - Wei Zong
- grid.21925.3d0000 0004 1936 9000Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - Jill R. Glausier
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA
| | - Sam-Moon Kim
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA ,grid.21925.3d0000 0004 1936 9000Center for Adolescent Reward, Rhythms, and Sleep, University of Pittsburgh, Pittsburgh, PA 15219 USA
| | - Micah A. Shelton
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA
| | - BaDoi N. Phan
- grid.147455.60000 0001 2097 0344Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213 USA
| | - Chaitanya Srinivasan
- grid.147455.60000 0001 2097 0344Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213 USA
| | - Andreas R. Pfenning
- grid.147455.60000 0001 2097 0344Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213 USA ,grid.147455.60000 0001 2097 0344Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213 USA
| | - George C. Tseng
- grid.21925.3d0000 0004 1936 9000Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - David A. Lewis
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA
| | - Marianne L. Seney
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA ,grid.21925.3d0000 0004 1936 9000Center for Adolescent Reward, Rhythms, and Sleep, University of Pittsburgh, Pittsburgh, PA 15219 USA
| | - Ryan W. Logan
- grid.189504.10000 0004 1936 7558Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118 USA ,grid.189504.10000 0004 1936 7558Center for Systems Neuroscience, Boston University, Boston, MA 02118 USA
| |
Collapse
|
9
|
Guo L, Hu A, Zhao X, Xiang X. Reduction of Orexin-A Is Associated With Anxiety and the Level of Depression of Male Methamphetamine Users During the Initial Withdrawal Period. Front Psychiatry 2022; 13:900135. [PMID: 35859609 PMCID: PMC9289462 DOI: 10.3389/fpsyt.2022.900135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/10/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Orexin has been linked to the regulation of reward and motivation in animals, but there have been few human studies to validate its regulatory effects. We aimed to determine how orexin-A levels changed during different stages of withdrawal, as well as the relationship between orexin-A levels and withdrawal symptoms in male METH users. METHODS This study included 76 METH users and 35 control participants. The METH users were divided into three groups: group 1 (abstinence within 1 week, n = 23), group 2 (abstinence between 1 week and 3 months, n = 38), and group 3 (abstinence over 3 months, n = 15). At baseline and 1 month of enrollment, the plasma orexin-A level was examined. To track the withdrawal symptoms, self-report questionnaires (anxiety, depression, craving, and sleep quality) were collected at two points. RESULTS The orexin-A levels of groups 1 (p < 0.001) and 2 (p < 0.001) were lower than that of the controls at baseline but not group 3. One month later, the orexin-A levels of group 2 (p < 0.05) significantly increased, while no significant changes in those of groups 1 and 3 were observed. Furthermore, the orexin-A levels of group 1 were positively linked with depression (p < 0.01) and anxiety (p < 0.01) at baseline. CONCLUSIONS The decrease in orexin-A levels was only transitory during the initial abstinence phase, and it was eventually restored near to normal with continued abstinence among the male METH users. Furthermore, a lower concentration of orexin-A may serve as a risk factor for negative emotions during METH withdrawal.
Collapse
Affiliation(s)
- Lei Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Aqian Hu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoxi Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaojun Xiang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
10
|
Gipson CD, Dunn KE, Bull A, Ulangkaya H, Hossain A. Establishing preclinical withdrawal syndrome symptomatology following heroin self-administration in male and female rats. Exp Clin Psychopharmacol 2021; 29:636-649. [PMID: 32297787 PMCID: PMC8405057 DOI: 10.1037/pha0000375] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Opioid use disorder (OUD) is a significant health problem, and understanding mechanisms of various aspects of OUD including drug use and withdrawal is important. Preclinical models provide an ideal opportunity to evaluate mechanisms underlying opioid withdrawal. Current models are limited by their reliance upon forced opioid administration, focus on the acute (and not protracted) syndrome, and exclusion of females. In this study, male and female rats self-administered heroin (maintenance dose of 12.5 μg/kg/infusion) and opioid withdrawal after abrupt discontinuation was measured. In Phase 1, acute withdrawal symptoms were rated in male and female rats at 0, 16, 48, and 72 hr after the last self-administration session. Total somatic signs increased until 48 hr (predominantly in females), and heroin intake positively correlated with total somatic signs at the 48 and 72 hr timepoints. Measures of hyperactivity and anxiety-like behavior increased by 16 and 48 hr, respectively. In Phase 2, symptoms were assessed at baseline, acute, and protracted (168 and 312 hr after self-administration) timepoints in a subset of male and female rats from Phase 1. The total number of somatic signs did not differ across timepoints, though females displayed significantly higher body temperature at all timepoints compared with males, indicating sex-specific protracted withdrawal symptomatology. These data provide a thorough characterization of rodent opioid withdrawal symptomatology after self-administration and abrupt discontinuation that serve as a foundation for future studies designed to mimic the human experience, and demonstrate the importance of characterizing acute and protracted withdrawal with sex-specificity in preclinical models of opioid self-administration. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
- Cassandra D. Gipson
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY
| | - Kelly E. Dunn
- Behavioral Pharmacology Research Unit, Johns Hopkins University, Baltimore, MD
| | - Amanda Bull
- Department of Psychology, Arizona State University, Tempe, AZ
| | - Hanaa Ulangkaya
- Department of Psychology, Arizona State University, Tempe, AZ
| | - Aronee Hossain
- Department of Psychology, Arizona State University, Tempe, AZ
| |
Collapse
|
11
|
Circadian rhythm influences naloxone induced morphine withdrawal and neuronal activity of lateral paragigantocellularis nucleus. Behav Brain Res 2021; 414:113450. [PMID: 34265318 DOI: 10.1016/j.bbr.2021.113450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/10/2021] [Accepted: 07/08/2021] [Indexed: 11/23/2022]
Abstract
Investigations have shown that the circadian rhythm can affect the mechanisms associated with drug dependence. In this regard, we sought to assess the negative consequence of morphine withdrawal syndrome on conditioned place aversion (CPA) and lateral paragigantocellularis (LPGi) neuronal activity in morphine-dependent rats during light (8:00-12:00) and dark (20:00-24:00) cycles. Male Wistar rats (250-300 g) were received 10 mg/kg morphine or its vehicle (Saline, 2 mL/kg/12 h, s.c.) in 13 consecutive days for behavioral assessment tests. Then, naloxone-induced conditioned place aversion and physical signs of withdrawal syndrome were evaluated during light and dark cycles. In contrast to the behavioral part, we performed in vivo extracellular single-unit recording for investigating the neural response of LPGi to naloxone in morphine-dependent rats on day 10 of morphine/saline exposure. Results showed that naloxone induced conditioned place aversion in both light and dark cycles, but the CPA score during the light cycle was larger. Moreover, the intensity of physical signs of morphine withdrawal syndrome was more severe during the light cycle (rest phase) compare to the dark one. In electrophysiological experiments, results indicated that naloxone evoked both excitatory and inhibitory responses in LPGi neurons and the incremental effect of naloxone on LPGi activity was stronger in the light cycle. Also, the neurons with the excitatory response exhibited higher baseline activity in the dark cycle, but the neurons with the inhibitory response showed higher baseline activity in the light cycle. Interestingly, the baseline firing rate of neurons recorded in the light cycle was significantly different in response (excitatory/inhibitory) -dependent manner. We concluded that naloxone-induced changes in LPGi cellular activity and behaviors of morphine-dependent rats can be affected by circadian rhythm and the internal clock.
Collapse
|
12
|
Wang XL, Wang DQ, Jiao FC, Ding KM, Ji YB, Lu L, Yuan K, Gao GF, Li SX. Diurnal rhythm disruptions induced by chronic unpredictable stress relate to depression-like behaviors in rats. Pharmacol Biochem Behav 2021; 204:173156. [PMID: 33675839 DOI: 10.1016/j.pbb.2021.173156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 11/15/2022]
Abstract
The relationship between circadian rhythms and mood disorders has been established. Circadian dysregulations are believed to exacerbate the severity of mood disorders and vice versa. Although many studies on diurnal changes of clock genes in animal model of depression have been performed from the RNA level, only a few studies have been carried out from the protein level. In this study, we investigated the diurnal changes induced by chronic unpredictable stress (CUS) using free-running wheel test and Western Blotting (WB). Besides, we examined the depression-like behaviors of rats by sucrose preference test (SPT) and forced swim test (FST). We found that CUS induced significant reductions in the quantity of free-running wheel activity and rhythmic disruptions of clock proteins in hippocampus. Furthermore, we found that the amplitude of PER1 in CA1 was positively related to the severity of depression-like behaviors. These results suggest that CUS results in both changes in diurnal rhythms and in depression-like behaviors and that it is suggested that these changes are related.
Collapse
Affiliation(s)
- Xin-Ling Wang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China; National Institute on Drug Dependence and Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing 100191, China
| | - De-Quan Wang
- National Institute on Drug Dependence and Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing 100191, China
| | - Fu-Chao Jiao
- Qingdao Agricultural University, Qingdao 266109, China
| | - Kai-Mo Ding
- National Institute on Drug Dependence and Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing 100191, China; Zhenjiang Mental Health Center, Zhenjiang, Jiangsu Province, China
| | - Yan-Bin Ji
- National Institute on Drug Dependence and Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing 100191, China; Center of Psychiatry, Anhui Medical University, Mental Health Center of Anhui Province, Hefei 230032, China
| | - Lin Lu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China; National Institute on Drug Dependence and Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing 100191, China; Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - Kai Yuan
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - George Fu Gao
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China; Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Su-Xia Li
- National Institute on Drug Dependence and Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing 100191, China.
| |
Collapse
|
13
|
Tamura EK, Oliveira-Silva KS, Ferreira-Moraes FA, Marinho EAV, Guerrero-Vargas NN. Circadian rhythms and substance use disorders: A bidirectional relationship. Pharmacol Biochem Behav 2021; 201:173105. [PMID: 33444601 DOI: 10.1016/j.pbb.2021.173105] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 01/23/2023]
Abstract
The circadian system organizes circadian rhythms (biological cycles that occur around 24 h) that couple environmental cues (zeitgebers) with internal functions of the organism. The misalignment between circadian rhythms and external cues is known as chronodisruption and contributes to the development of mental, metabolic and other disorders, including cancer, cardiovascular diseases and addictive disorders. Drug addiction represents a global public health concern and affects the health and well-being of individuals, families and communities. In this manuscript, we reviewed evidence indicating a bidirectional relationship between the circadian system and the development of addictive disorders. We provide information on the interaction between the circadian system and drug addiction for each drug or drug class (alcohol, cannabis, hallucinogens, psychostimulants and opioids). We also describe evidence showing that drug use follows a circadian pattern, which changes with the progression of addiction. Furthermore, clock gene expression is also altered during the development of drug addiction in many brain areas related to drug reward, drug seeking and relapse. The regulation of the glutamatergic and dopaminergic neurocircuitry by clock genes is postulated to be the main circadian mechanism underlying the escalation of drug addiction. The bidirectional interaction between the circadian system and drug addiction seems to be mediated by the effects caused by each drug or class of drugs of abuse. These studies provide new insights on the development of successful strategies aimed at restoring/stabilizing circadian rhythms to reduce the risk for addiction development and relapse.
Collapse
Affiliation(s)
- Eduardo K Tamura
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil.
| | - Kallyane S Oliveira-Silva
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Felipe A Ferreira-Moraes
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Eduardo A V Marinho
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Natalí N Guerrero-Vargas
- Department of Anatomy, Faculty of Medicine, Universidad Nacional Autonóma de México, Av Universidad 3000, Ciudad Universitaria, México City 04510, Mexico
| |
Collapse
|
14
|
Willner MJ, Xiao Y, Kim HS, Chen X, Xu B, Leong KW. Modeling SARS-CoV-2 infection in individuals with opioid use disorder with brain organoids. J Tissue Eng 2021; 12:2041731420985299. [PMID: 33738089 PMCID: PMC7934045 DOI: 10.1177/2041731420985299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/13/2020] [Indexed: 01/10/2023] Open
Abstract
The COVID-19 pandemic has aggravated a preexisting epidemic: the opioid crisis. Much literature has shown that the circumstances imposed by COVID-19, such as social distancing regulations, medical and financial instability, and increased mental health issues, have been detrimental to those with opioid use disorder (OUD). In addition, unexpected neurological sequelae in COVID-19 patients suggest that COVID-19 compromises neuroimmunity, induces hypoxia, and causes respiratory depression, provoking similar effects as those caused by opioid exposure. Combined conditions of COVID-19 and OUD could lead to exacerbated complications. With limited human in vivo options to study these complications, we suggest that iPSC-derived brain organoid models may serve as a useful platform to investigate the physiological connection between COVID-19 and OUD. This mini-review highlights the advances of brain organoids in other neuropsychiatric and infectious diseases and suggests their potential utility for investigating OUD and COVID-19, respectively.
Collapse
Affiliation(s)
- Moshe J Willner
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Hye Sung Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, Republic of Korea
| | - Xuejing Chen
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Physics, Tsinghua University, Beijing, China
| | - Bin Xu
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
15
|
Wang XL, Wang DQ, Jiao FC, Ding KM, Ji YB, Lu L, Yuan K, Gao GF, Li SX. Diurnal rhythm disruptions induced by chronic unpredictable stress relate to depression-like behaviors in rats. Pharmacol Biochem Behav 2020; 194:172939. [PMID: 32437704 DOI: 10.1016/j.pbb.2020.172939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 11/27/2022]
Abstract
The relationship between circadian rhythms and mood disorders has been established, circadian dysregulations are believed to exacerbate the severity of mood disorders and vice versa. Although many studies on diurnal changes of clock genes in animal model of depression have been performed from the RNA level, only a few studies have been carried out from the protein level. In this study, we investigated the diurnal changes induced by chronic unpredictable stress (CUS) using various methods, including free-running wheel test, enzyme-linked immunosorbent assay (ELISA) and Western Blotting (WB). Besides, we examined the depression-like behaviors of rats by sucrose preference test (SPT) and forced swim test (FST). We found that CUS induced significant reductions in the quantity of free-running wheel activity and the amplitude of melatonin secretion rhythm. We also found that CUS induced rhythmic disruptions of clock proteins in hippocampus. Furthermore, we found that the amplitude of PER1 in CA1 was positively related to the severity of depression-like behaviors. These results suggest that stress results in both changes in circadian rhythms and in depression-like behaviors and that it is suggested that these changes are related.
Collapse
Affiliation(s)
- Xin-Ling Wang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing101408, China; National Institute on Drug Dependence, Peking University, Beijing100191, China; Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing100191, China
| | - De-Quan Wang
- National Institute on Drug Dependence, Peking University, Beijing100191, China; Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing100191, China
| | - Fu-Chao Jiao
- Qingdao Agricultural University, Qingdao266109, China
| | - Kai-Mo Ding
- National Institute on Drug Dependence, Peking University, Beijing100191, China; Zhenjiang Mental Health Center, Zhenjiang, Jiangsu Province, China; Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing100191, China
| | - Yan-Bin Ji
- National Institute on Drug Dependence, Peking University, Beijing100191, China; Center of Psychiatry, Anhui Medical University, Mental Health Center of Anhui Province, Hefei230032, China; Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing100191, China
| | - Lin Lu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing101408, China; National Institute on Drug Dependence, Peking University, Beijing100191, China; Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health, Peking University, Beijing100191, China; Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing100191, China; Peking University Sixth Hospital, Peking University, Beijing100191, China
| | - Kai Yuan
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health, Peking University, Beijing100191, China; Peking University Sixth Hospital, Peking University, Beijing100191, China
| | - George Fu Gao
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing101408, China; Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing100101, China; Chinese Center for Disease Control and Prevention, Beijing102206, China.
| | - Su-Xia Li
- National Institute on Drug Dependence, Peking University, Beijing100191, China; Beijing Key Laboratory on Drug Dependence Research, Peking University, Beijing100191, China.
| |
Collapse
|
16
|
Roviš D, Vasiljev V, Jenko-Pražnikar Z, Petelin A, Drevenšek G, Peruč D, Černelič-Bizjak M. Mental health and drug use severity: the role of substance P, neuropeptide Y, self-reported childhood history of trauma, parental bonding and current resiliency. J Ment Health 2019; 30:88-96. [PMID: 31347421 DOI: 10.1080/09638237.2019.1644492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Many risk factors lead to opioid use and drug-related problems. One of the challenges to understand behavioural factors, drug problems and psychopathology is to identify biological markers that are suitable for research on broad substance abuse and dependence involving human participants. AIMS The study has examined the relationships between the self-reported childhood history of trauma, parental bonding, psychopathology, impulsivity, current resiliency, two neuropeptides, possible markers of behaviour and emotion regulation, and severity of drug-related problems. METHODS One hundred and sixty-seven individuals with a history of opioid use completed questionnaires. Serum neuropeptide Y (NPY) and substance P (SP) levels were analysed. Moderating and mediating relationships between variables were examined using structural equation modelling (SEM). RESULTS Antisocial features, depression, impulsivity, SP, NPY, emotional neglect and resilience are associated with severity of drug-related problems. SP is associated with antisocial personality traits. CONCLUSIONS The novelty of this study is the proposed possible link between biochemical markers, antisocial features and behavioural and emotional regulation. Serum NPY and SP levels have a potential to be used as a biomarker in opioid users before and in the treatment process to account for interactions between biological vulnerabilities and childhood risk factors in predicting behavioural adjustment and more severe drug-related problems.
Collapse
Affiliation(s)
- Darko Roviš
- Department of Social Medicine and Epidemiology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Vanja Vasiljev
- Department of Social Medicine and Epidemiology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Zala Jenko-Pražnikar
- Department of Nutritional Counselling - Dietetics, Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Ana Petelin
- Department of Nutritional Counselling - Dietetics, Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Gorazd Drevenšek
- Department of Psychology, Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Koper, Slovenia
| | - Dolores Peruč
- Department of Social Medicine and Epidemiology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Maša Černelič-Bizjak
- Department of Nutritional Counselling - Dietetics, Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| |
Collapse
|
17
|
Zhang P, Moye LS, Southey BR, Dripps I, Sweedler JV, Pradhan A, Rodriguez-Zas SL. Opioid-Induced Hyperalgesia Is Associated with Dysregulation of Circadian Rhythm and Adaptive Immune Pathways in the Mouse Trigeminal Ganglia and Nucleus Accumbens. Mol Neurobiol 2019; 56:7929-7949. [PMID: 31129808 DOI: 10.1007/s12035-019-01650-5] [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] [Received: 02/21/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
The benefits of opioid-based treatments to mitigate chronic pain can be hindered by the side effects of opioid-induced hyperalgesia (OIH) that can lead to higher consumption and risk of addiction. The present study advances the understanding of the molecular mechanisms associated with OIH by comparing mice presenting OIH symptoms in response to chronic morphine exposure (OIH treatment) relative to control mice (CON treatment). Using RNA-Seq profiles, gene networks were inferred in the trigeminal ganglia (TG), a central nervous system region associated with pain signaling, and in the nucleus accumbens (NAc), a region associated with reward dependency. The biological process of nucleic acid processing was over-represented among the 122 genes that exhibited a region-dependent treatment effect. Within the 187 genes that exhibited a region-independent treatment effect, circadian rhythm processes were enriched among the genes over-expressed in OIH relative to CON mice. This enrichment was supported by the differential expression of the period circadian clock 2 and 3 genes (Per2 and Per3). Transcriptional regulators in the PAR bZip family that are influenced by the circadian clock and that modulate neurotransmission associated with pain and drug addiction were also over-expressed in OIH relative to CON mice. Also notable was the under-expression in OIH relative to CON mice of the Toll-like receptor, nuclear factor-kappa beta, and interferon gamma genes and enrichment of the adaptive immune processes. The results from the present study offer insights to advance the effective use of opioids for pain management while minimizing hyperalgesia.
Collapse
Affiliation(s)
- Pan Zhang
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Isaac Dripps
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Amynah Pradhan
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Sandra L Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
18
|
Wang DQ, Wang XL, Wang CY, Wang Y, Li SX, Liu KZ. Effects of chronic cocaine exposure on the circadian rhythmic expression of the clock genes in reward-related brain areas in rats. Behav Brain Res 2019; 363:61-69. [DOI: 10.1016/j.bbr.2019.01.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/09/2019] [Accepted: 01/22/2019] [Indexed: 12/15/2022]
|
19
|
Jeon SY, Kim YH, Kim SJ, Suh SK, Cha HJ. Abuse potential of 2-(4-iodo-2, 5-dimethoxyphenyl)N-(2-methoxybenzyl)ethanamine (25INBOMe); in vivo and ex vivo approaches. Neurochem Int 2019; 125:74-81. [PMID: 30769030 DOI: 10.1016/j.neuint.2019.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/10/2019] [Accepted: 02/12/2019] [Indexed: 01/08/2023]
Abstract
25INBOMe ("25-I", "N-Bomb"), one of new psychoactive substances (NPSs), is being abused for recreational purpose. However, the liability for abuse or dependence has not been systematically studied yet. The objective of the present study was to evaluate rewarding and reinforcing effects of 25INBOMe using conditioned place preference (CPP) and self-administration (SA) paradigms. In addition, ultrasonic vocalizations (USVs) were measured to investigate relationships between USVs and emotional state regarding dependence on psychoactive substances. To understand molecular mechanism involved in its action, dopamine (DA) level changes were analyzed using synaptosomes extracted from the striatal region of the brain. Expression level changes of SGK1 (serum/glucocorticoid regulated kinase 1) and PER2 (period circadian protein homolog 2), two putative biomarkers for drug dependence, were also analyzed. Results showed that 25INBOMe increased both CPP (0.3 mg/kg) and SA (0.03 mg/kg/infusion) and produced higher frequencies in USVs analysis. It also increased DA levels in the striatal region and changed expression levels of SGK1 and PER2. Results of the present study suggest that 25INBOMe might produce rewarding and reinforcing effects, indicating its dependence liability. In addition, frequencies of USV might be associated with emotional state of mice induced by psychoactive substances regarding substance dependence. This is the first systemic preclinical report on the dependence liability of 25INBOMe and the first attempt to introduce a possible relationship between USVs and emotional state of mice regarding substance dependency. Further studies are needed to clarify the mechanism involved in 25INBOMe dependency and determine the usefulness of USV measurement as a method for evaluating dependence liability.
Collapse
Affiliation(s)
- Seo Young Jeon
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea
| | - Young-Hoon Kim
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea
| | - Sung Jin Kim
- Cosmetics Policy Division, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea
| | - Soo Kyung Suh
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea
| | - Hye Jin Cha
- National Institute of Drug and Safety Evaluation, Ministry of Food and Drug Safety, Osong, Cheongju, Republic of Korea.
| |
Collapse
|
20
|
Roy K, Bhattacharyya P, Deb I. Naloxone precipitated morphine withdrawal and clock genes expression in striatum: A comparative study in three different protocols for the development of morphine dependence. Neurosci Lett 2018; 685:24-29. [DOI: 10.1016/j.neulet.2018.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/03/2018] [Accepted: 07/16/2018] [Indexed: 12/21/2022]
|
21
|
Kim SH, Park HG, Jeong SH, Kang UG, Ahn YM, Kim YS. Electroconvulsive Seizure Alters the Expression and Daily Oscillation of Circadian Genes in the Rat Frontal Cortex. Psychiatry Investig 2018; 15:717-726. [PMID: 29945428 PMCID: PMC6056691 DOI: 10.30773/pi.2018.01.18.2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/18/2018] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE Electroconvulsive therapy (ECT) is the most effective treatment for mood disorders. Accumulating evidence has suggested the important role of circadian genes in mood disorders. However, the effects of ECT on circadian genes have not been systemically investigated. METHODS We examined the expression and daily oscillation of major circadian genes in the rat frontal cortex after electroconvulsive seizure (ECS). RESULTS Firstly, mRNA and protein level were investigated at 24 hr after single ECS (E1X) and repeated ECS treatements for 10 days (E10X), which showed more remarkable changes after E10X than E1X. mRNA expression of Rorα, Bmal1, Clock, Per1, and Cry1 was decreased, while Rev-erbα expression was increased at 24 hr after E10X compared to sham. The proteins showed similar pattern of changes. Next, the effects on oscillation and rhythm properties (mesor, amplitude, and acrophase) were examined, which also showed more prominent changes after E10X than E1X. After E10X, mesor of Rorα, Bmal1, and Cry1 was reduced, and that of Rev-erbα was increased. Five genes, Rev-erbα, Bmal1, Per1, Per2, and Cry2, showed earlier acrophase after E10X. CONCLUSION The findings suggest that repeated ECS induces reduced expression and phase advance of major circadian genes in the in vivo rat frontal cortex.
Collapse
Affiliation(s)
- Se Hyun Kim
- Department of Neuropsychiatry, Dongguk University International Hospital, Dongguk University Medical School, Goyang, Republic of Korea
| | - Hong Geun Park
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seong Hoon Jeong
- Department of Psychiatry, Eulji University School of Medicine, Eulji University Hospital, Daejeon, Republic of Korea
| | - Ung Gu Kang
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong Min Ahn
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong Sik Kim
- Department of Neuropsychiatry, Dongguk University International Hospital, Dongguk University Medical School, Goyang, Republic of Korea
| |
Collapse
|
22
|
Onaolapo OJ, Onaolapo AY. Melatonin in drug addiction and addiction management: Exploring an evolving multidimensional relationship. World J Psychiatry 2018; 8:64-74. [PMID: 29988891 PMCID: PMC6033744 DOI: 10.5498/wjp.v8.i2.64] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/06/2018] [Accepted: 05/10/2018] [Indexed: 02/05/2023] Open
Abstract
Melatonin is a pleiotropic signalling molecule that regulates several physiological functions, and synchronises biological rhythms. Recent evidences are beginning to reveal that a dysregulation of endogenous melatonin rhythm or action may play a larger role in the aetiology and behavioural expression of drug addiction, than was previously considered. Also, the findings from a number of animal studies suggest that exogenous melatonin supplementation and therapeutic manipulation of melatonin/melatonin receptor interactions may be beneficial in the management of behavioural manifestations of drug addiction. However, repeated exogenous melatonin administration may cause a disruption of its endogenous rhythm and be associated with potential drawbacks that might limit its usefulness. In this review, we examine the roles of melatonin and its receptors in addictive behaviours; discussing how our understanding of melatonin’s modulatory effects on the brain rewards system and crucial neurotransmitters such as dopamine has evolved over the years. Possible indications(s) for melatonergic agents in addiction management, and how manipulations of the endogenous melatonin system may be of benefit are also discussed. Finally, the potential impediments to application of melatonin in the management of addictive behaviours are considered.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Ladoke Akintola University of Technology, Osogbo 230263, Osun State, Nigeria
| | - Adejoke Y Onaolapo
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho 210211, Oyo State, Nigeria
| |
Collapse
|
23
|
Fan Y, Liang X, Wang R, Song L. Role of endogenous melatoninergic system in development of hyperalgesia and tolerance induced by chronic morphine administration in rats. Brain Res Bull 2017; 135:105-112. [DOI: 10.1016/j.brainresbull.2017.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/11/2017] [Accepted: 10/03/2017] [Indexed: 02/07/2023]
|
24
|
Abstract
Reward-related learning, including that associated with drugs of abuse, is largely mediated by the dopaminergic mesolimbic pathway. Mesolimbic neurophysiology and motivated behavior, in turn, are modulated by the circadian timing system which generates ∼24-h rhythms in cellular activity. Both drug taking and seeking and mesolimbic dopaminergic neurotransmission can vary widely over the day. Moreover, circadian clock genes are expressed in ventral tegmental area dopaminergic cells and in mesolimbic target regions where they can directly modulate reward-related neurophysiology and behavior. There also exists a reciprocal influence between drug taking and circadian timing as the administration of drugs of abuse can alter behavioral rhythms and circadian clock gene expression in mesocorticolimbic structures. These interactions suggest that manipulations of the circadian timing system may have some utility in the treatment of substance abuse disorders. Here, the literature on bidirectional interactions between the circadian timing system and drug taking is briefly reviewed, and potential chronotherapeutic considerations for the treatment of addiction are discussed.
Collapse
|
25
|
Christiansen SL, Bouzinova EV, Fahrenkrug J, Wiborg O. Altered Expression Pattern of Clock Genes in a Rat Model of Depression. Int J Neuropsychopharmacol 2016; 19:pyw061. [PMID: 27365111 PMCID: PMC5137278 DOI: 10.1093/ijnp/pyw061] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Abnormalities in circadian rhythms may be causal factors in development of major depressive disorder. The biology underlying a causal relationship between circadian rhythm disturbances and depression is slowly being unraveled. Although there is no direct evidence of dysregulation of clock gene expression in depressive patients, many studies have reported single-nucleotide polymorphisms in clock genes in these patients. METHODS In the present study we investigated whether a depression-like state in rats is associated with alternations of the diurnal expression of clock genes. The validated chronic mild stress (CMS) animal model of depression was used to investigate rhythmic expression of three clock genes: period genes 1 and 2 (Per1 and Per2) and Bmal1. Brain and liver tissue was collected from 96 animals after 3.5 weeks of CMS (48 control and 48 depression-like rats) at a 4h sampling interval within 24h. We quantified expression of clock genes on brain sections in the prefrontal cortex, nucleus accumbens, pineal gland, suprachiasmatic nucleus, substantia nigra, amygdala, ventral tegmental area, subfields of the hippocampus, and the lateral habenula using in situ hybridization histochemistry. Expression of clock genes in the liver was monitored by real-time quantitative polymerase chain reaction (PCR). RESULTS We found that the effect of CMS on clock gene expression was selective and region specific. Per1 exhibits a robust diurnal rhythm in most regions of interest, whereas Bmal1 and in particular Per2 were susceptible to CMS. CONCLUSION The present results suggest that altered expression of investigated clock genes is likely associated with the induction of a depression-like state in the CMS model.
Collapse
Affiliation(s)
| | | | | | - O Wiborg
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark (Drs Christiansen, Bouzinova, and Wiborg); Department of Clinical Biochemistry, Faculty of Health Sciences, University of Copenhagen, Denmark (Dr Fahrenkrug).
| |
Collapse
|
26
|
Pačesová D, Novotný J, Bendová Z. The effect of chronic morphine or methadone exposure and withdrawal on clock gene expression in the rat suprachiasmatic nucleus and AA-NAT activity in the pineal gland. Physiol Res 2016; 65:517-25. [PMID: 27070740 DOI: 10.33549/physiolres.933183] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The circadian rhythms of many behavioral and physiological functions are regulated by the major circadian pacemaker in the suprachiasmatic nucleus. Long-term opiate addiction and drug withdrawal may affect circadian rhythmicity of various hormones or the sleep/activity pattern of many experimental subjects; however, limited research has been done on the long-term effects of sustained opiate administration on the intrinsic rhythmicity in the suprachiasmatic nucleus and pineal gland. Here we compared the effects of repeated daily treatment of rats with morphine or methadone and subsequent naloxone-precipitated withdrawal on the expression of the Per1, Per2, and Avp mRNAs in the suprachiasmatic nucleus and on arylalkylamine N-acetyltransferase activity in the pineal gland. We revealed that 10-day administration and withdrawal of both these drugs failed to affect clock genes and Avp expression in the SCN. Our results indicate that opioid-induced changes in behavioral and physiological rhythms originate in brain structures downstream of the suprachiasmatic nucleus regulatory output pathway. Furthermore, we observed that acute withdrawal from methadone markedly extended the period of high night AA-NAT activity in the pineal gland. This suggests that withdrawal from methadone, a widely used drug for the treatment of opioid dependence, may have stronger impact on melatonin synthesis than withdrawal from morphine.
Collapse
Affiliation(s)
- D Pačesová
- Department of Physiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic.
| | | | | |
Collapse
|
27
|
Sun HQ, Li SX, Chen FB, Zhang Y, Li P, Jin M, Sun Y, Wang F, Mi WF, Shi L, Yue JL, Yang FD, Lu L. Diurnal neurobiological alterations after exposure to clozapine in first-episode schizophrenia patients. Psychoneuroendocrinology 2016; 64:108-116. [PMID: 26630391 DOI: 10.1016/j.psyneuen.2015.11.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 11/12/2015] [Accepted: 11/13/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Irregular circadian rhythm and some of its most characteristic symptoms are frequently observed in patients with schizophrenia. However, changes in the expression of clock genes or neuropeptides that are related to the regulation of circadian rhythm may influence the susceptibility to recurrence after antipsychotic treatment in schizophrenia, but this possibility has not been investigated. METHODS Blood samples were collected from 15 healthy male controls and 13 male schizophrenia patients at 4h intervals for 24h before and after treatment with clozapine for 8 weeks. The outcome measures included the relative expression of clock gene mRNA PERIOD1 (PER1), PERIOD2 (PER2), PERIOD3 (PER3) and the levels of plasma cortisol, orexin, and insulin. RESULTS Compared with healthy controls, schizophrenia patients presented disruptions in diurnal rhythms of the expression of PER1, PER3, and NPAS2 and the release of orexin, accompanied by a delayed phase in the expression of PER2, decreases in PER3 and NPAS2 expression, and an increase in cortisol levels at baseline. Several of these disruptions (i.e., in PER1 and PER3 expression) persisted after 8 weeks of clozapine treatment, similar to the decreases in the 24-h expression of PER3 and NPAS2. Clozapine treatment for 8 weeks significantly decreased the 24-h levels of PER2 and increased the 24-h levels of insulin. CONCLUSION These persistent neurobiological changes that occur after 8 weeks of clozapine treatment may contribute to the vulnerability to recurrence and efficacy of long-term maintenance treatment in schizophrenia.
Collapse
Affiliation(s)
- Hong-Qiang Sun
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing 100191, China
| | - Su-Xia Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Fang-Bin Chen
- 102 Military Hospital of China, Changzhou 213000, China
| | - Yan Zhang
- 102 Military Hospital of China, Changzhou 213000, China
| | - Peng Li
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing 100191, China; 102 Military Hospital of China, Changzhou 213000, China
| | - Mei Jin
- 102 Military Hospital of China, Changzhou 213000, China
| | - Yan Sun
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Wei-Feng Mi
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing 100191, China
| | - Le Shi
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing 100191, China; National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Jing-Li Yue
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing 100191, China
| | - Fu-De Yang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Lin Lu
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing 100191, China; National Clinical Research Center for Mental Disorder, Peking University, Beijing 100191, China.
| |
Collapse
|
28
|
Short photoperiod condition increases susceptibility to stress in adolescent male rats. Behav Brain Res 2015; 300:38-44. [PMID: 26655789 DOI: 10.1016/j.bbr.2015.12.002] [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: 11/06/2015] [Revised: 11/30/2015] [Accepted: 12/03/2015] [Indexed: 11/22/2022]
Abstract
The seasonality of depressive symptoms is prevalent in children and adolescents. However, the mechanisms that underlie such susceptibility to seasonal influences on mood disorders are unclear. We examined the effects of a short photoperiod condition on the susceptibility to subchronic unpredictable mild stress (SCUS) and rhythmic alterations of plasma corticosterone (CORT), melatonin, and neuropeptide Y (NPY) in adolescent male rats. Compared with the 12h/12h light/dark photoperiod control (CON) rats, the 8h/16h photoperiod SCUS rats exhibited significant anhedonia, a core symptom of human depression, together with a blunted diurnal rhythm and elevation of 24h CORT, melatonin, and NPY levels. The 8h/16h photoperiod condition also blunted the rhythmicity of CORT, caused a phase inversion of melatonin, and caused a phase delay of NPY compared with 12h/12h CON rats. Such abnormalities of plasma CORT, NPY, and melatonin might cause adolescent individuals to present higher stress reactivity and greater vulnerability to stress over their lifetimes. The present study provides evidence of the susceptibility to the seasonality of stress-related disorders in adolescence.
Collapse
|
29
|
Perreau-Lenz S, Spanagel R. Clock genes × stress × reward interactions in alcohol and substance use disorders. Alcohol 2015; 49:351-7. [PMID: 25943583 PMCID: PMC4457607 DOI: 10.1016/j.alcohol.2015.04.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 04/13/2015] [Indexed: 12/31/2022]
Abstract
Adverse life events and highly stressful environments have deleterious consequences for mental health. Those environmental factors can potentiate alcohol and drug abuse in vulnerable individuals carrying specific genetic risk factors, hence producing the final risk for alcohol- and substance-use disorders development. The nature of these genes remains to be fully determined, but studies indicate their direct or indirect relation to the stress hypothalamo-pituitary-adrenal (HPA) axis and/or reward systems. Over the past decade, clock genes have been revealed to be key-players in influencing acute and chronic alcohol/drug effects. In parallel, the influence of chronic stress and stressful life events in promoting alcohol and substance use and abuse has been demonstrated. Furthermore, the reciprocal interaction of clock genes with various HPA-axis components, as well as the evidence for an implication of clock genes in stress-induced alcohol abuse, have led to the idea that clock genes, and Period genes in particular, may represent key genetic factors to consider when examining gene × environment interaction in the etiology of addiction. The aim of the present review is to summarize findings linking clock genes, stress, and alcohol and substance abuse, and to propose potential underlying neurobiological mechanisms.
Collapse
Affiliation(s)
- Stéphanie Perreau-Lenz
- Institute of Psychopharmacology, Central Institute for Mental Health, Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany; SRI International, Center for Neuroscience, Biosciences Division, Menlo Park, CA, USA.
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute for Mental Health, Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
30
|
Webb IC, Lehman MN, Coolen LM. Diurnal and circadian regulation of reward-related neurophysiology and behavior. Physiol Behav 2015; 143:58-69. [PMID: 25708277 DOI: 10.1016/j.physbeh.2015.02.034] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 12/16/2022]
Abstract
Here, we review work over the past two decades that has indicated drug reward is modulated by the circadian system that generates daily (i.e., 24h) rhythms in physiology and behavior. Specifically, drug-self administration, psychomotor stimulant-induced conditioned place preference, and locomotor sensitization vary widely across the day in various species. These drug-related behavioral rhythms are associated with rhythmic neural activity and dopaminergic signaling in the mesocorticolimbic pathways, with a tendency toward increased activity during the species typical wake period. While the mechanisms responsible for such cellular rhythmicity remain to be fully identified, circadian clock genes are expressed in these brain areas and can function locally to modulate both dopaminergic neurotransmission and drug-associated behavior. In addition, neural and endocrine inputs to these brain areas contribute to cellular and reward-related behavioral rhythms, with the medial prefrontal cortex playing a pivotal role. Acute or chronic administration of drugs of abuse can also alter clock gene expression in reward-related brain regions. Emerging evidence suggests that drug craving in humans is under a diurnal regulation and that drug reward may be influenced by clock gene polymorphisms. These latter findings, in particular, indicate that the development of therapeutic strategies to modulate the circadian influence on drug reward may prove beneficial in the treatment of substance abuse disorders.
Collapse
Affiliation(s)
- Ian C Webb
- Dept. of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Michael N Lehman
- Dept. of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lique M Coolen
- Dept. of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA; Dept. of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
31
|
Li SX, Wei YM, Shi HS, Luo YX, Ding ZB, Xue YX, Lu L, Yu CX. Glycogen synthase kinase-3β in the ventral tegmental area mediates diurnal variations in cocaine-induced conditioned place preference in rats. Addict Biol 2014; 19:996-1005. [PMID: 23750993 DOI: 10.1111/adb.12068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cocaine sensitization and reward are reported to be under the influence of diurnal rhythm. However, no previous studies have reported brain areas that play a role as modulators and underlie the mechanism of diurnal variations in cocaine reward. We examined (1) the diurnal rhythm of glycogen synthase kinase-3β (GSK-3β) activity in the suprachiasmatic nucleus (SCN) and reward-related brain areas in naive rats; (2) the effect of day and night on the acquisition of cocaine-induced conditioned place preference (CPP); (3) the influence of cocaine-induced CPP on GSK-3β activity in the SCN and reward-related brain areas; and (4) the effect of the GSK-3β inhibitor SB216763 microinjected bilaterally into the ventral tegmental area (VTA) on cocaine-induced CPP. A significant diurnal rhythm of GSK-3β activity was found in the SCN and reward-related brain areas, with diurnal variations in cocaine-induced CPP. GSK-3β activity in the SCN and reward-related brain areas exhibited marked diurnal variations in rats treated with saline. GSK-3β activity in rats treated with cocaine exhibited distinct diurnal variations only in the prefrontal cortex and VTA. Cocaine decreased the expression of phosphorylated GSK-3β (i.e. increased GSK-3β activity) only in the VTA in rats trained and tested at ZT4 and ZT16. SB216763 microinjected into the VTA bilaterally eliminated the diurnal variations in cocaine-induced CPP, but did not affect the acquisition of cocaine-induced CPP. These findings suggest that the VTA may be a critical area involved in the diurnal variations in cocaine-induced CPP, and GSK-3β may be a regulator of diurnal variations in cocaine-induced CPP.
Collapse
Affiliation(s)
- Su-Xia Li
- National Institute on Drug Dependence, Peking University, China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Hamilton KL, Harris AC, Gewirtz JC. Affective and neuroendocrine effects of withdrawal from chronic, long-acting opiate administration. Brain Res 2013; 1538:73-82. [PMID: 24076207 PMCID: PMC4053187 DOI: 10.1016/j.brainres.2013.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/04/2013] [Accepted: 09/19/2013] [Indexed: 11/22/2022]
Abstract
Although the long-acting opiate methadone is commonly used to treat drug addiction, relatively little is known about the effects of withdrawal from this drug in preclinical models. The current study examined affective, neuroendocrine, and somatic signs of withdrawal from the longer-acting methadone derivative l-alpha-acetylmethydol (LAAM) in rats. Anxiety-like behavior during both spontaneous and antagonist-precipitated withdrawal was measured by potentiation of the startle reflex. Withdrawal elevated corticosterone and somatic signs and blunted circadian variations in baseline startle responding. In addition, fear to an explicit, Pavlovian conditioned stimulus (fear-potentiated startle) was enhanced. These data suggest that anxiety-like behavior as measured using potentiated startle responding does not emerge spontaneously during withdrawal from chronic opiate exposure - in contrast to withdrawal from acute drug exposure - but rather is manifested as exaggerated fear in response to explicit threat cues.
Collapse
Affiliation(s)
- Kathryn L. Hamilton
- Department of Psychology, University of Minnesota, 75 East River Road, Minneapolis, MN 55455, USA,
| | - Andrew C. Harris
- Minneapolis Medical Research Foundation and the Department of Medicine, University of Minnesota; 701 Park Avenue, Minneapolis, MN 55415, USA,
| | - Jonathan C. Gewirtz
- Departments of Psychology and Neuroscience, University of Minnesota, 75 East River Road, Minneapolis, MN 55455, USA,
| |
Collapse
|
33
|
Li SX, Liu LJ, Xu LZ, Gao L, Wang XF, Zhang JT, Lu L. Diurnal alterations in circadian genes and peptides in major depressive disorder before and after escitalopram treatment. Psychoneuroendocrinology 2013; 38:2789-2799. [PMID: 24001941 DOI: 10.1016/j.psyneuen.2013.07.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 07/12/2013] [Accepted: 07/13/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Strong links exist between circadian disturbances and some of the most characteristic symptoms of clinical major depressive disorder (MDD). However, changes in the expression of clock genes or neuropeptides related to the regulation of circadian rhythm that may influence the susceptibility to recurrence after antidepressant treatment in MDD have not been investigated. METHODS Blood samples were collected at 4h intervals for 24h from 12 male healthy controls and 12 male MDD patients before and after treatment with escitalopram for 8 weeks. The outcome measures included the relative expression of clock gene mRNA (PERIOD1, PERIOD2, PERIOD3, CRY1, BMAL1, NPAS2, and GSK-3β), and the levels of serum melatonin, vasoactive intestinal polypeptide (VIP), cortisol, adrenocorticotropic hormone (ACTH), insulin-like growth factor-1 (IGF-1), and growth hormone (GH). RESULTS Compared with healthy controls, MDD patients showed disruptions in the diurnal rhythms of the expression of PERIOD1, PERIOD2, CRY1, BMAL1, NPAS2, and GSK-3β and disruptions in the diurnal rhythms of the release of melatonin, VIP, cortisol, ACTH, IGF-1, and GH. Several of these disruptions (i.e., PER1, CRY1, melatonin, VIP, cortisol, ACTH, and IGF-1) persisted 8 weeks after escitalopram treatment, similar to the increase in the 24h levels of VIP and decreases in the 24h levels of cortisol and ACTH. CONCLUSION These persistent neurobiological changes may play a role in MDD symptoms that are thought to contribute to the vulnerability to recurrence and long-term maintenance therapy.
Collapse
Affiliation(s)
- Su-Xia Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China.
| | | | | | | | | | | | | |
Collapse
|
34
|
Different levels in orexin concentrations and risk factors associated with higher orexin levels: comparison between detoxified opiate and methamphetamine addicts in 5 Chinese cities. BIOMED RESEARCH INTERNATIONAL 2013; 2013:282641. [PMID: 24102051 PMCID: PMC3786501 DOI: 10.1155/2013/282641] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 08/01/2013] [Indexed: 11/17/2022]
Abstract
This study sought to explore the degree of orexin levels in Chinese opiate and methamphetamine addicts and the differences between them. The cross-sectional study was conducted among detoxified drug addicts from Mandatory Detoxification Center (MDC) in five Chinese cities. Orexin levels were assayed with radioimmunoassay (RIA). Mann-Whitney U test and Kruskal-Wallis test were used to detect differences across groups, and logistic regression was used to explore the association between orexin levels and characteristics of demographic and drug abuse. Between November 2009 and January 2011, 285 opiates addicts, 112 methamphetamine addicts, and 79 healthy controls were enrolled. At drug withdrawal period, both opiate and methamphetamine addicts had lower median orexin levels than controls, and median orexin levels in opiate addicts were higher than those in methamphetamine addicts (all above P < 0.05). Adjusted odds of the above median concentration of orexin were higher for injection than "chasing the dragon" (AOR = 3.1, 95% CI = 1.2-7.9). No significant factors associated with orexin levels of methamphetamine addicts were found. Development of intervention method on orexin system by different administration routes especially for injected opiate addicts at detoxification phase may be significant and was welcome.
Collapse
|
35
|
Vidaki A, Daniel B, Court DS. Forensic DNA methylation profiling—Potential opportunities and challenges. Forensic Sci Int Genet 2013; 7:499-507. [DOI: 10.1016/j.fsigen.2013.05.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 05/14/2013] [Indexed: 11/26/2022]
|
36
|
Feng YM, Jia YF, Su LY, Wang D, Lv L, Xu L, Yao YG. Decreased mitochondrial DNA copy number in the hippocampus and peripheral blood during opiate addiction is mediated by autophagy and can be salvaged by melatonin. Autophagy 2013; 9:1395-406. [PMID: 23800874 DOI: 10.4161/auto.25468] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Drug addiction is a chronic brain disease that is a serious social problem and causes enormous financial burden. Because mitochondrial abnormalities have been associated with opiate addiction, we examined the effect of morphine on mtDNA levels in rat and mouse models of addiction and in cultured cells. We found that mtDNA copy number was significantly reduced in the hippocampus and peripheral blood of morphine-addicted rats and mice compared with control animals. Concordantly, decreased mtDNA copy number and elevated mtDNA damage were observed in the peripheral blood from opiate-addicted patients, indicating detrimental effects of drug abuse and stress. In cultured rat pheochromocytoma (PC12) cells and mouse neurons, morphine treatment caused many mitochondrial defects, including a reduction in mtDNA copy number that was mediated by autophagy. Knockdown of the Atg7 gene was able to counteract the loss of mtDNA copy number induced by morphine. The mitochondria-targeted antioxidant melatonin restored mtDNA content and neuronal outgrowth and prevented the increase in autophagy upon morphine treatment. In mice, coadministration of melatonin with morphine ameliorated morphine-induced behavioral sensitization, analgesic tolerance and mtDNA content reduction. During drug withdrawal in opiate-addicted patients and improvement of protracted abstinence syndrome, we observed an increase of serum melatonin level. Taken together, our study indicates that opioid addiction is associated with mtDNA copy number reduction and neurostructural remodeling. These effects appear to be mediated by autophagy and can be salvaged by melatonin.
Collapse
Affiliation(s)
- Yue-Mei Feng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province; Kunming Institute of Zoology; Kunming, Yunnan China; University of Chinese Academy of Sciences; Beijing, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Differential regulation of the period genes in striatal regions following cocaine exposure. PLoS One 2013; 8:e66438. [PMID: 23776671 PMCID: PMC3679086 DOI: 10.1371/journal.pone.0066438] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/09/2013] [Indexed: 11/19/2022] Open
Abstract
Several studies have suggested that disruptions in circadian rhythms contribute to the pathophysiology of multiple psychiatric diseases, including drug addiction. In fact, a number of the genes involved in the regulation of circadian rhythms are also involved in modulating the reward value for drugs of abuse, like cocaine. Thus, we wanted to determine the effects of chronic cocaine on the expression of several circadian genes in the Nucleus Accumbens (NAc) and Caudate Putamen (CP), regions of the brain known to be involved in the behavioral responses to drugs of abuse. Moreover, we wanted to explore the mechanism by which these genes are regulated following cocaine exposure. Here we find that after repeated cocaine exposure, expression of the Period (Per) genes and Neuronal PAS Domain Protein 2 (Npas2) are elevated, in a somewhat regionally selective fashion. Moreover, NPAS2 (but not CLOCK (Circadian Locomotor Output Cycles Kaput)) protein binding at Per gene promoters was enhanced following cocaine treatment. Mice lacking a functional Npas2 gene failed to exhibit any induction of Per gene expression after cocaine, suggesting that NPAS2 is necessary for this cocaine-induced regulation. Examination of Per gene and Npas2 expression over twenty-four hours identified changes in diurnal rhythmicity of these genes following chronic cocaine, which were regionally specific. Taken together, these studies point to selective disruptions in Per gene rhythmicity in striatial regions following chronic cocaine treatment, which are mediated primarily by NPAS2.
Collapse
|
38
|
Erami E, Azhdari-Zarmehri H, Rahmani A, Ghasemi-Dashkhasan E, Semnanian S, Haghparast A. Blockade of orexin receptor 1 attenuates the development of morphine tolerance and physical dependence in rats. Pharmacol Biochem Behav 2012; 103:212-9. [DOI: 10.1016/j.pbb.2012.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 08/12/2012] [Accepted: 08/15/2012] [Indexed: 11/30/2022]
|
39
|
Richardson KA, Aston-Jones G. Lateral hypothalamic orexin/hypocretin neurons that project to ventral tegmental area are differentially activated with morphine preference. J Neurosci 2012; 32:3809-17. [PMID: 22423101 PMCID: PMC3321304 DOI: 10.1523/jneurosci.3917-11.2012] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 12/12/2011] [Accepted: 01/13/2012] [Indexed: 11/21/2022] Open
Abstract
Orexin (or hypocretin) is synthesized exclusively in dorsomedial, perifornical, and lateral hypothalamus (LH). These neurons are implicated in several functions, including reward processing. We examined the ventral tegmental area (VTA) as a possible site of orexin action for drug preference during protracted morphine abstinence, and studied functional topography of orexin projections to VTA. Male Sprague Dawley rats were used to investigate whether orexin cells that project to VTA exhibit Fos activation with morphine conditioned place preference (CPP), and whether these cells exhibit increased Fos with morphine CPP during protracted abstinence. Unilateral injections of a retrograde tracer (WGA-Au, 350-400 nl) were made into the VTA or a nonreward area, locus ceruleus, and morphine or placebo pellets were implanted for 14 d. Approximately 2 weeks after pellet removal (post dependence), CPP conditioning and testing were conducted. Triple labeling for WGA-Au, Fos, and orexin revealed that the percentage of VTA-projecting orexin neurons Fos activated on the CPP test day significantly increased in post-dependent (vs nondependent) rats, and was exclusive to LH orexin neurons (not dorsomedial or perifornical). Post-dependent animals showed a positive correlation between CPP scores and percentages of Fos-activated, caudal VTA-projecting LH orexin cells. Unlike afferents to caudal VTA, percentages of rostral VTA-projecting, LH orexin cells that were Fos activated showed a positive correlation with CPP only in nondependent animals. Fos in LC-projecting orexin cells was not correlated with CPP in any group. These results indicate that VTA is a heterogeneous and functionally significant target of orexin neurons for morphine reward during protracted abstinence.
Collapse
Affiliation(s)
- Kimberlei A. Richardson
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059 and
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Gary Aston-Jones
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
40
|
Glaser AM, Reyes-Vázquez C, Prieto-Gómez B, Burau K, Dafny N. Morphine administration and abrupt cessation alter the behavioral diurnal activity pattern. Pharmacol Biochem Behav 2012; 101:544-52. [PMID: 22386964 DOI: 10.1016/j.pbb.2012.02.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 02/13/2012] [Accepted: 02/18/2012] [Indexed: 01/09/2023]
Abstract
In mammals, there is an underlying mechanism that dictates the organism's biological functions and daily activity schedule, known as circadian rhythms, which play a major role in maintaining steady metabolism, homeostasis, and immunity. Limited research has been done investigating the effects of continuous opiate administration on the circadian rhythm activity pattern. A change in circadian activity pattern is suggested as an experimental model to demonstrate long-term effect of the drug. The objective of this study was to investigate the effects of morphine treatment on the long term activity (24 h) of the animal as well as the activity after abrupt removal, since prescribed medication containing morphine is widely used and abused and its long term effects are not known. Male Sprague-Dawley rats were contained in stable conditions with a standard light/dark cycle recordings taken before, during and after morphine pellet implantation. Cosinor analysis was used to fit a 24-hour curve to the activity pattern. Results indicate that morphine pellet administration alters the mesor, amplitude, the day-time and night-time activity levels, and demonstrates a remarkable change in the maximal circadian rhythm timing during the withdrawal period. The question whether morphine changes the circadian rhythm or a change in circadian rhythm results in tolerance and withdrawal is discussed.
Collapse
Affiliation(s)
- Andrea M Glaser
- Neurobiology and Anatomy, University of Texas Medical School at Houston, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
This paper is the thirty-third consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2010 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, USA.
| |
Collapse
|
42
|
Bayerlein K, Kraus T, Leinonen I, Pilniok D, Rotter A, Hofner B, Schwitulla J, Sperling W, Kornhuber J, Biermann T. Orexin A expression and promoter methylation in patients with alcohol dependence comparing acute and protracted withdrawal. Alcohol 2011; 45:541-7. [PMID: 21621370 DOI: 10.1016/j.alcohol.2011.02.306] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 02/11/2011] [Accepted: 02/21/2011] [Indexed: 12/29/2022]
Abstract
The orexins (hypocretins) are neuropeptides deriving from the lateral hypothalamus and may be of importance within the context of drug craving, withdrawal, and relapse. Therefore, the orexin A expression and promoter methylation in peripheral blood cells of 68 patients (41 male and 27 female patients at three different time points during withdrawal and 27 patients during stationary dehabituation therapy) suffering from alcohol dependence were assessed by quantitative reverse transcription-polymerase chain reaction and bisulfite sequencing. There was a statistically significant difference of orexin A expression between the three time points of withdrawal and long-term (LT) abstinence (F=4.16, P=.011). This difference was most prominent in comparison with LT abstinence (t=-3.08, P=.0032). Expression was significantly associated with the severity of withdrawal symptoms measured with the Withdrawal Syndrome Scale for Alcohol and Related Psychoactive Drugs (WSA) (t=2.17, P=.0356). The stronger the withdrawal symptoms, the lower the orexin A expression (F=4.69, P=.036). Body mass index (t=2.15, P=.041), the severity of withdrawal measured with the WSA (t=2.595, P=.0133), craving measured either by the Obsessive Compulsive Drinking Scale (t=2.77, P=.0085) or the Lübecker Craving Questionnaire (t=-2.23, P=.0314) had a significant influence on orexin A expression taking into account mean methylation of the CpG island of the orexin A promoter during withdrawal. Orexin A may be a possible candidate to further elucidate mechanisms of alcohol withdrawal taking into account energy homoeostasis in the circuit of reward and motivation.
Collapse
Affiliation(s)
- Kristina Bayerlein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Nakatome M, Orii M, Hamajima M, Hirata Y, Uemura M, Hirayama S, Isobe I. Methylation analysis of circadian clock gene promoters in forensic autopsy specimens. Leg Med (Tokyo) 2011; 13:205-9. [PMID: 21596611 DOI: 10.1016/j.legalmed.2011.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 03/15/2011] [Accepted: 03/15/2011] [Indexed: 12/31/2022]
Abstract
DNA methylation in gene promoter regions influences gene expression. Circadian clock genes play an important role in the formation of a biological clock and aberrant methylation of these genes contributes to several disorders. In this study, we examined forensic autopsy specimens to determine whether DNA methylation status in the promoter regions of nine circadian clock genes (Per1, Per2, Per3, Cry1, Cry2, Bmal1, Clock, Tim, and Ck1e) is related to a change in acquired diathesis and/or causes of death. Methylation-specific PCR and direct sequencing methods revealed that the promoters of Per1, Cry2, Bmal1, Clock, and Ck1e were unmethylated in all the forensic autopsy specimens, while the promoters of Per2, Per3, Cry1, and Tim were partially methylated. Methylation status varied between individuals and between tissues in the same patient. A detailed analysis of methylation patterns in the Cry1 promoter region revealed that the patterns also varied between individuals and the Cry1 promoter had highly methylated patterns in two cases that had been exposed to methamphetamine. These results suggest that the methylation status of clock gene promoters varies between individuals. Methamphetamine use may influence methylation in the Cry1 gene promoter region and disturb circadian rhythmicity.
Collapse
Affiliation(s)
- Masato Nakatome
- Department of Legal Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan.
| | | | | | | | | | | | | |
Collapse
|
44
|
Hood S, Cassidy P, Mathewson S, Stewart J, Amir S. Daily morphine injection and withdrawal disrupt 24-h wheel running and PERIOD2 expression patterns in the rat limbic forebrain. Neuroscience 2011; 186:65-75. [PMID: 21536108 DOI: 10.1016/j.neuroscience.2011.04.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 03/30/2011] [Accepted: 04/19/2011] [Indexed: 12/12/2022]
Abstract
Symptoms of opiate withdrawal include disturbances in circadian rhythms. We examined in male Wistar rats (n=48) the effects of a daily, mid-morning morphine injection (5-40 mg/kg, i.p.) and its withdrawal on 24-h wheel-running activity and on the expression of the clock protein, PERIOD2 (PER2), in the suprachiasmatic nucleus (SCN), oval nucleus of the bed nucleus of the stria terminalis (BNSTov), central amygdala (CEA), and dorsal striatum. Rats were killed over 2 days at 10, 22, 46, and 58 h after the last daily morphine injection at zeitgeber times (ZT) 1 or ZT13. Daily morphine injections and their withdrawal suppressed nighttime wheel running, but did not entrain any increase in activity in advance of the injection. Neither morphine injection nor its withdrawal affected PER2 expression in the SCN, whereas the normal daily peaks of PER2 in the BNSTov, CEA, and dorsal striatum were blunted both during morphine administration and its withdrawal. Treatment with a dopaminergic agonist (the D2/3 agonist, quinpirole, 1.0 mg/kg) or a noradrenergic agonist (alpha2 agonist, clonidine, 0.1 mg/kg) in morphine withdrawal did not restore normal PER2 patterns in each affected region; however, both quinpirole and clonidine themselves altered normal daily PER2 expression patterns in morphine-naive rats. These findings confirm and extend previous observations that opiates disrupt daily patterns of clock gene expression in the limbic forebrain. Furthermore, catecholaminergic drugs, which have been previously found to alleviate symptoms of opiate withdrawal, do not alleviate the effects of morphine withdrawal on PER2, but do modulate daily patterns of PER2 expression in saline controls.
Collapse
Affiliation(s)
- S Hood
- Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | | | | | | | | |
Collapse
|