1
|
Krishnan AS, Reichenberger DA, Strayer SM, Master L, Russell MA, Buxton OM, Hale L, Chang AM. Childhood sleep is prospectively associated with adolescent alcohol and marijuana use. Ann Epidemiol 2024; 98:25-31. [PMID: 39043321 PMCID: PMC11387129 DOI: 10.1016/j.annepidem.2024.07.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/15/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Prior studies have examined the cross-sectional relationship between adolescent sleep and substance use; however, fewer have explored the long-term connections between childhood sleep and adolescent substance use. METHODS This study investigated both cross-sectional associations during adolescence and prospective associations between childhood weeknight sleep and later alcohol and marijuana use in the Future of Families and Child Wellbeing Study, a diverse national birth cohort of urban children from 20 cities with populations greater than 200,000. Parents reported their child's bedtime at ages 3, 5, and 9 and their child's sleep duration at ages 5 and 9. RESULTS At age 15, adolescents self-reported their bedtime, sleep duration, and alcohol and marijuana use (n = 1514). Logistic regression analyses for each substance use outcome at age 15 were adjusted for sex, age at time of assessment, race/ethnicity, income-relative-to-poverty threshold, family structure, and caregiver education level. At age 15, later bedtime (AOR=1.39; 95 % CI=1.22, 1.57) and shorter sleep duration (AOR=1.28; 95 % CI=1.14, 1.43) were associated with greater odds of consuming a full drink of alcohol more than once, and later bedtime was associated with greater odds of trying marijuana (AOR=1.35; 95 % CI=1.20, 1.51). Unexpectedly, later bedtimes at age 3 were associated with lower odds of drinking alcohol by age 15 (AOR=0.74; 95 % CI=0.59, 0.92). In contrast, later bedtimes at age 9 were associated with greater odds of drinking alcohol (AOR=1.45; 95 % CI=1.11, 1.90). Additionally, later bedtime at age 5 (AOR=1.26; 95 % CI=1.01, 1.58) and shorter sleep duration at age 9 (AOR=1.19; 95 % CI=1.04, 1.36) were associated with greater odds of trying marijuana. CONCLUSION Taken together, these associations support the importance of protecting childhood sleep habits to reduce the likelihood of substance use starting as early as mid-adolescence. IMPLICATIONS AND CONTRIBUTION In this longitudinal cohort study, adolescents were more likely to have consumed alcohol or tried marijuana by age 15 if they had later bedtimes and shorter sleep duration during childhood and adolescence. Protecting sleep health throughout childhood may reduce the likelihood of substance use during early adolescence.
Collapse
Affiliation(s)
- Akshay S Krishnan
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - David A Reichenberger
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States.
| | - Stephen M Strayer
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - Lindsay Master
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - Michael A Russell
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - Orfeu M Buxton
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - Lauren Hale
- Program in Public Health, Stony Brook University, Stony Brook, NY, United States; Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Anne-Marie Chang
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
2
|
Martins-de-Passos TO, Mesas AE, Beneit N, Díaz-Goñi V, Peral-Martinez F, Cekrezi S, Martinez-Vizcaino V, Jimenez-Lopez E. Are Sleep Parameters and Chronotype Associated with Eating Disorder Risk? A Cross-Sectional Study of University Students in Spain. J Clin Med 2024; 13:5482. [PMID: 39336969 PMCID: PMC11432122 DOI: 10.3390/jcm13185482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Objectives: Eating disorders (EDs) have emerged as a growing public health concern. However, the role of sleep in this context remains underexplored. The aim of this cross-sectional study was to determine the associations between sleep parameters and chronotype with ED risk in a sample of university students in Spain. Methods: ED risk was assessed via the Sick, Control, One stone, Fat, Food Questionnaire, and sleep quality was assessed via the Pittsburgh Sleep Quality Index. Other sleep parameters and chronotypes were self-reported. Sociodemographic, body composition, lifestyle, and depressive symptom data were collected. Logistic and linear regression models adjusted for the main confounders were used to estimate the odds ratios (ORs) and 95% confidence intervals (CIs) of the study associations. Results: A total of 403 students (70.2% female) aged 18 to 30 years participated in the study. Those reporting poor sleep quality (OR = 1.85, 95% CI 1.08-3.17, p = 0.025) and ≤6 h of night-time sleep duration (OR = 4.14, 95% CI 2.00-8.57, p < 0.01) were more likely to be at risk of EDs in the adjusted analyses. The association between night-time sleep duration and the risk of ED did not remain significant when we adjusted for sleep quality. In addition, an evening chronotype was associated with an increased risk of EDs (OR = 1.68, 95% CI 1.07-2.66, p = 0.039) only before adjustment for confounders. Conclusions: Among university students, poorer sleep quality was cross-sectionally associated with EDs. Future prospective studies are needed to examine whether promoting sleep quality may serve as an effective strategy for preventing the risk of EDs.
Collapse
Affiliation(s)
| | - Arthur E Mesas
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16071 Cuenca, Spain
| | - Nuria Beneit
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16071 Cuenca, Spain
| | - Valentina Díaz-Goñi
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16071 Cuenca, Spain
| | | | - Shkelzen Cekrezi
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16071 Cuenca, Spain
| | - Vicente Martinez-Vizcaino
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16071 Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3480559, Chile
| | - Estela Jimenez-Lopez
- Health and Social Research Center, Universidad de Castilla-La Mancha, 16071 Cuenca, Spain
- Department of Psychiatry, Hospital Virgen de La Luz, 16002 Cuenca, Spain
- Center for Biomedical Research Network in Mental Health (CIBERSAM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
3
|
Xu Y, Lin Y, Yu M, Zhou K. The nucleus accumbens in reward and aversion processing: insights and implications. Front Behav Neurosci 2024; 18:1420028. [PMID: 39184934 PMCID: PMC11341389 DOI: 10.3389/fnbeh.2024.1420028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
The nucleus accumbens (NAc), a central component of the brain's reward circuitry, has been implicated in a wide range of behaviors and emotional states. Emerging evidence, primarily drawing from recent rodent studies, suggests that the function of the NAc in reward and aversion processing is multifaceted. Prolonged stress or drug use induces maladaptive neuronal function in the NAc circuitry, which results in pathological conditions. This review aims to provide comprehensive and up-to-date insights on the role of the NAc in motivated behavior regulation and highlights areas that demand further in-depth analysis. It synthesizes the latest findings on how distinct NAc neuronal populations and pathways contribute to the processing of opposite valences. The review examines how a range of neuromodulators, especially monoamines, influence the NAc's control over various motivational states. Furthermore, it delves into the complex underlying mechanisms of psychiatric disorders such as addiction and depression and evaluates prospective interventions to restore NAc functionality.
Collapse
Affiliation(s)
| | | | | | - Kuikui Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
4
|
Ge MJ, Chen G, Zhang ZQ, Yu ZH, Shen JX, Pan C, Han F, Xu H, Zhu XL, Lu YP. Chronic restraint stress induces depression-like behaviors and alterations in the afferent projections of medial prefrontal cortex from multiple brain regions in mice. Brain Res Bull 2024; 213:110981. [PMID: 38777132 DOI: 10.1016/j.brainresbull.2024.110981] [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: 11/19/2023] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION The medial prefrontal cortex (mPFC) forms output pathways through projection neurons, inversely receiving adjacent and long-range inputs from other brain regions. However, how afferent neurons of mPFC are affected by chronic stress needs to be clarified. In this study, the effects of chronic restraint stress (CRS) on the distribution density of mPFC dendrites/dendritic spines and the projections from the cortex and subcortical brain regions to the mPFC were investigated. METHODS In the present study, C57BL/6 J transgenic (Thy1-YFP-H) mice were subjected to CRS to establish an animal model of depression. The infralimbic (IL) of mPFC was selected as the injection site of retrograde AAV using stereotactic technique. The effects of CRS on dendrites/dendritic spines and afferent neurons of the mPFC IL were investigaed by quantitatively assessing the distribution density of green fluorescent (YFP) positive dendrites/dendritic spines and red fluorescent (retrograde AAV recombinant protein) positive neurons, respectively. RESULTS The results revealed that retrograde tracing virus labeled neurons were widely distributed in ipsilateral and contralateral cingulate cortex (Cg1), second cingulate cortex (Cg2), prelimbic cortex (PrL), infralimbic cortex, medial orbital cortex (MO), and dorsal peduncular cortex (DP). The effects of CRS on the distribution density of mPFC red fluorescence positive neurons exhibited regional differences, ranging from rostral to caudal or from top to bottom. Simultaneously, CRS resulted a decrease in the distribution density of basal, proximal and distal dendrites, as well as an increase in the loss of dendritic spines of the distal dendrites in the IL of mPFC. Furthermore, varying degrees of red retrograde tracing virus fluorescence signals were observed in other cortices, amygdala, hippocampus, septum/basal forebrain, hypothalamus, thalamus, mesencephalon, and brainstem in both ipsilateral and contralateral brain. CRS significantly reduced the distribution density of red fluorescence positive neurons in other cortices, hippocampus, septum/basal forebrain, hypothalamus, and thalamus. Conversely, CRS significantly increased the distribution density of red fluorescence positive neurons in amygdala. CONCLUSION Our results suggest a possible mechanism that CRS leads to disturbances in synaptic plasticity by affecting multiple inputs to the mPFC, which is characterized by a decrease in the distribution density of dendrites/dendritic spines in the IL of mPFC and a reduction in input neurons of multiple cortices to the IL of mPFC as well as an increase in input neurons of amygdala to the IL of mPFC, ultimately causing depression-like behaviors.
Collapse
Affiliation(s)
- Ming-Jun Ge
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Geng Chen
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Zhen-Qiang Zhang
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Zong-Hao Yu
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Jun-Xian Shen
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Chuan Pan
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Fei Han
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Hui Xu
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China; Anhui College of Traditional Chinese Medicine, No. 18 Wuxiashan West Road, Wuhu 241002, China
| | - Xiu-Ling Zhu
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China; Department of Anatomy, Wannan Medical College, No. 22 Wenchang West Road, Wuhu 241002, China
| | - Ya-Ping Lu
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China.
| |
Collapse
|
5
|
Wang Y, Li D, Widjaja J, Guo R, Cai L, Yan R, Ozsoy S, Allocca G, Fang J, Dong Y, Tseng GC, Huang C, Huang YH. An EEG Signature of MCH Neuron Activities Predicts Cocaine Seeking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586887. [PMID: 38586019 PMCID: PMC10996698 DOI: 10.1101/2024.03.27.586887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Background Identifying biomarkers that predict substance use disorder (SUD) propensity may better strategize anti-addiction treatment. The melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus (LH) critically mediates interactions between sleep and substance use; however, their activities are largely obscured in surface electroencephalogram (EEG) measures, hindering the development of biomarkers. Methods Surface EEG signals and real-time Ca2+ activities of LH MCH neurons (Ca2+MCH) were simultaneously recorded in male and female adult rats. Mathematical modeling and machine learning were then applied to predict Ca2+MCH using EEG derivatives. The robustness of the predictions was tested across sex and treatment conditions. Finally, features extracted from the EEG-predicted Ca2+MCH either before or after cocaine experience were used to predict future drug-seeking behaviors. Results An EEG waveform derivative - a modified theta-to-delta ratio (EEG Ratio) - accurately tracks real-time Ca2+MCH in rats. The prediction was robust during rapid eye movement sleep (REMS), persisted through REMS manipulations, wakefulness, circadian phases, and was consistent across sex. Moreover, cocaine self-administration and long-term withdrawal altered EEG Ratio suggesting shortening and circadian redistribution of synchronous MCH neuron activities. In addition, features of EEG Ratio indicative of prolonged synchronous MCH neuron activities predicted lower subsequent cocaine seeking. EEG Ratio also exhibited advantages over conventional REMS measures for the predictions. Conclusions The identified EEG Ratio may serve as a non-invasive measure for assessing MCH neuron activities in vivo and evaluating REMS; it may also serve as a potential biomarker predicting drug use propensity.
Collapse
Affiliation(s)
- Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Danyang Li
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | | | - Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Rongzhen Yan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Sahin Ozsoy
- Somnivore Pty. Ltd., Bacchus Marsh, VIC, Australia 3340
| | - Giancarlo Allocca
- Somnivore Pty. Ltd., Bacchus Marsh, VIC, Australia 3340
- Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia 3010
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia
| | - Jidong Fang
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, Hershey, PA 17033
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Chengcheng Huang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219; 15260; 15213
| |
Collapse
|
6
|
He Y, Huang YH, Schlüter OM, Dong Y. Cue- versus reward-encoding basolateral amygdala projections to nucleus accumbens. eLife 2023; 12:e89766. [PMID: 37963179 PMCID: PMC10645419 DOI: 10.7554/elife.89766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
In substance use disorders, drug use as unconditioned stimulus (US) reinforces drug taking. Meanwhile, drug-associated cues (conditioned stimulus [CS]) also gain incentive salience to promote drug seeking. The basolateral amygdala (BLA) is implicated in both US- and CS-mediated responses. Here, we show that two genetically distinct BLA neuronal types, expressing Rspo2 versus Ppp1r1b, respectively, project to the nucleus accumbens (NAc) and form monosynaptic connections with both dopamine D1 and D2 receptor-expressing neurons. While intra-NAc stimulation of Rspo2 or Ppp1r1b presynaptic terminals establishes intracranial self-stimulation (ICSS), only Ppp1r1b-stimulated mice exhibit cue-induced ICSS seeking. Furthermore, increasing versus decreasing the Ppp1r1b-to-NAc, but not Rspo2-to-NAc, subprojection increases versus decreases cue-induced cocaine seeking after cocaine withdrawal. Thus, while both BLA-to-NAc subprojections contribute to US-mediated responses, the Ppp1r1b subprojection selectively encodes CS-mediated reward and drug reinforcement. Such differential circuit representations may provide insights into precise understanding and manipulation of drug- versus cue-induced drug seeking and relapse.
Collapse
Affiliation(s)
- Yi He
- Departments of Neuroscience, University of PittsburghPittsburghUnited States
| | - Yanhua H Huang
- Departments of Psychiatry, University of PittsburghPittsburghUnited States
| | - Oliver M Schlüter
- Departments of Neuroscience, University of PittsburghPittsburghUnited States
| | - Yan Dong
- Departments of Neuroscience, University of PittsburghPittsburghUnited States
- Departments of Psychiatry, University of PittsburghPittsburghUnited States
| |
Collapse
|
7
|
Akhlaghi M, Kohanmoo A. Sleep deprivation in development of obesity, effects on appetite regulation, energy metabolism, and dietary choices. Nutr Res Rev 2023:1-21. [PMID: 37905402 DOI: 10.1017/s0954422423000264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Sleep deprivation, which is a decrease in duration and quality of sleep, is a common problem in today's life. Epidemiological and interventional investigations have suggested a link between sleep deprivation and overweight/obesity. Sleep deprivation affects homeostatic and non-homoeostatic regulation of appetite, with the food reward system playing a dominant role. Factors such as sex and weight status affect this regulation; men and individuals with excess weight seem to be more sensitive to reward-driven and hedonistic regulation of food intake. Sleep deprivation may also affect weight through affecting physical activity and energy expenditure. In addition, sleep deprivation influences food selection and eating behaviours, which are mainly managed by the food reward system. Sleep-deprived individuals mostly crave for palatable energy-dense foods and have low desire for fruit and vegetables. Consumption of meals may not change but energy intake from snacks increases. The individuals have more desire for snacks with high sugar and saturated fat content. The relationship between sleep and the diet is mutual, implying that diet and eating behaviours also affect sleep duration and quality. Consuming healthy diets containing fruit and vegetables and food sources of protein and unsaturated fats and low quantities of saturated fat and sugar may be used as a diet strategy to improve sleep. Since the effects of sleep deficiency differ between animals and humans, only evidence from human subject studies has been included, controversies are discussed and the need for future investigations is highlighted.
Collapse
Affiliation(s)
- Masoumeh Akhlaghi
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Kohanmoo
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Mutti C, Malagutti G, Maraglino V, Misirocchi F, Zilioli A, Rausa F, Pizzarotti S, Spallazzi M, Rosenzweig I, Parrino L. Sleep Pathologies and Eating Disorders: A Crossroad for Neurology, Psychiatry and Nutrition. Nutrients 2023; 15:4488. [PMID: 37892563 PMCID: PMC10610508 DOI: 10.3390/nu15204488] [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: 09/17/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
The intricate connection between eating behaviors and sleep habits is often overlooked in clinical practice, despite their profound interdependence. Sleep plays a key role in modulating psychological, hormonal and metabolic balance and exerting an influence on food choices. Conversely, various eating disorders may affect sleep continuity, sometimes promoting the development of sleep pathologies. Neurologists, nutritionists and psychiatrists tend to focus on these issues separately, resulting in a failure to recognize the full extent of the clinical conditions. This detrimental separation can lead to underestimation, misdiagnosis and inappropriate therapeutic interventions. In this review, we aim to provide a comprehensive understanding of the tangled relationship between sleep, sleep pathologies and eating disorders, by incorporating the perspective of sleep experts, psychologists and psychiatrists. Our goal is to identify a practical crossroad integrating the expertise of all the involved specialists.
Collapse
Affiliation(s)
- Carlotta Mutti
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125 Parma, Italy
| | - Giulia Malagutti
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125 Parma, Italy
| | - Valentina Maraglino
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125 Parma, Italy
| | - Francesco Misirocchi
- Neurology Unit, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy (A.Z.)
| | - Alessandro Zilioli
- Neurology Unit, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy (A.Z.)
| | - Francesco Rausa
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125 Parma, Italy
| | - Silvia Pizzarotti
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125 Parma, Italy
| | - Marco Spallazzi
- Neurology Unit, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy (A.Z.)
| | - Ivana Rosenzweig
- Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London WC2R 2LS, UK
| | - Liborio Parrino
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125 Parma, Italy
- Neurology Unit, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy (A.Z.)
| |
Collapse
|
9
|
Kim K, Kim H, Kong J, Kim JB. Enhanced functional connectivity in the reward circuitry in healthy adults with weekend catch-up sleep. Hum Brain Mapp 2023; 44:4927-4937. [PMID: 37466297 PMCID: PMC10472906 DOI: 10.1002/hbm.26429] [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: 12/18/2022] [Revised: 05/16/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023] Open
Abstract
We aimed to identify structural and functional changes in healthy adults with catch-up sleep (CUS), we applied seed-based functional connectivity (FC) analysis using resting-state functional magnetic resonance imaging (MRI). We hypothesized that deficits in reward processing could be a fundamental mechanism underlying the motivation of taking CUS. Then, 55 healthy adults voluntarily (34 with CUS and 21 without CUS) participated in this study. Voxel-based morphometry was performed to explore region of gray matter volume (GMV) difference between CUS and non-CUS groups. Between-group comparison of FC was then carried out using resting-state functional MRI analysis seeding at the region of volume difference. Moreover, the region of volume difference and the strength of FC were correlated with scores of questionnaires for reward-seeking behavior and clinical variables. CUS group had a higher reward-seeking tendency, and increased GMV in the bilateral nucleus accumbens and right superior frontal gyrus relative to non-CUS group. FC analysis seeding at the bilateral accumbens revealed increases of FC in the bilateral medial prefrontal cortex in CUS group compared to non-CUS group. The questionnaire scores reflecting the reward-seeking tendency were correlated with the FC strength between bilateral accumbens and medial prefrontal cortex. Our results indicate that CUS is associated with reward-seeking tendency and increased GMV and FC in regions responsible for reward network. Our findings suggest that enhanced reward network could be the crucial mechanism underlying taking CUS and might be implicated in the detrimental effects of circadian misalignment.
Collapse
Affiliation(s)
- Keun‐Tae Kim
- Department of NeurologyKorea University Anam Hospital, Korea University College of MedicineSeoulRepublic of Korea
| | - Hayom Kim
- Department of NeurologyKorea University Anam Hospital, Korea University College of MedicineSeoulRepublic of Korea
| | - Jooheon Kong
- Department of NeurologyKorea University Anam Hospital, Korea University College of MedicineSeoulRepublic of Korea
| | - Jung Bin Kim
- Department of NeurologyKorea University Anam Hospital, Korea University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
10
|
Rolle CE, Ng GY, Nho YH, Barbosa DAN, Shivacharan RS, Gold JI, Bassett DS, Halpern CH, Buch V. Accumbens connectivity during deep-brain stimulation differentiates loss of control from physiologic behavioral states. Brain Stimul 2023; 16:1384-1391. [PMID: 37734587 PMCID: PMC10811591 DOI: 10.1016/j.brs.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Loss of control (LOC) eating, the subjective sense that one cannot control what or how much one eats, characterizes binge-eating behaviors pervasive in obesity and related eating disorders. Closed-loop deep-brain stimulation (DBS) for binge eating should predict LOC and trigger an appropriately timed intervention. OBJECTIVE/HYPOTHESIS This study aimed to identify a sensitive and specific biomarker to detect LOC onset for DBS. We hypothesized that changes in phase-locking value (PLV) predict the onset of LOC-associated cravings and distinguish them from potential confounding states. METHODS Using DBS data recorded from the nucleus accumbens (NAc) of two patients with binge eating disorder (BED) and severe obesity, we compared PLV between inter- and intra-hemispheric NAc subregions for three behavioral conditions: craving (associated with LOC eating), hunger (not associated with LOC), and sleep. RESULTS In both patients, PLV in the high gamma frequency band was significantly higher for craving compared to sleep and significantly higher for hunger compared to craving. Maximum likelihood classifiers achieved accuracies above 88% when differentiating between the three conditions. CONCLUSIONS High-frequency inter- and intra-hemispheric PLV in the NAc is a promising biomarker for closed-loop DBS that differentiates LOC-associated cravings from physiologic states such as hunger and sleep. Future trials should assess PLV as a LOC biomarker across a larger cohort and a wider patient population transdiagnostically.
Collapse
Affiliation(s)
- Camarin E Rolle
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Pennsylvania Hospital, Spruce Building 3rd Floor, 801 Spruce Street, Philadelphia, PA 19107, USA; Department of Surgery, Corporal Michael J. Crescenz Veterans Affairs Medical Center, 3900 Woodland Ave, Philadelphia, PA, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Grace Y Ng
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Pennsylvania Hospital, Spruce Building 3rd Floor, 801 Spruce Street, Philadelphia, PA 19107, USA; Department of Surgery, Corporal Michael J. Crescenz Veterans Affairs Medical Center, 3900 Woodland Ave, Philadelphia, PA, USA; Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
| | - Young-Hoon Nho
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Pennsylvania Hospital, Spruce Building 3rd Floor, 801 Spruce Street, Philadelphia, PA 19107, USA; Department of Surgery, Corporal Michael J. Crescenz Veterans Affairs Medical Center, 3900 Woodland Ave, Philadelphia, PA, USA
| | - Daniel A N Barbosa
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Pennsylvania Hospital, Spruce Building 3rd Floor, 801 Spruce Street, Philadelphia, PA 19107, USA; Department of Surgery, Corporal Michael J. Crescenz Veterans Affairs Medical Center, 3900 Woodland Ave, Philadelphia, PA, USA
| | - Rajat S Shivacharan
- Department of Neurosurgery, Stanford University School of Medicine, 453 Quarry Road Office 245C, Stanford, CA 94304, USA
| | - Joshua I Gold
- Department of Neuroscience, University of Pennsylvania, 3700 Hamilton Walk, Richards D407, Philadelphia, PA 19104, USA
| | - Dani S Bassett
- Departments of Bioengineering, Physics and Astronomy, Electrical and Systems Engineering, Neurology, and Psychiatry, University of Pennsylvania, 210 S. 33rd St, Skirkanich Hall 240, Philadelphia, PA 19104, USA; Santa Fe Institute, 1399 Hyde Park Rd, Santa Fe, NM 87501, USA
| | - Casey H Halpern
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Pennsylvania Hospital, Spruce Building 3rd Floor, 801 Spruce Street, Philadelphia, PA 19107, USA; Department of Surgery, Corporal Michael J. Crescenz Veterans Affairs Medical Center, 3900 Woodland Ave, Philadelphia, PA, USA
| | - Vivek Buch
- Department of Neurosurgery, Stanford University School of Medicine, 453 Quarry Road Office 245C, Stanford, CA 94304, USA.
| |
Collapse
|
11
|
Li F, Liu C, Qin S, Wang X, Wan Q, Li Z, Wang L, Yang H, Jiang J, Wu W. The nucleus accumbens functional connectivity in patients with insomnia using resting-state fMRI. Front Neurosci 2023; 17:1234477. [PMID: 37650097 PMCID: PMC10464489 DOI: 10.3389/fnins.2023.1234477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Background The aim of this study was to investigate the functional abnormalities between the nucleus accumbens (NAc) and the whole brain in individuals with Insomnia Disorder (ID) using resting-state functional magnetic resonance imaging (fMRI). Additionally, the study aimed to explore the underlying neural mechanisms of ID. Methods We enrolled 18 participants with ID and 16 normal controls (NC). Resting-state functional connectivity (FC) between the NAc and the whole brain voxels was calculated and compared between the two groups to identify differential brain region. Receiver operating characteristic (ROC) curve analysis was employed to assess the ability of differential features to distinguish between groups. Furthermore, Pearson correlation analysis was performed to examine the relationship between neurocognitive scores and differential features. Results The ID group exhibited significantly reduced FC values in several brain regions, including the right supplementary motor area, the bilateral middle frontal gyrus, the bilateral median cingulate and paracingulate gyri and the left precuneus. The area under the curve (AUC) of the classification model based on FC in these brain regions was 83.3%. Additionally, the abnormal functional changes observed in ID patients were positively correlated with the Fatigue Severity Scale (R = 0.650, p = 0.004). Conclusion These findings suggest that the NAc may play a crucial role in the diagnosis of ID and could serve as a potential imaging biomarker, providing insights into the underlying neural mechanisms of the disorder.
Collapse
Affiliation(s)
- Fangjie Li
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengyong Liu
- Department of Acupuncture-Moxibustion and Rehabilitation, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Shan Qin
- Department of Acupuncture-Moxibustion and Rehabilitation, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Xiaoqiu Wang
- Physical Examination Center, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Qingyun Wan
- Department of Acupuncture-Moxibustion and Rehabilitation, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Zhuoyuan Li
- School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Luyao Wang
- Institute of Biomedical Engineering, School of Life Science, Shanghai University, Shanghai, China
| | - Huayuan Yang
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiehui Jiang
- Institute of Biomedical Engineering, School of Life Science, Shanghai University, Shanghai, China
| | - Wenzhong Wu
- Department of Acupuncture-Moxibustion and Rehabilitation, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
12
|
Glickman B, LaLumiere RT. Theoretical Considerations for Optimizing the Use of Optogenetics with Complex Behavior. Curr Protoc 2023; 3:e836. [PMID: 37439512 PMCID: PMC10406170 DOI: 10.1002/cpz1.836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Optogenetic approaches have allowed researchers to address complex questions about behavior that were previously unanswerable. However, as optogenetic procedures involve a large parameter space across multiple dimensions, it is crucial to consider such parameters in conjunction with the behaviors under study. Here, we discuss strategies to optimize optogenetic approaches with complex behavior by identifying critical experimental design considerations, including frequency specificity, temporal precision, activity-controlled optogenetics, stimulation pattern, and cell-type specificity. We highlight potential limitations or theoretical considerations to be made when manipulating each of these factors of optogenetic experiments. This overview emphasizes the importance of optimizing optogenetic study design to enhance the conclusions that can be drawn about the neuroscience of behavior. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Bess Glickman
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242
| | - Ryan T. LaLumiere
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
13
|
Ten-Blanco M, Flores Á, Cristino L, Pereda-Pérez I, Berrendero F. Targeting the orexin/hypocretin system for the treatment of neuropsychiatric and neurodegenerative diseases: from animal to clinical studies. Front Neuroendocrinol 2023; 69:101066. [PMID: 37015302 DOI: 10.1016/j.yfrne.2023.101066] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/06/2023]
Abstract
Orexins (also known as hypocretins) are neuropeptides located exclusively in hypothalamic neurons that have extensive projections throughout the central nervous system and bind two different G protein-coupled receptors (OX1R and OX2R). Since its discovery in 1998, the orexin system has gained the interest of the scientific community as a potential therapeutic target for the treatment of different pathological conditions. Considering previous basic science research, a dual orexin receptor antagonist, suvorexant, was the first orexin agent to be approved by the US Food and Drug Administration to treat insomnia. In this review, we discuss and update the main preclinical and human studies involving the orexin system with several psychiatric and neurodegenerative diseases. This system constitutes a nice example of how basic scientific research driven by curiosity can be the best route to the generation of new and powerful pharmacological treatments.
Collapse
Affiliation(s)
- Marc Ten-Blanco
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - África Flores
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Neurosciences Institute, University of Barcelona and Bellvitge University Hospital-IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Inmaculada Pereda-Pérez
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Fernando Berrendero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
14
|
Eacret D, Manduchi E, Noreck J, Tyner E, Fenik P, Dunn AD, Schug J, Veasey SC, Blendy JA. Mu-opioid receptor-expressing neurons in the paraventricular thalamus modulate chronic morphine-induced wake alterations. Transl Psychiatry 2023; 13:78. [PMID: 36869037 PMCID: PMC9984393 DOI: 10.1038/s41398-023-02382-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Disrupted sleep is a symptom of many psychiatric disorders, including substance use disorders. Most drugs of abuse, including opioids, disrupt sleep. However, the extent and consequence of opioid-induced sleep disturbance, especially during chronic drug exposure, is understudied. We have previously shown that sleep disturbance alters voluntary morphine intake. Here, we examine the effects of acute and chronic morphine exposure on sleep. Using an oral self-administration paradigm, we show that morphine disrupts sleep, most significantly during the dark cycle in chronic morphine, with a concomitant sustained increase in neural activity in the Paraventricular Nucleus of the Thalamus (PVT). Morphine binds primarily to Mu Opioid Receptors (MORs), which are highly expressed in the PVT. Translating Ribosome Affinity Purification (TRAP)-Sequencing of PVT neurons that express MORs showed significant enrichment of the circadian entrainment pathway. To determine whether MOR + cells in the PVT mediate morphine-induced sleep/wake properties, we inhibited these neurons during the dark cycle while mice were self-administering morphine. This inhibition decreased morphine-induced wakefulness but not general wakefulness, indicating that MORs in the PVT contribute to opioid-specific wake alterations. Overall, our results suggest an important role for PVT neurons that express MORs in mediating morphine-induced sleep disturbance.
Collapse
Affiliation(s)
- Darrell Eacret
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elisabetta Manduchi
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julia Noreck
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma Tyner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Polina Fenik
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Amelia D Dunn
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Schug
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sigrid C Veasey
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Olejniczak I, Begemann K, Wilhelm I, Oster H. The circadian neurobiology of reward. Acta Physiol (Oxf) 2023; 237:e13928. [PMID: 36625310 DOI: 10.1111/apha.13928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/29/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
Circadian clocks are important regulators of physiology and behavior. In the brain, circadian clocks have been described in many centers of the central reward system. They affect neurotransmitter signaling, neuroendocrine circuits, and the sensitivity to external stimulation. Circadian disruption affects reward signaling, promoting the development of behavioral and substance use disorders. In this review, we summarize our current knowledge of circadian clock-reward crosstalk. We show how chronodisruption affects reward signaling in different animal models. We then translate these findings to circadian aspects of human reward (dys-) function and its clinical implications. Finally, we devise approaches to and challenges in implementing the concepts of circadian medicine in the therapy of substance use disorders.
Collapse
Affiliation(s)
- Iwona Olejniczak
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| | - Kimberly Begemann
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| | - Ines Wilhelm
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany.,Translational Psychiatry Unit, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| |
Collapse
|
16
|
Guo R, Vaughan DT, Rojo ALA, Huang YH. Sleep-mediated regulation of reward circuits: implications in substance use disorders. Neuropsychopharmacology 2023; 48:61-78. [PMID: 35710601 PMCID: PMC9700806 DOI: 10.1038/s41386-022-01356-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 12/11/2022]
Abstract
Our modern society suffers from both pervasive sleep loss and substance abuse-what may be the indications for sleep on substance use disorders (SUDs), and could sleep contribute to the individual variations in SUDs? Decades of research in sleep as well as in motivated behaviors have laid the foundation for us to begin to answer these questions. This review is intended to critically summarize the circuit, cellular, and molecular mechanisms by which sleep influences reward function, and to reveal critical challenges for future studies. The review also suggests that improving sleep quality may serve as complementary therapeutics for treating SUDs, and that formulating sleep metrics may be useful for predicting individual susceptibility to SUDs and other reward-associated psychiatric diseases.
Collapse
Affiliation(s)
- Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Allen Institute, Seattle, WA, 98109, USA
| | - Dylan Thomas Vaughan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana Lourdes Almeida Rojo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
17
|
Anastasiades PG, de Vivo L, Bellesi M, Jones MW. Adolescent sleep and the foundations of prefrontal cortical development and dysfunction. Prog Neurobiol 2022; 218:102338. [PMID: 35963360 PMCID: PMC7616212 DOI: 10.1016/j.pneurobio.2022.102338] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022]
Abstract
Modern life poses many threats to good-quality sleep, challenging brain health across the lifespan. Curtailed or fragmented sleep may be particularly damaging during adolescence, when sleep disruption by delayed chronotypes and societal pressures coincides with our brains preparing for adult life via intense refinement of neural connectivity. These vulnerabilities converge on the prefrontal cortex, one of the last brain regions to mature and a central hub of the limbic-cortical circuits underpinning decision-making, reward processing, social interactions and emotion. Even subtle disruption of prefrontal cortical development during adolescence may therefore have enduring impact. In this review, we integrate synaptic and circuit mechanisms, glial biology, sleep neurophysiology and epidemiology, to frame a hypothesis highlighting the implications of adolescent sleep disruption for the neural circuitry of the prefrontal cortex. Convergent evidence underscores the importance of acknowledging, quantifying and optimizing adolescent sleep's contributions to normative brain development and to lifelong mental health.
Collapse
Affiliation(s)
- Paul G Anastasiades
- University of Bristol, Translational Health Sciences, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Luisa de Vivo
- University of Bristol, School of Physiology, Pharmacology & Neuroscience, University Walk, Bristol BS8 1TD, UK; University of Camerino, School of Pharmacy, via Gentile III Da Varano, Camerino 62032, Italy
| | - Michele Bellesi
- University of Bristol, School of Physiology, Pharmacology & Neuroscience, University Walk, Bristol BS8 1TD, UK; University of Camerino, School of Bioscience and Veterinary Medicine, via Gentile III Da Varano, Camerino 62032, Italy
| | - Matt W Jones
- University of Bristol, School of Physiology, Pharmacology & Neuroscience, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
18
|
O'Connor RM, Kenny PJ. Utility of 'substance use disorder' as a heuristic for understanding overeating and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2022; 118:110580. [PMID: 35636576 DOI: 10.1016/j.pnpbp.2022.110580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
Rates of obesity and obesity-associated diseases have increased dramatically in countries with developed economies. Substance use disorders (SUDs) are characterized by the persistent use of the substance despite negative consequences. It has been hypothesized that overconsumption of palatable energy dense food can elicit SUD-like maladaptive behaviors that contribute to persistent caloric intake beyond homeostatic need even in the face of negative consequences. Palatable food and drugs of abuse act on many of the same motivation-related circuits in the brain, and can induce, at least superficially, similar molecular, cellular, and physiological adaptations on these circuits. As such, applying knowledge about the neurobiological mechanisms of SUDs may serve as useful heuristic to better understand the persistent overconsumption of palatable food that contributes to obesity. However, many important differences exist between the actions of drugs of abuse and palatable food in the brain. This warrants caution when attributing weight gain and obesity to the manifestation of a putative SUD-related behavioral disorder. Here, we describe similarities and differences between compulsive drug use in SUDs and overconsumption in obesity and consider the merit of the concept of "food addiction".
Collapse
Affiliation(s)
- Richard M O'Connor
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States of America
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States of America.
| |
Collapse
|
19
|
Yu J, Sesack SR, Huang Y, Schlüter OM, Grace AA, Dong Y. Contingent Amygdala Inputs Trigger Heterosynaptic LTP at Hippocampus-To-Accumbens Synapses. J Neurosci 2022; 42:6581-6592. [PMID: 35840324 PMCID: PMC9410749 DOI: 10.1523/jneurosci.0838-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/14/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
Abstract
The nucleus accumbens shell (NAcSh) is a key brain region where environmental cues acquire incentive salience to reinforce motivated behaviors. Principal medium spiny neurons (MSNs) in the NAcSh receive extensive glutamatergic projections from limbic regions, among which, the ventral hippocampus (vH) transmits information enriched in contextual cues, and the basolateral amygdala (BLA) encodes real-time arousing states. The vH and BLA project convergently to NAcSh MSNs, both activated in a time-locked manner on a cue-conditioned motivational action. In brain slices prepared from male and female mice, we show that co-activation of the two projections induces long-term potentiation (LTP) at vH-to-NAcSh synapses without affecting BLA-to-NAcSh synapses, revealing a heterosynaptic mechanism through which BLA signals persistently increase the temporally contingent vH-to-NAcSh transmission. Furthermore, this LTP is more prominent in dopamine D1 receptor-expressing (D1) MSNs than D2 MSNs and can be prevented by inhibition of either D1 receptors or dopaminergic terminals in NAcSh. This heterosynaptic LTP may provide a dopamine-guided mechanism through which vH-encoded cue inputs that are contingent to BLA activation acquire increased circuit representation to reinforce behavior.SIGNIFICANCE STATEMENT In motivated behaviors, environmental cues associated with arousing stimuli acquire increased incentive salience, processes mediated in part by the nucleus accumbens (NAc). NAc principal neurons receive glutamatergic projections from the ventral hippocampus (vH) and basolateral amygdala (BLA), which transmit information encoding contextual cues and affective states, respectively. Our results show that co-activation of the two projections induces long-term potentiation (LTP) at vH-to-NAc synapses without affecting BLA-to-NAc synapses, revealing a heterosynaptic mechanism through which BLA signals potentiate the temporally contingent vH-to-NAc transmission. Furthermore, this LTP is prevented by inhibition of either D1 receptors or dopaminergic axons. This heterosynaptic LTP may provide a dopamine-guided mechanism through which vH-encoded cue inputs that are contingent to BLA activation acquire increased circuit representation to reinforce behavior.
Collapse
Affiliation(s)
- Jun Yu
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Susan R Sesack
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Yanhua Huang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Oliver M Schlüter
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Anthony A Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
20
|
López-Muciño LA, García-García F, Cueto-Escobedo J, Acosta-Hernández M, Venebra-Muñoz A, Rodríguez-Alba JC. Sleep loss and addiction. Neurosci Biobehav Rev 2022; 141:104832. [PMID: 35988803 DOI: 10.1016/j.neubiorev.2022.104832] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022]
Abstract
Reducing sleep hours is a risk factor for developing cardiovascular, metabolic, and psychiatric disorders. Furthermore, previous studies have shown that reduction in sleep time is a factor that favors relapse in addicted patients. Additionally, animal models have demonstrated that both sleep restriction and sleep deprivation increase the preference for alcohol, methylphenidate, and the self-administration of cocaine. Therefore, the present review discusses current knowledge about the influence of sleep hours reduction on addictivebehaviors; likewise, we discuss the neuronal basis underlying the sleep reduction-addiction relationship, like the role of the orexin and dopaminergic system and neuronal plasticity (i.e., delta FosB expression). Potentially, chronic sleep restriction could increase brain vulnerability and promote addictive behavior.
Collapse
Affiliation(s)
- Luis Angel López-Muciño
- Health Sciences Ph.D. Program, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Fabio García-García
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Jonathan Cueto-Escobedo
- Department of Clinical and Translational Research, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Mario Acosta-Hernández
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Arturo Venebra-Muñoz
- Laboratory of Neurobiology of Addiction and Brain Plasticity, Faculty of Science, Autonomous University of Mexico State, Edomex 50295, Mexico.
| | - Juan Carlos Rodríguez-Alba
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| |
Collapse
|
21
|
Fan BF, Hao B, Dai YD, Xue L, Shi YW, Liu L, Xuan SM, Yang N, Wang XG, Zhao H. Deficiency of Tet3 in nucleus accumbens enhances fear generalization and anxiety-like behaviors in mice. Brain Pathol 2022; 32:e13080. [PMID: 35612904 PMCID: PMC9616092 DOI: 10.1111/bpa.13080] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/02/2022] [Indexed: 11/30/2022] Open
Abstract
Stress‐induced neuroepigenetic programming gains growing more and more interest in the studies of the etiology of posttraumatic stress disorder (PTSD). However, seldom attention is focused on DNA demethylation in fear memory generalization, which is the core characteristic of PTSD. Here, we show that ten‐eleven translocation protein 3 (TET3), the most abundant DNA demethylation enzyme of the TET family in neurons, senses environmental stress and bridges neuroplasticity with behavioral adaptation during fear generalization. Foot shock strength dependently induces fear generalization and TET3 expression in nucleus accumbens (NAc) in mice. Inhibition of DNA demethylation by infusing demethyltransferase inhibitors or AAV‐Tet3‐shRNA virus in NAc enhances the fear generalization and anxiety‐like behavior. Furthermore, TET3 knockdown impairs the dendritic spine density, PSD length, and thickness of neurons, decreases DNA hydroxymethylation (5hmC), reduces the expression of synaptic plasticity‐related genes including Homer1, Cdkn1a, Cdh8, Vamp8, Reln, Bdnf, while surprisingly increases immune‐related genes Stat1, B2m, H2‐Q7, H2‐M2, C3, Cd68 shown by RNA‐seq. Notably, knockdown of TET3 in NAc activates microglia and CD39‐P2Y12R signaling pathway, and inhibition of CD39 reverses the effects of TET3 knockdown on the fear memory generalization and anxiety. Overexpression of TET3 by Crispr‐dSaCas9 virus delivery to activate endogenous Tet3 in NAc increases dendritic spine density of neurons in NAc and reverses fear memory generalization and anxiety‐like behavior in mice. These results suggest that TET3 modulates fear generalization and anxiety via regulating synaptic plasticity and CD39 signaling pathway.
Collapse
Affiliation(s)
- Bu-Fang Fan
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bo Hao
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yun-Da Dai
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Xue
- Department of Psychology, School of Public Medicine, Southern Medical University, Guangzhou, China
| | - Yan-Wei Shi
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lu Liu
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shou-Min Xuan
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ning Yang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Guang Wang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hu Zhao
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
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: 14] [Impact Index Per Article: 7.0] [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
|
23
|
Sleep dysregulation in binge eating disorder and "food addiction": the orexin (hypocretin) system as a potential neurobiological link. Neuropsychopharmacology 2021; 46:2051-2061. [PMID: 34145404 PMCID: PMC8505614 DOI: 10.1038/s41386-021-01052-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022]
Abstract
It has been proposed that binge eating reflects a pathological compulsion driven by the "addictive" properties of foods. Proponents of this argument highlight the large degree of phenomenological and diagnostic overlap between binge eating disorder (BED) and substance use disorders (SUDs), including loss of control over how much is consumed and repeated unsuccessful attempts to abstain from consumption, as well as commonalities in brain structures involved in food and drug craving. To date, very little attention has been given to an additional behavioral symptom that BED shares with SUDs-sleep dysregulation-and the extent to which this may contribute to the pathophysiology of BED. Here, we review studies examining sleep outcomes in patients with BED, which collectively point to a heightened incidence of sleep abnormalities in BED. We identify the orexin (hypocretin) system as a potential neurobiological link between compulsive eating and sleep dysregulation in BED, and provide a comprehensive update on the evidence linking this system to these processes. Finally, drawing on evidence from the SUD literature indicating that the orexin system exhibits significant plasticity in response to drugs of abuse, we hypothesize that chronic palatable food consumption likewise increases orexin system activity, resulting in dysregulated sleep/wake patterns. Poor sleep, in turn, is predicted to exacerbate binge eating, contributing to a cycle of uncontrolled food consumption. By extension, we suggest that pharmacotherapies normalizing orexin signaling, which are currently being trialed for the treatment of SUDs, might also have utility in the clinical management of BED.
Collapse
|
24
|
Cocaine-induced neural adaptations in the lateral hypothalamic melanin-concentrating hormone neurons and the role in regulating rapid eye movement sleep after withdrawal. Mol Psychiatry 2021; 26:3152-3168. [PMID: 33093653 PMCID: PMC8060355 DOI: 10.1038/s41380-020-00921-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022]
Abstract
Sleep abnormalities are often a prominent contributor to withdrawal symptoms following chronic drug use. Notably, rapid eye movement (REM) sleep regulates emotional memory, and persistent REM sleep impairment after cocaine withdrawal negatively impacts relapse-like behaviors in rats. However, it is not understood how cocaine experience may alter REM sleep regulatory machinery, and what may serve to improve REM sleep after withdrawal. Here, we focus on the melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus (LH), which regulate REM sleep initiation and maintenance. Using adult male Sprague-Dawley rats trained to self-administer intravenous cocaine, we did transcriptome profiling of LH MCH neurons after long-term withdrawal using RNA-sequencing, and performed functional assessment using slice electrophysiology. We found that 3 weeks after withdrawal from cocaine, LH MCH neurons exhibit a wide range of gene expression changes tapping into cell membrane signaling, intracellular signaling, and transcriptional regulations. Functionally, they show reduced membrane excitability and decreased glutamatergic receptor activity, consistent with increased expression of voltage-gated potassium channel gene Kcna1 and decreased expression of metabotropic glutamate receptor gene Grm5. Finally, chemogenetic or optogenetic stimulations of LH MCH neural activity increase REM sleep after long-term withdrawal with important differences. Whereas chemogenetic stimulation promotes both wakefulness and REM sleep, optogenetic stimulation of these neurons in sleep selectively promotes REM sleep. In summary, cocaine exposure persistently alters gene expression profiles and electrophysiological properties of LH MCH neurons. Counteracting cocaine-induced hypoactivity of these neurons selectively in sleep enhances REM sleep quality and quantity after long-term withdrawal.
Collapse
|
25
|
García-García F, Priego-Fernández S, López-Muciño LA, Acosta-Hernández ME, Peña-Escudero C. Increased alcohol consumption in sleep-restricted rats is mediated by delta FosB induction. Alcohol 2021; 93:63-70. [PMID: 33662520 DOI: 10.1016/j.alcohol.2021.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 02/10/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
The reduction of sleep hours is a public health problem in contemporary society. It is estimated that humans sleep between 1.5 and 2 h less, per night, than 100 years ago. The reduction of sleep hours is a risk factor for developing cardiovascular, metabolic, and psychiatric problems. Previous studies have shown that low sleep quality is a factor that favors relapse in addicted patients. In rodents, sleep deprivation increases the preference for methylphenidate and the self-administration of cocaine. However, it is unknown whether chronic sleep restriction induces voluntary alcohol consumption in rats and whether alcohol intake is associated with delta FosB expression in the brain reward circuit. Potentially, chronic sleep restriction could make the brain vulnerable and consequently promote addictive behavior. Therefore, the present study's objective was to evaluate alcohol consumption in a chronic sleep restriction model and determine the expression of delta FosB in brains of adult rats. For this purpose, male Wistar rats (300-350 g body weight) were divided into four experimental groups (n = 6 each group): control (without manipulation), sleep restriction (SR) for 7 days, SR and ethanol exposure (Ethanol + SR), and a group with just ethanol exposure (Ethanol). At the end of the management, rats were sacrificed, and the brains were dissected and processed for immunohistochemical detection of delta FosB. The results showed that SR stimulates alcohol consumption compared to unrestricted-sleep rats and induces a significant increase in the number of delta FosB-positive cells in brain nuclei within the motivation/brain reward circuit. These results suggest that chronic reduction of sleep hours is a risk factor for developing a preference for alcohol consumption.
Collapse
Affiliation(s)
- Fabio García-García
- Biomedicine Department, Health Sciences Institute, Veracruzana University, Xalapa, VER, Mexico.
| | - Sergio Priego-Fernández
- Health Sciences Program, Health Sciences Institute, Veracruzana University, Xalapa, VER, Mexico
| | - Luis Angel López-Muciño
- Health Sciences Program, Health Sciences Institute, Veracruzana University, Xalapa, VER, Mexico
| | | | - Carolina Peña-Escudero
- Health Sciences Program, Health Sciences Institute, Veracruzana University, Xalapa, VER, Mexico
| |
Collapse
|
26
|
Gong L, Yu S, Xu R, Liu D, Dai X, Wang Z, Hu Y, Yan B, Kui Y, Zhang B, Feng F. The abnormal reward network associated with insomnia severity and depression in chronic insomnia disorder. Brain Imaging Behav 2021; 15:1033-1042. [PMID: 32710331 DOI: 10.1007/s11682-020-00310-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Previous research has shown that acute sleep deprivation can influence the reward networks. However, it is unclear whether and how the intrinsic reward network is altered in chronic insomnia disorder (CID). In the present study, we aimed to investigate whether the reward network is altered in patients with CID using resting-state functional magnetic resonance imaging (fMRI) data. Forty-two patients with CID and 33 healthy controls (HCs) were enrolled and underwent resting-state fMRI. Nucleus accumbens (NAc) - based functional connectivity (NAFC) was evaluated to explore the differences in the reward network between the CID and HC groups. Pearson correlation analysis was used to evaluate the clinical significance of altered NAFC networks. Compared to those in the HC group, increased NAFC was found in the salience and limbic networks, while decreased NAFC was found in the default mode network (DMN) and within the reward circuit in patients with CID. In addition, decreased FC between the NAc and DMN was associated with insomnia severity, while NAFC within the reward network was associated with depression symptoms in patients with CID. These findings showed that the reward network is dysfunctional and associated with depression symptom in patients with CID. Future studies of CID should consider both insomnia and depression symptoms to disentangle the role of insomnia and depression in the relationship under study.
Collapse
Affiliation(s)
- Liang Gong
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Siyi Yu
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Ronghua Xu
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Duan Liu
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Xijian Dai
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Zhengyan Wang
- Department of Pain Management, Sichuan Integrative Medicine, Sichuan, 610041, Chengdu, China
| | - Youping Hu
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Bohua Yan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China
| | - Yu Kui
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Bei Zhang
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Fen Feng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
27
|
Le Merre P, Ährlund-Richter S, Carlén M. The mouse prefrontal cortex: Unity in diversity. Neuron 2021; 109:1925-1944. [PMID: 33894133 DOI: 10.1016/j.neuron.2021.03.035] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/28/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022]
Abstract
The prefrontal cortex (PFC) is considered to constitute the highest stage of neural integration and to be devoted to representation and production of actions. Studies in primates have laid the foundation for theories regarding the principles of prefrontal function and provided mechanistic insights. The recent surge of studies of the PFC in mice holds promise for evolvement of present theories and development of novel concepts, particularly regarding principles shared across mammals. Here we review recent empirical work on the mouse PFC capitalizing on the experimental toolbox currently privileged to studies in this species. We conclude that this line of research has revealed cellular and structural distinctions of the PFC and neuronal activity with direct relevance to theories regarding the functions of the PFC. We foresee that data-rich mouse studies will be key to shed light on the general prefrontal architecture and mechanisms underlying cognitive aspects of organized actions.
Collapse
Affiliation(s)
- Pierre Le Merre
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Marie Carlén
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden.
| |
Collapse
|
28
|
Chronic sleep fragmentation enhances habenula cholinergic neural activity. Mol Psychiatry 2021; 26:941-954. [PMID: 30980042 PMCID: PMC6790161 DOI: 10.1038/s41380-019-0419-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/21/2019] [Accepted: 03/26/2019] [Indexed: 01/25/2023]
Abstract
Sleep is essential to emotional health. Sleep disturbance, particularly REM sleep disturbance, profoundly impacts emotion regulation, but the underlying neural mechanisms remain elusive. Here we show that chronic REM sleep disturbance, achieved in mice by chronic sleep fragmentation (SF), enhanced neural activity in the medial habenula (mHb), a brain region increasingly implicated in negative affect. Specifically, after a 5-day SF procedure that selectively fragmented REM sleep, cholinergic output neurons (ChNs) in the mHb exhibited increased spontaneous firing rate and enhanced firing regularity in brain slices. The SF-induced firing changes remained intact upon inhibition of glutamate, GABA, acetylcholine, and histamine receptors, suggesting cell-autonomous mechanisms independent of synaptic transmissions. Moreover, the SF-induced hyperactivity was not because of enhanced intrinsic membrane excitability, but was accompanied by depolarized resting membrane potential in mHb ChNs. Furthermore, inhibition of TASK-3 (KCNK9) channels, a subtype of two-pore domain K+ channels, mimicked the SF effects by increasing the firing rate and regularity, as well as depolarizing the resting membrane potential in mHb ChNs in control-sleep mice. These effects of TASK-3 inhibition were absent in SF mice, suggesting reduced TASK-3 activity following SF. By contrast, inhibition of small-conductance Ca2+-activated K+ (SK) channels did not produce similar effects. Thus, SF compromised TASK-3 function in mHb ChNs, which likely led to depolarized resting membrane potential and increased spontaneous firing. These results not only demonstrate that selective REM sleep disturbance leads to hyperactivity of mHb ChNs, but also identify a key molecular substrate through which REM sleep disturbance may alter affect regulation.
Collapse
|
29
|
Meng Q, Tan X, Jiang C, Xiong Y, Yan B, Zhang J. Tracking Eye Movements During Sleep in Mice. Front Neurosci 2021; 15:616760. [PMID: 33716648 PMCID: PMC7947631 DOI: 10.3389/fnins.2021.616760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/04/2021] [Indexed: 12/02/2022] Open
Abstract
Eye movement is not only for adjusting the visual field and maintaining the stability of visual information on the retina, but also provides an external manifestation of the cognitive status of the brain. Recent studies showed similarity in eye movement patterns between wakefulness and rapid eye movement (REM) sleep, indicating that the brain status of REM sleep likely resembles that of awake status. REM sleep in humans could be divided into phasic REM and tonic REM sleep according to the difference in eye movement frequencies. Mice are the most commonly used animal model for studying neuronal and molecular mechanisms underlying sleep. However, there was a lack of details for eye movement patterns during REM sleep, hence it remains unknown whether REM sleep can be further divided into different stages in mice. Here we developed a device combining electroencephalogram (EEG), electromyogram (EMG) as well as eye movements recording in mice to study the eye movement patterns during sleep. We implanted a magnet beneath the conjunctiva of eye and tracked eye movements using a magnetic sensor. The magnetic signals showed strong correlation with video-oculography in head-fixed mice, indicating that the magnetic signals reflect the direction and magnitude of eye movement. We also found that the magnet implanted beneath the conjunctiva exhibited good biocompatibility. Finally, we examined eye movement in sleep–wake cycle, and discriminated tonic REM and phasic REM according to the frequency of eye movements, finding that compared to tonic REM, phasic REM exhibited higher oscillation power at 0.50 Hz, and lower oscillation power at 1.50–7.25 Hz and 9.50–12.00 Hz. Our device allowed to simultaneously record EEG, EMG, and eye movements during sleep and wakefulness, providing a convenient and high temporal-spatial resolution tool for studying eye movements in sleep and other researches in mice.
Collapse
Affiliation(s)
- Qingshuo Meng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinrong Tan
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chengyong Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanyu Xiong
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Biao Yan
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiayi Zhang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Smith MT, Mun CJ, Remeniuk B, Finan PH, Campbell CM, Buenaver LF, Robinson M, Fulton B, Tompkins DA, Tremblay JM, Strain EC, Irwin MR. Experimental sleep disruption attenuates morphine analgesia: findings from a randomized trial and implications for the opioid abuse epidemic. Sci Rep 2020; 10:20121. [PMID: 33208831 PMCID: PMC7674501 DOI: 10.1038/s41598-020-76934-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/03/2020] [Indexed: 11/08/2022] Open
Abstract
Preclinical studies demonstrate that sleep disruption diminishes morphine analgesia and modulates reward processing. We sought to translate these preclinical findings to humans by examining whether sleep disruption alters morphine's analgesic and hedonic properties. We randomized 100 healthy adults to receive morphine versus placebo after two nights of undisturbed sleep (US) and two nights of forced awakening (FA) sleep disruption. Sleep conditions were counterbalanced, separated by a two-week washout. The morning after both sleep conditions, we tested cold pressor pain tolerance before and 40-min after double-blind injection of .08 mg/kg morphine or placebo. The primary outcome was the analgesia index, calculated as the change in cold pressor hand withdrawal latency (HWL) before and after drug injection. Secondary outcomes were ratings of feeling "high," drug "liking," and negative drug effects. We found a significant sleep condition by drug interaction on the analgesia index (95% CI - 0.57, - 0.001). After US, subjects receiving morphine demonstrated significantly longer HWL compared to placebo (95% CI 0.23, 0.65), but not after FA (95% CI - 0.05, 0.38). Morphine analgesia was diminished threefold under FA, relative to US. After FA, females (95% CI - 0.88, - 0.05), but not males (95% CI - 0.23, 0.72), reported decreased subjective "high" effects compared to US. After FA, females (95% CI 0.05, 0.27), but not males (95% CI - 0.10, 0.11), administered morphine reported increased negative drug effects compared to US. These data demonstrate that sleep disruption attenuates morphine analgesia in humans and suggest that sleep disturbed males may be at greatest risk for problematic opioid use.
Collapse
Affiliation(s)
- Michael T Smith
- Division of Behavioral Medicine, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21225, USA.
| | - Chung Jung Mun
- Division of Behavioral Medicine, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21225, USA
| | - Bethany Remeniuk
- Division of Behavioral Medicine, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21225, USA
| | - Patrick H Finan
- Division of Behavioral Medicine, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21225, USA
| | - Claudia M Campbell
- Division of Behavioral Medicine, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21225, USA
| | - Luis F Buenaver
- Division of Behavioral Medicine, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21225, USA
| | | | - Brook Fulton
- Division of Behavioral Medicine, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21225, USA
| | | | | | - Eric C Strain
- Division of Behavioral Medicine, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21225, USA
| | - Michael R Irwin
- Cousins Center for Psychoneuroimmunology, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, 90024, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine At UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
31
|
Brunet JF, McNeil J, Doucet É, Forest G. The association between REM sleep and decision-making: Supporting evidences. Physiol Behav 2020; 225:113109. [DOI: 10.1016/j.physbeh.2020.113109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/17/2020] [Accepted: 07/25/2020] [Indexed: 01/08/2023]
|
32
|
Tabibnia G. An affective neuroscience model of boosting resilience in adults. Neurosci Biobehav Rev 2020; 115:321-350. [DOI: 10.1016/j.neubiorev.2020.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 05/09/2020] [Accepted: 05/10/2020] [Indexed: 12/11/2022]
|
33
|
Wang Y, Liu Z, Cai L, Guo R, Dong Y, Huang YH. A Critical Role of Basolateral Amygdala-to-Nucleus Accumbens Projection in Sleep Regulation of Reward Seeking. Biol Psychiatry 2020; 87:954-966. [PMID: 31924324 PMCID: PMC7210061 DOI: 10.1016/j.biopsych.2019.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/09/2019] [Accepted: 10/27/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Sleep impacts reward-motivated behaviors partly by retuning the brain reward circuits. The nucleus accumbens (NAc) is a reward processing hub sensitive to acute sleep deprivation. Glutamatergic transmission carrying reward-associated signals converges in the NAc and regulates various aspects of reward-motivated behaviors. The basolateral amygdala projection (BLAp) innervates broad regions of the NAc and critically regulates reward seeking. METHODS Using slice electrophysiology, we measured how acute sleep deprivation alters transmission at BLAp-NAc synapses in male C57BL/6 mice. Moreover, using SSFO (stabilized step function opsin) and DREADDs (designer receptors exclusively activated by designer drugs) (Gi) to amplify and reduce transmission, respectively, we tested behavioral consequences following bidirectional manipulations of BLAp-NAc transmission. RESULTS Acute sleep deprivation increased sucrose self-administration in mice and altered the BLAp-NAc transmission in a topographically specific manner. It selectively reduced glutamate release at the rostral BLAp (rBLAp) onto ventral and lateral NAc (vlNAc) synapses, but spared caudal BLAp onto medial NAc synapses. Furthermore, experimentally facilitating glutamate release at rBLAp-vlNAc synapses suppressed sucrose reward seeking. Conversely, mimicking sleep deprivation-induced reduction of rBLAp-vlNAc transmission increased sucrose reward seeking. Finally, facilitating rBLAp-vlNAc transmission per se did not promote either approach motivation or aversion. CONCLUSIONS Sleep acts on rBLAp-vINAc transmission gain control to regulate established reward seeking but does not convey approach motivation or aversion on its own.
Collapse
Affiliation(s)
- Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA,These authors contributed equally to this work
| | - Zheng Liu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,These authors contributed equally to this work
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Yan Dong
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
34
|
Neisewander JL. Sleep Deprivation Dysregulates Brain Reward Circuitry: Implications for Psychiatric Disorders. Biol Psychiatry 2020; 87:938-939. [PMID: 32446315 DOI: 10.1016/j.biopsych.2020.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/28/2022]
|
35
|
Sleep quality improves during treatment with repetitive transcranial magnetic stimulation (rTMS) in patients with cocaine use disorder: a retrospective observational study. BMC Psychiatry 2020; 20:153. [PMID: 32252720 PMCID: PMC7137315 DOI: 10.1186/s12888-020-02568-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sleep disturbance is a prominent and common complaint in people with cocaine use disorder (CUD), either during intake or withdrawal. Repetitive transcranial magnetic stimulation (rTMS) has shown promise as a treatment for CUD. Thus, we evaluated the relationship between self-perceived sleep quality and cocaine use pattern variables in outpatients with CUD undergoing an rTMS protocol targeted at the left dorsolateral prefrontal cortex. METHODS This is a retrospective observational study including 87 patients diagnosed with CUD according to the DSM-5 criteria. Scores in Pittsburgh Sleep Quality Index (PSQI), Cocaine Craving Questionnaire (CCQ), Beck Depression Inventory-II (BDI-II), Self-rating Anxiety Scale (SAS), and Symptoms checklist 90-Revised (outcome used: Global Severity Index, GSI) were recorded at baseline, and after 5, 30, 60, and 90 days of rTMS treatment. Cocaine use was assessed by self-report and regular urine screens. RESULTS Sleep disturbances (PSQI scores > 5) were common in patients at baseline (mean ± SD; PSQI score baseline: 9.24 ± 3.89; PSQI > 5 in 88.5% of patients). PSQI scores significantly improved after rTMS treatment (PSQI score Day 90: 6.12 ± 3.32). Significant and consistent improvements were also seen in craving and in negative-affect symptoms compared to baseline. Considering the lack of a control group, in order to help the conceptualization of the outcomes, we compared the results to a wait-list group (n = 10). No significant improvements were observed in the wait-list group in any of the outcome measures. CONCLUSIONS The present findings support the therapeutic role of rTMS interventions for reducing cocaine use and accompanying symptoms such as sleep disturbance and negative-affect symptoms. TRIAL REGISTRATION ClinicalTrials.gov.NCT03733821.
Collapse
|
36
|
Early-life stress alters sleep structure and the excitatory-inhibitory balance in the nucleus accumbens in aged mice. Chin Med J (Engl) 2020; 132:1582-1590. [PMID: 31045908 PMCID: PMC6616240 DOI: 10.1097/cm9.0000000000000279] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: Exposure to adverse experiences in early life may profoundly reshape the neurodevelopmental trajectories of the brain and lead to long-lasting behavioral and neural alterations. One deleterious effect of early-life stress that manifests in later life is sleep disturbance, but this has not been examined in aged mice and the underlying neural mechanisms remain unknown. Considering the important role of the nucleus accumbens (NAc) in the sleep-wake regulation, this study aimed to assess the effects of early-life stress on the sleep behaviors in aged mice and the potential involvement of the NAc in stress-induced sleep abnormalities. Methods: Twenty aged male C57BL/6 mice (>16 months, n = 10 per group) were used in this study. During post-natal days 2 to 9, dams were provided with either sufficient (control) or a limited nesting and bedding materials (stressed). When the mice were 16 to 17 months old, their sleep-wake behaviors were recorded over 24 h using electroencephalogram and electromyelogram. The amount of each sleep-wake stage, mean duration, and stage transition was analyzed. Then, five animals were randomly chosen from each group and were used to measure the expression levels of vesicular glutamate transporter-1 (VGluT1) and vesicular transporters of γ-aminobutyric acid (VGAT) in the NAc using immunohistochemistry. Group comparisons were carried out using Student t test or analysis of variances when appropriate. Results: Compared with the control mice, the early-life stressed aged mice spent less time awake over 24 h (697.97 ± 77.47 min vs. 631.33 ± 34.73 min, t17 = 2.376, P = 0.030), accordingly, non-rapid eye movement sleep time was increased (667.37 ± 62.07 min vs. 723.54 ± 39.21 min, t17 = 2.326, P = 0.033) and mean duration of rapid eye movement sleep was prolonged (73.00 ± 8.98 min vs. 89.39 ± 12.69 min, t17 = 3.277, P = 0.004). Meanwhile, we observed decreased VGluT1/VGAT ratios in the NAc in the stressed group (F(1, 16) = 81.04, P < 0.001). Conclusion: Early adverse experiences disrupt sleep behaviors in aged mice, which might be associated with the excitatory-inhibitory imbalance in the NAc.
Collapse
|
37
|
McDevitt DS, Jonik B, Graziane NM. Morphine Differentially Alters the Synaptic and Intrinsic Properties of D1R- and D2R-Expressing Medium Spiny Neurons in the Nucleus Accumbens. Front Synaptic Neurosci 2019; 11:35. [PMID: 31920618 PMCID: PMC6932971 DOI: 10.3389/fnsyn.2019.00035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 12/06/2019] [Indexed: 12/31/2022] Open
Abstract
Exposure to opioids reshapes future reward and motivated behaviors partially by altering the functional output of medium spiny neurons (MSNs) in the nucleus accumbens shell. Here, we investigated how morphine, a highly addictive opioid, alters synaptic transmission and intrinsic excitability on dopamine D1-receptor (D1R) expressing and dopamine D2-receptor (D2R) expressing MSNs, the two main output neurons in the nucleus accumbens shell. Using whole-cell electrophysiology recordings, we show, that 24 h abstinence following repeated non-contingent administration of morphine (10 mg/kg, i.p.) in mice reduces the miniature excitatory postsynaptic current (mEPSC) frequency and miniature inhibitory postsynaptic current (mIPSC) frequency on D2R-MSNs, with concomitant increases in D2R-MSN intrinsic membrane excitability. We did not observe any changes in synaptic or intrinsic changes on D1R-MSNs. Last, in an attempt to determine the integrated effect of the synaptic and intrinsic alterations on the overall functional output of D2R-MSNs, we measured the input-output efficacy by measuring synaptically-driven action potential firing. We found that both D1R-MSN and D2R-MSN output was unchanged following morphine treatment.
Collapse
Affiliation(s)
- Dillon S McDevitt
- Departments of Anesthesiology and Perioperative Medicine, and Pharmacology, Penn State College of Medicine, Hershey, PA, United States.,Neuroscience Graduate Program, Penn State College of Medicine, Hershey, PA, United States
| | - Benjamin Jonik
- Medical Student Research Program, Penn State College of Medicine, Hershey, PA, United States
| | - Nicholas M Graziane
- Departments of Anesthesiology and Perioperative Medicine, and Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
38
|
Timing of Morphine Administration Differentially Alters Paraventricular Thalamic Neuron Activity. eNeuro 2019; 6:ENEURO.0377-19.2019. [PMID: 31801741 PMCID: PMC6920517 DOI: 10.1523/eneuro.0377-19.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 12/29/2022] Open
Abstract
The paraventricular thalamic nucleus (PVT) is a brain region involved in regulating arousal, goal-oriented behaviors, and drug seeking, all key factors playing a role in substance use disorder. Given this, we investigated the temporal effects of administering morphine, an opioid with strongly addictive properties, on PVT neuronal function in mice using acute brain slices. Here, we show that morphine administration and electrophysiological recordings that occur during periods of animal inactivity (light cycle) elicit increases in PVT neuronal function during a 24-h abstinence time point. Furthermore, we show that morphine-induced increases in PVT neuronal activity at 24-h abstinence are occluded when morphine administration and recordings are performed during an animals' active state (dark cycle). Based on our electrophysiological results combined with previous findings demonstrating that PVT neuronal activity regulates drug-seeking behaviors, we investigated whether timing morphine administration with periods of vigilance (dark cycle) would decrease drug-seeking behaviors in an animal model of substance use disorder. We found that context-induced morphine-seeking behaviors were intact regardless of the time morphine was administered (e.g., light cycle or dark cycle). Our electrophysiological results suggest that timing morphine with various states of arousal may impact the firing of PVT neurons during abstinence. Although, this may not impact context-induced drug-seeking behaviors.
Collapse
|
39
|
Harkness JH, Bushana PN, Todd RP, Clegern WC, Sorg BA, Wisor JP. Sleep disruption elevates oxidative stress in parvalbumin-positive cells of the rat cerebral cortex. Sleep 2019; 42:5145871. [PMID: 30371896 DOI: 10.1093/sleep/zsy201] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Indexed: 11/13/2022] Open
Abstract
We used a novel automated sleep disruption (SD) apparatus to determine the impact of SD on sleep and molecular markers of oxidative stress in parvalbumin (PV) neurons in the rat prefrontal cortex (PFC). Rats were subjected to two 6 hr SD sessions from zeitgeber time (ZT) 0 to ZT6, one by the gentle handling method and the other by an automated agitator running the length of the rat's home cage floor (a novel SD method). The same rats were later subjected to a 12 hr SD session from ZT0 to ZT12. Sleep was disrupted with both methods, although rats slept less during gentle handling than during the automated condition. Immediately after both SD sessions, rats displayed compensatory sleep characterized by elevated slow-wave activity. We measured in the prelimbic prefrontal cortex (prelimbic PFC; 6 and 12 hr SD) and orbital frontal cortex (12 hr SD) the intensity of the oxidative stress marker, 8-oxo-2'-deoxyguanosine (8-oxo-dG) as well as the staining intensity of PV and the PV cell-associated perineuronal net marker, Wisteria floribunda agglutinin (WFA). In the prelimbic PFC, 6 hr SD increased the intensity of 8-oxo-dG, PV, and WFA. After 12 hr SD, the intensity of 8-oxo-dG was elevated in all neurons. PV intensity was elevated only in neurons colabeled with 8-oxo-dG or WFA, and no changes were found in WFA intensity. We conclude that in association with SD-induced sleep drive, PV neurons in the prelimbic PFC exhibit oxidative stress.
Collapse
Affiliation(s)
- John H Harkness
- Department of Integrative Physiology and Neuroscience, Translational Addiction Research Center, Washington State University, Vancouver, WA
| | - Priyanka N Bushana
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Spokane, WA
| | - Ryan P Todd
- Department of Integrative Physiology and Neuroscience, Translational Addiction Research Center, Washington State University, Vancouver, WA
| | - William C Clegern
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Spokane, WA
| | - Barbara A Sorg
- Department of Integrative Physiology and Neuroscience, Translational Addiction Research Center, Washington State University, Vancouver, WA
| | - Jonathan P Wisor
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Spokane, WA
| |
Collapse
|
40
|
Ahrens AM, Ahmed OJ. Neural circuits linking sleep and addiction: Animal models to understand why select individuals are more vulnerable to substance use disorders after sleep deprivation. Neurosci Biobehav Rev 2019; 108:435-444. [PMID: 31756346 DOI: 10.1016/j.neubiorev.2019.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 10/26/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
Abstract
Individuals differ widely in their drug-craving behaviors. One reason for these differences involves sleep. Sleep disturbances lead to an increased risk of substance use disorders and relapse in only some individuals. While animal studies have examined the impact of sleep on reward circuitry, few have addressed the role of individual differences in the effects of altered sleep. There does, however, exist a rodent model of individual differences in reward-seeking behavior: the sign/goal-tracker model of Pavlovian conditioned approach. In this model, only some rats show the key behavioral traits associated with addiction, including impulsivity and poor attentional control, making this an ideal model system to examine individually distinct sleep-reward interactions. Here, we describe how the limbic neural circuits responsible for individual differences in incentive motivation overlap with those involved in sleep-wake regulation, and how this model can elucidate the common underlying mechanisms. Consideration of individual differences in preclinical models would improve our understanding of how sleep interacts with motivational systems, and why sleep deprivation contributes to addiction in only select individuals.
Collapse
Affiliation(s)
| | - Omar J Ahmed
- Dept. of Psychology, United States; Neuroscience Graduate Program, United States; Michigan Center for Integrative Research in Critical Care, United States; Kresge Hearing Research Institute, United States; Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
41
|
Burunat E. Love is a physiological motivation (like hunger, thirst, sleep or sex). Med Hypotheses 2019; 129:109225. [PMID: 31371074 DOI: 10.1016/j.mehy.2019.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/17/2019] [Accepted: 05/12/2019] [Indexed: 10/26/2022]
Abstract
The multitude of terms associated with love has given rise to a false perception of love. In this paper, only maternal and romantic love are considered. Love is usually regarded as a feeling, motivation, addiction, passion, and, above all, an emotion. This confusion has consequences in the lives of human beings, leading not only to divorces, suicides, femicides but possibly also to a number of mental illnesses and suffering. Therefore, it is crucial to first clarify what is meant by emotion, motivation and love. This work aims to finally place love within the category of physiological motivations, such as hunger, thirst, sleep, or sex, on the basis that love is also essential for human survival, especially in childhood. Love is presented from an evolutionary perspective. Some other similarities between love and other physiological motivations are pointed out, such as its importance for appropriate human development, both its ontogeny and its permanence, and the long-lasting consequences of abuse and neglect. There are summarized reasons that account for this, such as the fact that physiological motivations are essential for survival and that love is an essential motivation for the survival of human offspring. Other reasons are that minimum changes in the quantity and quality of love alters development, that there can be a variety of neurophysiological and behavioural states within a motivation, and that motivations (also love) appear and change throughout development. Also, motivations and love sometimes may lead to an addictive behaviour. Finally, it is recognized that once physiological motivations (and love) appear, they become permanent. In a third section, some potential social, cultural, clinical and scientific consequences of the proposed consideration of love as a motivation are discussed. Accordingly, love's recognition as a motivation in the clinical field would imply a better understanding of its disorders and its inclusion in classifications manuals such as The Diagnostic and Statistical Manual of Mental Disorders (DSM), or in the International Classification of Diseases (ICD). Considering love as a motivation rather than an emotion could also impact the results of scientific research (an example is included). A comprehensive understanding of these questions could potentially allow for a new therapeutic approach in the treatment of mental illness, while offering an all-inclusive evolutionary explanation of cultural phenomena such as the origin and diffusion of both language and art. Love should be understood as a physiological motivation, like hunger, sleep or sex, and not as an emotion as it is commonly considered.
Collapse
Affiliation(s)
- Enrique Burunat
- School of Health Sciences/School of Psychology, Department of Clinical Psychology, Psychobiology and Methodology, University of La Laguna, P.O. Box 456, 38200 Santa Cruz de Tenerife, Canary Islands, Spain.
| |
Collapse
|
42
|
Fang X, Zhan G, Zhang J, Xu H, Zhu B, Hu Y, Yang C, Luo A. Abnormalities in Inflammatory Cytokines Confer Susceptible to Chronic Neuropathic Pain-related Anhedonia in a Rat Model of Spared Nerve Injury. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2019; 17:189-199. [PMID: 30905119 PMCID: PMC6478091 DOI: 10.9758/cpn.2019.17.2.189] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/31/2018] [Accepted: 02/05/2018] [Indexed: 12/15/2022]
Abstract
Objective Patients with chronic neuropathic pain (CNP) have a higher incidence to develop depression. However, its pathogenesis has not yet been fully elucidated. Here we aimed to investigate the role of inflammatory cytokines in CNP-related anhedonia, which is a core symptom of depression, and to explore the effects of ketamine and parecoxib on pain and anhedonia. Methods A rat model of spared nerve injury (SNI) was constructed to mimic CNP. Hierarchical cluster analysis of sucrose preference test (SPT) was applied to classify the SNI rats into anhedonia susceptible and unsusceptible. Inflammatory cytokines in medial prefrontal cortex (mPFC) of brain, serum and L2–5 spinal cord were measured. Moreover, effects of ketamine or parecoxib on mechanical withdrawal test (MWT) and SPT in anhedonia susceptible rats were detected. Results Tumor necrosis factor (TNF)-α was increased in mPFC, serum and and spinal cord of anhedonia susceptible rats. Furthermore, anhedonia susceptible and unsusceptible rats both increased the interleukin (IL)-1β level in mPFC, serum and spinal cord. IL-6 was altered in serum and spinal cord, but not in mPFC. IL-10 was significantly altered in mPFC and serum, but not in spinal cord. Additionally, ketamine treatment significantly attenuated the decreased results of MWT and SPT in anhedonia susceptible rats, and that parecoxib significantly improved the MWT score, but failed to alter the result of SPT. Conclusion These findings suggest that abnormalities in inflammatory cytokines confer susceptible to anhedonia in a rat model of SNI. Ketamine, a fast-acting antidepressant, has pharmacological benefits to alleviate pain and anhedonia symptoms.
Collapse
Affiliation(s)
- Xi Fang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yimin Hu
- Department of Anestesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Mu P, Huang YH. Cholinergic system in sleep regulation of emotion and motivation. Pharmacol Res 2019; 143:113-118. [PMID: 30894329 DOI: 10.1016/j.phrs.2019.03.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/28/2019] [Accepted: 03/15/2019] [Indexed: 01/22/2023]
Abstract
Sleep profoundly regulates our emotional and motivational state of mind. Human brain imaging and animal model studies are providing initial insights on the underlying neural mechanisms. Here, we focus on the brain cholinergic system, including cholinergic neurons in the basal forebrain, ventral striatum, habenula, and brain stem. Although much is learned about cholinergic regulations of emotion and motivation, less is known on their interactions with sleep. Specifically, we present an anatomical framework that highlights cholinergic signaling in the integrated reward-arousal/sleep circuitry, and identify the knowledge gaps on the potential roles of cholinergic system in sleep-mediated regulation of emotion and motivation. Sleep impacts every aspect of brain functions. It not only restores cognitive control, but also retunes emotional and motivational regulation [1]. Sleep disturbance is a comorbidity and sometimes a predicting factor for various psychiatric diseases including major depressive disorder, anxiety, post-traumatic stress disorder, and drug addiction [2-9]. Although it is well recognized that sleep prominently shapes emotional and motivational regulation, the underlying neural mechanisms remain elusive. The brain cholinergic system is essential for a diverse variety of functions including cognition, learning and memory, sensory and motor processing, sleep and arousal, reward processing, and emotion regulation [10-14]. Although cholinergic functions in cognition, learning and memory, motor control, and sleep and arousal have been well established, its interaction with sleep in regulating emotion and motivation has not been extensively studied. Here we review current evidence on sleep-mediated regulation of emotion and motivation, and reveal knowledge gaps on potential contributions from the cholinergic system.
Collapse
Affiliation(s)
- Ping Mu
- College of Life Sciences, Ludong University, 186 Hongqi Middle Road, Yantai, Shandong, 264025, China.
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, 15219, PA, United States.
| |
Collapse
|
44
|
Wood J, LaPalombara Z, Ahmari SE. Monoamine abnormalities in the SAPAP3 knockout model of obsessive-compulsive disorder-related behaviour. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0023. [PMID: 29352023 DOI: 10.1098/rstb.2017.0023] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2017] [Indexed: 01/05/2023] Open
Abstract
Obsessive-compulsive disorder (OCD) is a leading cause of illness-related disability, but the neural mechanisms underlying OCD symptoms are unclear. One potential mechanism of OCD pathology is monoamine dysregulation. Because of the difficulty of studying monoamine signalling in patients, animal models offer a viable alternative to understanding this aspect of OCD pathophysiology. We used HPLC to characterize post-mortem monoamine levels in lateral orbitofrontal cortex (OFC), medial OFC, medial prefrontal cortex and dorsal and ventral striatum of SAPAP-3 knockout (KO) mice, a well-validated model of compulsive-like behaviours in OCD. As predicted from previous studies, excessive grooming was significantly increased in SAPAP-3 KO mice. Overall levels of the serotonin metabolite 5-hydroxyindoleacetic acid (HIAA) and the ratio of 5HIAA/serotonin (serotonin turnover) were increased in all cortical and striatal regions examined. In addition, dihydroxyphenylacetic acid/dopamine ratio was increased in lateral OFC, and HVA/dopamine ratio was increased in lateral and medial OFC. No baseline differences in serotonin or dopamine tissue content were observed. These data provide evidence of monoaminergic dysregulation in a translational model of OCD symptoms and are consistent with aberrant cortical and striatal serotonin and dopamine release/metabolism in SAPAP-3 KO mice. These results are guiding ongoing experiments using circuit and cell-type specific manipulations of dopamine and serotonin to determine the contributions of these monoaminergic systems to compulsive behaviours, and serve here as a touchstone for an expanded discussion of these techniques for precise circuit dissection.This article is part of the discussion meeting issue 'Of mice and mental health: facilitating dialogue between basic and clinical neuroscientists'.
Collapse
Affiliation(s)
- Jesse Wood
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, 450 Technology Drive, Room 227, Pittsburgh, PA 15219, USA.,Center for Neuroscience Program and Center for the Neural Basis of Cognition, University of Pittsburgh, 450 Technology Drive, Room 227, Pittsburgh, PA 15219, USA
| | - Zoe LaPalombara
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, 450 Technology Drive, Room 227, Pittsburgh, PA 15219, USA.,Center for Neuroscience Program and Center for the Neural Basis of Cognition, University of Pittsburgh, 450 Technology Drive, Room 227, Pittsburgh, PA 15219, USA
| | - Susanne E Ahmari
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, 450 Technology Drive, Room 227, Pittsburgh, PA 15219, USA .,Center for Neuroscience Program and Center for the Neural Basis of Cognition, University of Pittsburgh, 450 Technology Drive, Room 227, Pittsburgh, PA 15219, USA
| |
Collapse
|
45
|
Müller Ewald VA, De Corte BJ, Gupta SC, Lillis KV, Narayanan NS, Wemmie JA, LaLumiere RT. Attenuation of cocaine seeking in rats via enhancement of infralimbic cortical activity using stable step-function opsins. Psychopharmacology (Berl) 2019; 236:479-490. [PMID: 30003306 PMCID: PMC6330160 DOI: 10.1007/s00213-018-4964-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/28/2018] [Indexed: 12/29/2022]
Abstract
RATIONALE The infralimbic cortex (IL) and its downstream projection target the nucleus accumbens shell (NAshell) mediate the active suppression of cocaine-seeking behavior. Although an optogenetic approach would be beneficial for stimulating the IL and its efferents to study their role during reinstatement of cocaine seeking, the use of channelrhodopsin introduces significant difficulties, as optimal stimulation parameters are not known. OBJECTIVES The present experiments utilized a stable step-function opsin (SSFO) to potentiate endogenous activity in the IL and in IL terminals in the NAshell during cocaine-seeking tests to determine how these manipulations affect cocaine-seeking behaviors. METHODS Rats first underwent 6-h access cocaine self-administration followed by 21-27 days in the homecage. Rats then underwent cue-induced and cocaine-primed drug-seeking tests during which the optogenetic manipulation was given. The same rats then underwent extinction training, followed by cue-induced and cocaine-primed reinstatements. RESULTS Potentiation of endogenous IL activity did not significantly alter cue-induced or cocaine-primed drug seeking following the homecage period. However, following extinction training, enhancement of endogenous IL activity attenuated cue-induced reinstatement by 35% and cocaine-primed reinstatement by 53%. Stimulation of IL terminals in the NAshell did not consistently alter cocaine-seeking behavior. CONCLUSION These results suggest the utility of an SSFO-based approach for enhancing activity in a structure without driving specific patterns of neuronal firing. However, the utility of an SSFO-based approach for axon terminal stimulation remains unclear. Moreover, these results suggest that the ability of the IL to reduce cocaine seeking depends, at least in part, on rats first having undergone extinction training.
Collapse
Affiliation(s)
- Victória A Müller Ewald
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, 52242, USA.
- W311 Seashore Hall, Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, 52242, USA.
| | - Benjamin J De Corte
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
| | - Subhash C Gupta
- Department of Psychiatry, University of Iowa, Iowa City, IA, 52242, USA
| | - Katherine V Lillis
- W311 Seashore Hall, Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, 52242, USA
| | - Nandakumar S Narayanan
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - John A Wemmie
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, 52242, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Ryan T LaLumiere
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, 52242, USA
- W311 Seashore Hall, Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
46
|
Abstract
Over the period of decades in the mid to late twentieth century, arousal-promoting functions were attributed to neuromodulators including serotonin, hypocretin, histamine, and noradrenaline. For some time, a relatively minor role in regulating sleep and wake states was ascribed to dopamine and the dopamine-producing cells of the ventral tegmental area, despite the fact that dopaminergic signaling is a major target, if not the primary target, for wake-promoting agents. In recent years, due to observations from human genetic studies, pharmacogenetic studies in animal models, and the increasingly sophisticated methods used to manipulate the nervous systems of experimental animals, it has become clear that dopaminergic signaling is central to the regulation of arousal. This chapter reviews this central role of dopaminergic signaling, and in particular its antagonistic interaction with adenosinergic signaling, in maintaining vigilance and in the response to wake-promoting therapeutics.
Collapse
Affiliation(s)
- Jonathan P Wisor
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA.
| |
Collapse
|
47
|
Logan RW, Hasler BP, Forbes EE, Franzen PL, Torregrossa MM, Huang YH, Buysse DJ, Clark DB, McClung CA. Impact of Sleep and Circadian Rhythms on Addiction Vulnerability in Adolescents. Biol Psychiatry 2018; 83:987-996. [PMID: 29373120 PMCID: PMC5972052 DOI: 10.1016/j.biopsych.2017.11.035] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/18/2017] [Accepted: 11/22/2017] [Indexed: 12/11/2022]
Abstract
Sleep homeostasis and circadian function are important maintaining factors for optimal health and well-being. Conversely, sleep and circadian disruptions are implicated in a variety of adverse health outcomes, including substance use disorders. These risks are particularly salient during adolescence. Adolescents require 8 to 10 hours of sleep per night, although few consistently achieve these durations. A mismatch between developmental changes and social/environmental demands contributes to inadequate sleep. Homeostatic sleep drive takes longer to build, circadian rhythms naturally become delayed, and sensitivity to the phase-shifting effects of light increases, all of which lead to an evening preference (i.e., chronotype) during adolescence. In addition, school start times are often earlier in adolescence and the use of electronic devices at night increases, leading to disrupted sleep and circadian misalignment (i.e., social jet lag). Social factors (e.g., peer influence) and school demands further impact sleep and circadian rhythms. To cope with sleepiness, many teens regularly consume highly caffeinated energy drinks and other stimulants, creating further disruptions in sleep. Chronic sleep loss and circadian misalignment enhance developmental tendencies toward increased reward sensitivity and impulsivity, increasing the likelihood of engaging in risky behaviors and exacerbating the vulnerability to substance use and substance use disorders. We review the neurobiology of brain reward systems and the impact of sleep and circadian rhythms changes on addiction vulnerability in adolescence and suggest areas that warrant additional research.
Collapse
Affiliation(s)
- Ryan W Logan
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; The Jackson Laboratory, Bar Harbor, Maine
| | - Brant P Hasler
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Erika E Forbes
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Peter L Franzen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mary M Torregrossa
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yanhua H Huang
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel J Buysse
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Duncan B Clark
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Colleen A McClung
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; The Jackson Laboratory, Bar Harbor, Maine.
| |
Collapse
|
48
|
Bueno-Junior LS, Leite JP. Input Convergence, Synaptic Plasticity and Functional Coupling Across Hippocampal-Prefrontal-Thalamic Circuits. Front Neural Circuits 2018; 12:40. [PMID: 29875637 PMCID: PMC5975431 DOI: 10.3389/fncir.2018.00040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/26/2018] [Indexed: 01/19/2023] Open
Abstract
Executive functions and working memory are long known to involve the prefrontal cortex (PFC), and two PFC-projecting areas: midline/paramidline thalamus (MLT) and cornus ammonis 1 (CA1)/subiculum of the hippocampal formation (HF). An increasing number of rodent electrophysiology studies are examining these substrates together, thus providing circuit-level perspectives on input convergence, synaptic plasticity and functional coupling, as well as insights into cognition mechanisms and brain disorders. Our review article puts this literature into a method-oriented narrative. As revisited throughout the text, limbic thalamic and hippocampal afferents to the PFC gate one another’s inputs, which in turn are modulated by PFC interneurons and ascending monoaminergic projections. In addition, long-term synaptic plasticity, paired-pulse facilitation (PPF), and event-related potentials (ERP) dynamically vary across PFC-related circuits during learning paradigms and drug effects. Finally, thalamic-prefrontal loops, which have been shown to amplify both cognitive processes and limbic seizures, are also being implicated as relays in the prefrontal-hippocampal feedback, contributing to spatial navigation and decision making. Based on these issues, we conclude the review with a critical synthesis and some research directions.
Collapse
Affiliation(s)
- Lezio S Bueno-Junior
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Joao P Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
49
|
Miller OH, Bruns A, Ben Ammar I, Mueggler T, Hall BJ. Synaptic Regulation of a Thalamocortical Circuit Controls Depression-Related Behavior. Cell Rep 2018; 20:1867-1880. [PMID: 28834750 DOI: 10.1016/j.celrep.2017.08.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 07/08/2017] [Accepted: 07/26/2017] [Indexed: 01/06/2023] Open
Abstract
The NMDA receptor (NMDAR) antagonist ketamine elicits a long-lasting antidepressant response in patients with treatment-resistant depression. Understanding how antagonism of NMDARs alters synapse and circuit function is pivotal to developing circuit-based therapies for depression. Using virally induced gene deletion, ex vivo optogenetic-assisted circuit analysis, and in vivo chemogenetics and fMRI, we assessed the role of NMDARs in the medial prefrontal cortex (mPFC) in controlling depression-related behavior in mice. We demonstrate that post-developmental genetic deletion of the NMDAR subunit GluN2B from pyramidal neurons in the mPFC enhances connectivity between the mPFC and limbic thalamus, but not the ventral hippocampus, and reduces depression-like behavior. Using intersectional chemogenetics, we show that activation of this thalamocortical circuit is sufficient to elicit a decrease in despair-like behavior. Our findings reveal that GluN2B exerts input-specific control of pyramidal neuron innervation and identify a medial dorsal thalamus (MDT)→mPFC circuit that controls depression-like behavior.
Collapse
Affiliation(s)
- Oliver H Miller
- Roche Innovation Center Basel, Basel 4070, Switzerland; Tulane Brain Institute, Tulane University, New Orleans, LA 70115, USA
| | - Andreas Bruns
- Roche Innovation Center Basel, Basel 4070, Switzerland
| | - Imen Ben Ammar
- Faculté de Médecine, Université de Strasbourg, Strasbourg 67000, France
| | | | - Benjamin J Hall
- Roche Innovation Center Basel, Basel 4070, Switzerland; Tulane Brain Institute, Tulane University, New Orleans, LA 70115, USA.
| |
Collapse
|
50
|
Murillo-Rodríguez E, Di Marzo V, Machado S, Rocha NB, Veras AB, Neto GAM, Budde H, Arias-Carrión O, Arankowsky-Sandoval G. Role of N-Arachidonoyl-Serotonin (AA-5-HT) in Sleep-Wake Cycle Architecture, Sleep Homeostasis, and Neurotransmitters Regulation. Front Mol Neurosci 2017; 10:152. [PMID: 28611585 PMCID: PMC5447686 DOI: 10.3389/fnmol.2017.00152] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/05/2017] [Indexed: 12/19/2022] Open
Abstract
The endocannabinoid system comprises several molecular entities such as endogenous ligands [anandamide (AEA) and 2-arachidonoylglycerol (2-AG)], receptors (CB1 and CB2), enzymes such as [fatty acid amide hydrolase (FAHH) and monoacylglycerol lipase (MAGL)], as well as the anandamide membrane transporter. Although the role of this complex neurobiological system in the sleep–wake cycle modulation has been studied, the contribution of the blocker of FAAH/transient receptor potential cation channel subfamily V member 1 (TRPV1), N-arachidonoyl-serotonin (AA-5-HT) in sleep has not been investigated. Thus, in the present study, varying doses of AA-5-HT (5, 10, or 20 mg/Kg, i.p.) injected at the beginning of the lights-on period of rats, caused no statistical changes in sleep patterns. However, similar pharmacological treatment given to animals at the beginning of the dark period decreased wakefulness (W) and increased slow wave sleep (SWS) as well as rapid eye movement sleep (REMS). Power spectra analysis of states of vigilance showed that injection of AA-5-HT during the lights-off period diminished alpha spectrum across alertness in a dose-dependent fashion. In opposition, delta power spectra was enhanced as well as theta spectrum, during SWS and REMS, respectively. Moreover, the highest dose of AA-5-HT decreased wake-related contents of neurotransmitters such as dopamine (DA), norepinephrine (NE), epinephrine (EP), serotonin (5-HT) whereas the levels of adenosine (AD) were enhanced. In addition, the sleep-inducing properties of AA-5-HT were confirmed since this compound blocked the increase in W caused by stimulants such as cannabidiol (CBD) or modafinil (MOD) during the lights-on period. Additionally, administration of AA-5-HT also prevented the enhancement in contents of DA, NE, EP, 5-HT and AD after CBD of MOD injection. Lastly, the role of AA-5-HT in sleep homeostasis was tested in animals that received either CBD or MOD after total sleep deprivation (TSD). The injection of CBD or MOD increased alertness during sleep rebound period after TSD. However, AA-5-HT blocked this effect by allowing animals to display an enhancement in sleep across sleep rebound period. Overall, our findings provide evidence that AA-5-HT is an important modulator of sleep, sleep homeostasis and neurotransmitter contents.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac MayabMérida, Mexico.,Grupo de Investigación en Envejecimiento, División Ciencias de la Salud, Universidad Anáhuac MayabMérida, Mexico.,Grupo de Investigación Desarrollos Tecnológicos para la Salud, División de Ingeniería y Ciencias Exactas, Universidad Anáhuac MayabMérida, Mexico.,Intercontinental Neuroscience Research Group
| | - Vincenzo Di Marzo
- Intercontinental Neuroscience Research Group.,Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle RicerchePozzuoli, Italy
| | - Sergio Machado
- Intercontinental Neuroscience Research Group.,Laboratory of Panic and Respiration, Institute of Psychiatry, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,Postgraduate Program, Salgado de Oliveira UniversityRio de Janeiro, Brazil
| | - Nuno B Rocha
- Intercontinental Neuroscience Research Group.,Faculty of Health Sciences, Polytechnic Institute of PortoPorto, Portugal
| | - André B Veras
- Intercontinental Neuroscience Research Group.,Institute of Psychiatry, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,Dom Bosco Catholic UniversityRio de Janeiro, Brazil
| | - Geraldo A M Neto
- Intercontinental Neuroscience Research Group.,Laboratory of Panic and Respiration, Institute of Psychiatry, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Henning Budde
- Intercontinental Neuroscience Research Group.,Faculty of Human Sciences, Medical School HamburgHamburg, Germany.,Physical Activity, Physical Education, Health and Sport Research Centre (PAPESH), Sports Science Department, School of Science and Engineering Reykjavik UniversityReykjavik, Iceland.,Department of Health, Physical and Social Education, Lithuanian Sports UniversityKaunas, Lithuania
| | - Oscar Arias-Carrión
- Intercontinental Neuroscience Research Group.,Unidad de Trastornos del Movimiento y Sueño (TMS), Hospital General "Dr. Manuel Gea González"Ciudad de México, Mexico
| | - Gloria Arankowsky-Sandoval
- Intercontinental Neuroscience Research Group.,Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de YucatánMérida, Mexico
| |
Collapse
|