1
|
Deng Q, Li Y, Sun Z, Gao X, Zhou J, Ma G, Qu WM, Li R. Sleep disturbance in rodent models and its sex-specific implications. Neurosci Biobehav Rev 2024; 164:105810. [PMID: 39009293 DOI: 10.1016/j.neubiorev.2024.105810] [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: 12/11/2023] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Sleep disturbances, encompassing altered sleep physiology or disorders like insomnia and sleep apnea, profoundly impact physiological functions and elevate disease risk. Despite extensive research, the underlying mechanisms and sex-specific differences in sleep disorders remain elusive. While polysomnography serves as a cornerstone for human sleep studies, animal models provide invaluable insights into sleep mechanisms. However, the availability of animal models of sleep disorders is limited, with each model often representing a specific sleep issue or mechanism. Therefore, selecting appropriate animal models for sleep research is critical. Given the significant sex differences in sleep patterns and disorders, incorporating both male and female subjects in studies is essential for uncovering sex-specific mechanisms with clinical relevance. This review provides a comprehensive overview of various rodent models of sleep disturbance, including sleep deprivation, sleep fragmentation, and circadian rhythm dysfunction. We evaluate the advantages and disadvantages of each model and discuss sex differences in sleep and sleep disorders, along with potential mechanisms. We aim to advance our understanding of sleep disorders and facilitate sex-specific interventions.
Collapse
Affiliation(s)
- Qi Deng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yuhong Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xiang Gao
- Shanxi Bethune Hospital, Shanxi, China
| | | | - Guangwei Ma
- Peking University Sixth Hospital, Beijing, China
| | - Wei-Min Qu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China; Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Rena Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Rendine M, Cocci P, de Vivo L, Bellesi M, Palermo FA. Effects of Chronic Sleep Restriction on Transcriptional Sirtuin 1 Signaling Regulation in Male Mice White Adipose Tissue. Curr Issues Mol Biol 2024; 46:2144-2154. [PMID: 38534754 DOI: 10.3390/cimb46030138] [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: 02/08/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Chronic sleep restriction (CSR) is a prevalent issue in modern society that is associated with several pathological states, ranging from neuropsychiatric to metabolic diseases. Despite its known impact on metabolism, the specific effects of CSR on the molecular mechanisms involved in maintaining metabolic homeostasis at the level of white adipose tissue (WAT) remain poorly understood. Therefore, this study aimed to investigate the influence of CSR on sirtuin 1 (SIRT1) and the peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway in the WAT of young male mice. Both genes interact with specific targets involved in multiple metabolic processes, including adipocyte differentiation, browning, and lipid metabolism. The quantitative PCR (qPCR) results demonstrated a significant upregulation of SIRT-1 and some of its target genes associated with the transcriptional regulation of lipid homeostasis (i.e., PPARα, PPARγ, PGC-1α, and SREBF) and adipose tissue development (i.e., leptin, adiponectin) in CSR mice. On the contrary, DNA-binding transcription factors (i.e., CEBP-β and C-myc), which play a pivotal function during the adipogenesis process, were found to be down-regulated. Our results also suggest that the induction of SIRT1-dependent molecular pathways prevents weight gain. Overall, these findings offer new, valuable insights into the molecular adaptations of WAT to CSR, in order to support increased energy demand due to sleep loss.
Collapse
Affiliation(s)
- Marco Rendine
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133 Milan, Italy
| | - Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Luisa de Vivo
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy
| | - Michele Bellesi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1QU, UK
| | | |
Collapse
|
3
|
Moraes MM, Marques AL, Borges L, Hatanaka E, Heller D, Núñez-Espinosa C, Gonçalves DAP, Soares DD, Wanner SP, Mendes TT, Arantes RME. Sleep impairment and altered pattern of circadian biomarkers during a long-term Antarctic summer camp. Sci Rep 2023; 13:15959. [PMID: 37749123 PMCID: PMC10519969 DOI: 10.1038/s41598-023-42910-8] [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: 02/12/2023] [Accepted: 09/15/2023] [Indexed: 09/27/2023] Open
Abstract
Antarctic expeditions include isolation and exposure to cold and extreme photoperiods (with continuous natural light during summer) that may influence psychophysiological responses modulated by luminosity and sleep. We assessed changes in night sleep patterns by actigraphy, salivary biomarkers, and perceptual variables in seven participants in the following time points along a 50-day camping expedition in Antarctica (Nelson Island): Pre-Field (i.e., on the ship before camp), Field-1, Field-2, Field-3, Field-4 (from 1st to 10th, 11th to 20th, 21st to 35th and 36th to 50th days in camp, respectively), and Post-Field (on the ship after camp). We also characterized mood states, daytime sleepiness, and sleep quality by questionnaires. Staying in an Antarctic camp reduced sleep efficiency (5.2%) and increased the number of awakenings and wakefulness after sleep onset (51.8% and 67.1%, respectively). Furthermore, transient increases in time in bed (16.5%) and sleep onset latency (4.8 ± 4.0 min, from Pre- to Field-3) was observed. These changes were accompanied by an altered pattern of the emerging circadian marker β-Arrestin-1 and a trend to reduce nocturnal melatonin [57.1%; P = 0.066, with large effect size (ES) from Pre-Field to Field-2 (ES = 1.2) and Field-3 (ES = 1.2)]. All changes returned to Pre-Field values during the Post-Field. The volunteers reported sleep-related physical complaints (feeling of cold and pain, discomfort to breathe, and cough or loud snoring), excessive daytime sleepiness, and reduced vigor during the camp. Thus, a 50-day camp alters neuroendocrine regulation and induces physical discomfort, which may explain the impaired sleep pattern and the consequent daytime sleepiness and mood changes.
Collapse
Affiliation(s)
- Michele Macedo Moraes
- Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Center for Newborn Screening and Genetics Diagnosis, Faculty of Medicine, Universidade Federal de Minas Gerais (NUPAD-FM/UFMG), Belo Horizonte, MG, Brazil
| | - Alice Lamounier Marques
- Post-Graduation Program in Social Sciences in Development, Culture and Society, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brazil
| | - Leandro Borges
- Interdisciplinary Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo, SP, Brazil
| | - Elaine Hatanaka
- Interdisciplinary Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo, SP, Brazil
| | - Debora Heller
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Post-Graduate Studies in Dentistry, Universidade Cruzeiro Do Sul, São Paulo, SP, Brazil
- Department of Periodontology, School of Dentistry, UT Health San Antonio, San Antonio, TX, USA
| | - Cristian Núñez-Espinosa
- Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
- Centro Asistencial Docente y de Investigación, Universidad de Magallanes, Punta Arenas, Chile
- Interuniversity Center for Healthy Aging, Chilecito, Chile
| | - Dawit Albieiro Pinheiro Gonçalves
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danusa Dias Soares
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Samuel Penna Wanner
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thiago Teixeira Mendes
- Department of Physical Education, Faculty of Education, Universidade Federal da Bahia, Salvador, BA, Brazil
| | - Rosa Maria Esteves Arantes
- Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
- Center for Newborn Screening and Genetics Diagnosis, Faculty of Medicine, Universidade Federal de Minas Gerais (NUPAD-FM/UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
4
|
Cheng Y, Dempsey RE, Roodsari SK, Shuboni-Mulligan DD, George O, Sanford LD, Guo ML. Cocaine Regulates NLRP3 Inflammasome Activity and CRF Signaling in a Region- and Sex-Dependent Manner in Rat Brain. Biomedicines 2023; 11:1800. [PMID: 37509440 PMCID: PMC10376186 DOI: 10.3390/biomedicines11071800] [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: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Cocaine, one of the most abused drugs worldwide, is capable of activating microglia in vitro and in vivo. Several neuroimmune pathways have been suggested to play roles in cocaine-mediated microglial activation. Previous work showed that cocaine activates microglia in a region-specific manner in the brains of self-administered mice. To further characterize the effects of cocaine on microglia and neuroimmune signaling in vivo, we utilized the brains from both sexes of outbred rats with cocaine self-administration to explore the activation status of microglia, NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activity, corticotropin-releasing factor (CRF) signaling, and NF-κB levels in the striatum and hippocampus (HP). Age-matched rats of the same sex (drug naïve) served as controls. Our results showed that cocaine increased neuroinflammation in the striatum and HP of both sexes with a relatively higher increases in male brains. In the striatum, cocaine upregulated NLRP3 inflammasome activity and CRF levels in males but not in females. In contrast, cocaine increased NLRP3 inflammasome activity in the HP of females but not in males, and no effects on CRF signaling were observed in this region of either sex. Interestingly, cocaine increased NF-κB levels in the striatum and HP with no sex difference. Taken together, our results provide evidence that cocaine can exert region- and sex-specific differences in neuroimmune signaling in the brain. Targeting neuroimmune signaling has been suggested as possible treatment for cocaine use disorders (CUDs). Our current results indicate that sex should be taken into consideration when determining the efficacy of these new therapeutic approaches.
Collapse
Affiliation(s)
- Yan Cheng
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Rachael Elizabeth Dempsey
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Soheil Kazemi Roodsari
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Dorela D Shuboni-Mulligan
- Sleep Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Olivier George
- Department of Psychiatry, School of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Larry D Sanford
- Sleep Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ming-Lei Guo
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
5
|
Sleep-Disturbance-Induced Microglial Activation Involves CRH-Mediated Galectin 3 and Autophagy Dysregulation. Cells 2022; 12:cells12010160. [PMID: 36611953 PMCID: PMC9818437 DOI: 10.3390/cells12010160] [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: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Chronic sleep disturbances (CSDs) including insomnia, insufficient sleep time, and poor sleep quality are major public health concerns around the world, especially in developed countries. CSDs are major health risk factors linked to multiple neurodegenerative and neuropsychological diseases. It has been suggested that CSDs could activate microglia (Mg) leading to increased neuroinflammation levels, which ultimately lead to neuronal dysfunction. However, the detailed mechanisms underlying CSD-mediated microglial activation remain mostly unexplored. In this study, we used mice with three-weeks of sleep fragmentation (SF) to explore the underlying pathways responsible for Mg activation. Our results revealed that SF activates Mg in the hippocampus (HP) but not in the striatum and prefrontal cortex (PFc). SF increased the levels of corticotropin-releasing hormone (CRH) in the HP. In vitro mechanism studies revealed that CRH activation of Mg involves galectin 3 (Gal3) upregulation and autophagy dysregulation. CRH could disrupt lysosome membrane integrity resulting in lysosomal cathepsins leakage. CRHR2 blockage mitigated CRH-mediated effects on microglia in vitro. SF mice also show increased Gal3 levels and autophagy dysregulation in the HP compared to controls. Taken together, our results show that SF-mediated hippocampal Mg activation involves CRH mediated galectin 3 and autophagy dysregulation. These findings suggest that targeting the hippocampal CRH system might be a novel therapeutic approach to ameliorate CSD-mediated neuroinflammation and neurodegenerative diseases.
Collapse
|
6
|
Li MT, Robinson CL, Ruan QZ, Surapaneni S, Southerland W. The Influence of Sleep Disturbance on Chronic Pain. Curr Pain Headache Rep 2022; 26:795-804. [PMID: 36190680 DOI: 10.1007/s11916-022-01074-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to present an overview of common sleep disturbance pathologies and their impact on chronic pain, while examining various factors that are implicit in the relationship between sleep disturbance and chronic pain, including neurobiochemistry, anatomy, and systemic mediators, and reviewing recent and landmark literature. RECENT FINDINGS Earlier literature reviews and studies have introduced the bidirectional relationship between sleep disturbance and chronic pain; that is, impaired sleep may worsen chronic pain, and chronic pain causes sleep disturbance. However, more recent reviews and studies seem to show a more associative, rather than causative relationship. There have been recent studies that attempt to determine mechanisms that link sleep disturbance and chronic pain; the results of these studies were more varied, ultimately concluding that there may be a separate, yet-to-be discovered mechanism that shows the causative relationship between sleep disturbance and pain. There are several neurotransmitters that are involved in the mediation of chronic pain and sleep disturbance as separate entities, and some studies have shown that there may be mechanisms that govern both chronic pain and sleep disturbance as a single unit. Other neuroendocrine substances also serve to mediate chronic pain and sleep disturbance. All these substances are found to be associated with various sleep disorders and are also associated with chronic pain symptoms as well. Inflammation plays a role in chronic pain and sleep disturbance, with an increase in inflammatory substances and mediators associated with an increase or worsening in chronic pain symptoms and sleep disorders. The HPA axis plays a role in chronic pain and sleep disorders, influencing pain and sleep pathways through stress response, inflammation, and maintenance of homeostasis. There are several variables that influence both chronic pain and sleep disturbance, and more research into these variables may further our understanding into the complex pathways governing the influence of sleep disturbance on pain, and ultimately to improve treatment for this issue.
Collapse
Affiliation(s)
- Michael T Li
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Christopher Louis Robinson
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Qing Zhao Ruan
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sindhuja Surapaneni
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Warren Southerland
- Department of Anesthesia, The Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Monzon AD, Patton SR, Koren D. Childhood diabetes and sleep. Pediatr Pulmonol 2022; 57:1835-1850. [PMID: 34506691 DOI: 10.1002/ppul.25651] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/18/2021] [Accepted: 08/26/2021] [Indexed: 12/18/2022]
Abstract
Sleep modulates glucose metabolism, both in healthy states and in disease. Alterations in sleep duration (insufficient and excessive) and obstructive sleep apnea may have reciprocal ties with obesity, insulin resistance and Type 2 diabetes, as demonstrated by emerging evidence in children and adolescents. Type 1 diabetes is also associated with sleep disturbances due to the influence of wide glycemic fluctuations upon sleep architecture, the need to treat nocturnal hypoglycemia, and the need for glucose monitoring and insulin delivery technologies. In this article, we provide an extensive and critical review on published pediatric literature regarding these topics, reviewing both epidemiologic and qualitative data, and provide an overview of the pathophysiology linking sleep with disorders of glucose homeostasis.
Collapse
Affiliation(s)
- Alexandra D Monzon
- Department of Psychology and Applied Behavioral Science, Clinical Child Psychology Program, University of Kansas, Lawrence, Kansas, USA
| | - Susana R Patton
- Department of Biomedical Research, Center for Healthcare Delivery Science, Nemours Children's Health System, Jacksonville, Florida, USA
| | - Dorit Koren
- Department of Pediatrics, Pediatric Endocrinology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
8
|
The interplay between Sleep and Gut Microbiota. Brain Res Bull 2022; 180:131-146. [DOI: 10.1016/j.brainresbull.2021.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023]
|
9
|
Berdina O, Madaeva I, Bolshakova S, Sholokhov L, Rychkova L. Circadian Rhythm of Salivary Cortisol in Obese Adolescents With and Without Apnea: A Pilot Study. Front Pediatr 2022; 10:795635. [PMID: 35558378 PMCID: PMC9090445 DOI: 10.3389/fped.2022.795635] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/23/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Obstructive sleep apnea (OSA) and obesity are associated with stress system activation involving the hypothalamic-pituitary-adrenal (HPA) axis in adults, but these effects in childhood and adolescence remain unclear. We examined diurnal salivary cortisol as a measurement of the HPA axis function in obese adolescents with and without OSA and the relationships between cortisol levels, body weight, and parameters of polysomnography (PSG). METHODS After PSG, saliva samples were collected from obese participants (with and without OSA) and lean participants four times over a 24-h period, namely, at 7:00 h (m-sCort), 13:00 h (a-sCort), 19:00 h (e-sCort), and 23:00 h (n-sCort). An enzyme-linked immunosorbent assay (ELISA) was used to measure salivary cortisol levels. The mean values of cortisol levels and fixed-time point diurnal cortisol slope (DCS) were calculated and compared among the three study groups. Correlations between parameters were analyzed using Spearman's correlation coefficients. RESULTS Obese OSA participants had significantly higher e-sCort and n-sCort levels than both obese non-OSA participants and lean controls. However, m-sCort and a-sCort in these patients had a pronounced upward trend. M-sCort was significantly correlated with both the lowest oxygen saturation (SpO2) and time with SpO2 <90%. Moreover, in the obese OSA group, DCS was significantly flatter than in the other two groups. The a-sCort in obese non-OSA participants was significantly higher than that in the lean control group and, surprisingly, was positively correlated with the apnea/hypopnea index. Additionally, m-sCort was related to body weight. CONCLUSION This study provided further evidence for alterations in diurnal cortisol production in obese adolescents, which may indicate a chronically stressed HPA axis. However, there were significant differences in salivary cortisol parameters between participants with and without OSA. Furthermore, patients with OSA had more associations between time-point cortisol levels and OSA-related indices. Nonetheless, this research is a pilot study, and further investigations are necessary.
Collapse
Affiliation(s)
- Olga Berdina
- Laboratory of Somnology and Neurophysiology, Department of Personalized and Preventive Medicine, Scientific Centre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - Irina Madaeva
- Laboratory of Somnology and Neurophysiology, Department of Personalized and Preventive Medicine, Scientific Centre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - Svetlana Bolshakova
- Laboratory of Somnology and Neurophysiology, Department of Personalized and Preventive Medicine, Scientific Centre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - Leonid Sholokhov
- Laboratory of Physiology and Pathology of Endocrine System, Department of Reproductive Health Care, Scientific Centre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| | - Liubov Rychkova
- Scientific Centre for Family Health and Human Reproduction Problems, Irkutsk, Russia
| |
Collapse
|
10
|
Open Hip Abductor Repair Hitting the Sack-Abductor Tendon Repair Significantly Improves Sleep Quality. J Clin Med 2021; 10:jcm10215164. [PMID: 34768684 PMCID: PMC8584347 DOI: 10.3390/jcm10215164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/25/2021] [Accepted: 11/03/2021] [Indexed: 11/17/2022] Open
Abstract
PURPOSE To (1) describe the prevalence of abnormal sleep quality in patients with hip abductor tears (HAT), to (2) determine whether sleep quality improves after open HAT repair, and to (3) to report clinical short-term outcomes in patients undergoing open HAT repair. METHODS The data of 28 patients (29 hips) who underwant open HAT repair were prospectively analyzed at midterm follow-up. The Pittsburgh Sleep Quality Index (PSQI), modified Harris Hip Score (mHHS), the University of California, Los Angeles activity scale (UCLA), and Visual Analog Scale (VAS) for pain were determined via questionnaire. Paired t-tests were applied to compare preoperative and post-operative Patient-reported Outcome Measures (PROMs). Logistic regression was performed to determine the association between PSQI improvement achievement and demographic variables (laterality, sex, age, body-mass-index (BMI), and preoperative mHHS). The minimal clinically important difference (MCID) was calculated for the mHHS. RESULTS A total of 28 patients were included. Four patients (14.3%) suffered post-operative complications after open HAT repair. The predominance of patients was female (77.4%), with a mean age of 60 ± 13 years. The average follow-up was 30.35 ± 16.62 months. Preoperatively, 27 (96.4%) patients experienced poor sleep quality (PSQI > 5); at follow-up, 7 (25%) patients experienced poor sleep quality. Univariate logistical regression analysis demonstrated no significant association between preoperative demographic data and achieving postoperative PSQI < 5. The MCID of mHHS was calculated to be 12.5. Overall, 90% of patients achieved MCID for mHHS. CONCLUSION Preoperative sleep quality was impaired in 96.4% of HAT patients (PSQI > 5). However, these patients showed an improvement in sleep disturbances after open HAT repair in the early postoperative period. Ninety percent of patients showed significant improvements in mHHS and achieved the corresponding MCID. LEVEL OF EVIDENCE Case series; Level IV.
Collapse
|
11
|
Cheng Y, Kim WK, Wellman LL, Sanford LD, Guo ML. Short-Term Sleep Fragmentation Dysregulates Autophagy in a Brain Region-Specific Manner. Life (Basel) 2021; 11:life11101098. [PMID: 34685469 PMCID: PMC8538758 DOI: 10.3390/life11101098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 01/17/2023] Open
Abstract
In this study, we investigated autophagy, glial activation status, and corticotropin releasing factor (CRF) signaling in the brains of mice after 5 days of sleep fragmentation (SF). Three different brain regions including the striatum, hippocampus, and frontal cortex were selected for examination based on roles in sleep regulation and sensitivity to sleep disruption. For autophagy, we monitored the levels of various autophagic induction markers including beclin1, LC3II, and p62 as well as the levels of lysosomal associated membrane protein 1 and 2 (LAMP1/2) and the transcription factor EB (TFEB) which are critical for lysosome function and autophagy maturation stage. For the status of microglia and astrocytes, we determined the levels of Iba1 and GFAP in these brain regions. We also measured the levels of CRF and its cognate receptors 1 and 2 (CRFR1/2). Our results showed that 5 days of SF dysregulated autophagy in the striatum and hippocampus but not in the frontal cortex. Additionally, 5 days of SF activated microglia in the striatum but not in the hippocampus or frontal cortex. In the striatum, CRFR2 but not CRFR1 was significantly increased in SF-experienced mice. CRF did not alter its mRNA levels in any of the three brain regions assessed. Our findings revealed that autophagy processes are sensitive to short-term SF in a region-specific manner and suggest that autophagy dysregulation may be a primary initiator for brain changes and functional impairments in the context of sleep disturbances and disorders.
Collapse
Affiliation(s)
- Yan Cheng
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Drug Addiction Laboratory, Department Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Woong-Ki Kim
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Laurie L. Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Larry D. Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ming-Lei Guo
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Drug Addiction Laboratory, Department Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Correspondence: ; Tel.: +1-757-446-5891
| |
Collapse
|
12
|
Bowers SJ, Lambert S, He S, Lowry CA, Fleshner M, Wright KP, Turek FW, Vitaterna MH. Immunization with a heat-killed bacterium, Mycobacterium vaccae NCTC 11659, prevents the development of cortical hyperarousal and a PTSD-like sleep phenotype after sleep disruption and acute stress in mice. Sleep 2021; 44:6025170. [PMID: 33283862 DOI: 10.1093/sleep/zsaa271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/20/2020] [Indexed: 12/27/2022] Open
Abstract
STUDY OBJECTIVES Sleep deprivation induces systemic inflammation that may contribute to stress vulnerability and other pathologies. We tested the hypothesis that immunization with heat-killed Mycobacterium vaccae NCTC 11659 (MV), an environmental bacterium with immunoregulatory and anti-inflammatory properties, prevents the negative impacts of 5 days of sleep disruption on stress-induced changes in sleep, behavior, and physiology in mice. METHODS In a 2 × 2 × 2 experimental design, male C57BL/6N mice were given injections of either MV or vehicle on days -17, -10, and -3. On days 1-5, mice were exposed to intermittent sleep disruption, whereby sleep was disrupted for 20 h per day. Immediately following sleep disruption, mice were exposed to 1-h social defeat stress or novel cage (control) conditions. Object location memory (OLM) testing was conducted 24 h after social defeat, and tissues were collected 6 days later to measure inflammatory markers. Sleep was recorded using electroencephalography (EEG) and electromyography (EMG) throughout the experiment. RESULTS In vehicle-treated mice, only the combination of sleep disruption followed by social defeat (double hit): (1) increased brief arousals and NREM beta (15-30 Hz) EEG power in sleep immediately post-social defeat compared to baseline; (2) induced an increase in the proportion of rapid-eye-movement (REM) sleep and number of state shifts for at least 5 days post-social defeat; and (3) induced hyperlocomotion and lack of habituation in the OLM task. Immunization with MV prevented most of these sleep and behavioral changes. CONCLUSIONS Immunization with MV ameliorates a stress-induced sleep and behavioral phenotype that shares features with human posttraumatic stress disorder.
Collapse
Affiliation(s)
- Samuel J Bowers
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL.,Department of Neurobiology, Northwestern University, Evanston, IL
| | - Sophie Lambert
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - Shannon He
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL.,Department of Neurobiology, Northwestern University, Evanston, IL
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Kenneth P Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO.,Sleep and Chronobiology Laboratory, University of Colorado Boulder, Boulder, CO
| | - Fred W Turek
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL.,Department of Neurobiology, Northwestern University, Evanston, IL.,The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Martha H Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL.,Department of Neurobiology, Northwestern University, Evanston, IL
| |
Collapse
|
13
|
Choshen-Hillel S, Ishqer A, Mahameed F, Reiter J, Gozal D, Gileles-Hillel A, Berger I. Acute and chronic sleep deprivation in residents: Cognition and stress biomarkers. MEDICAL EDUCATION 2021; 55:174-184. [PMID: 32697336 PMCID: PMC7854866 DOI: 10.1111/medu.14296] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/29/2020] [Accepted: 07/16/2020] [Indexed: 06/10/2023]
Abstract
OBJECTIVES Insufficient sleep affects circadian hormonal profiles and inflammatory markers and may modulate attention, executive functioning and decision-making. Medical professionals and specifically resident physicians, who are involved in long-term nightshift schedules during their post-graduate training, are prone to acute and chronic sleep deprivation and disruption, putting them at risk for making medical errors. The aim of the study was to evaluate the impact of chronic and acute-on-chronic sleep deprivation and disruption among residents on selected physiological and cognitive measures. METHODS Thirty-three medical and surgical residents were evaluated twice - at baseline and after a 26-hour shift. Eighteen young attending physicians who did not engage in nightshift schedules served as controls and were evaluated once. Measures included morning cortisol and high-sensitivity C-reactive protein (hs-CRP), computerised tests of attention and behaviour, the Behaviour Rating Inventory of Executive Function, a risk-taking questionnaire and the Pittsburgh Sleep Quality Index. RESULTS Residents, but not attendings, reported chronic sleep disruption and deprivation. Residents at baseline exhibited reduced morning cortisol levels and elevated hs-CRP levels, compared to attendings. Residents at baseline had impaired global executive function compared to attendings. A nightshift with acute sleep deprivation further reduced residents' executive function. Residents at baseline and after a nightshift demonstrated increased impulsivity and slower processing time than attendings. Residents and attendings did not differ in risk-taking tendencies which were assessed in a separate cohort. CONCLUSIONS In a real-life setting, resident physicians exhibit increased low-grade systemic inflammation (hs-CRP) and impaired HPA-axis function. Their chronic sleep curtailment is associated with greater impulsivity, slower cognitive processing, and impaired executive function. Future research is warranted to understand how improving working schedule by increasing sleep duration may minimise the short-term and potential long-term risks to physicians in training.
Collapse
Affiliation(s)
- Shoham Choshen-Hillel
- School of Business Administration and the Federmann Center for the Study of Rationality, Hebrew University of Jerusalem
| | - Ahmad Ishqer
- Pediatric Pulmonary & Sleep Unit, Hadassah-Hebrew University Medical Center
| | - Fadi Mahameed
- Pediatric Pulmonary & Sleep Unit, Hadassah-Hebrew University Medical Center
| | - Joel Reiter
- Pediatric Pulmonary & Sleep Unit, Hadassah-Hebrew University Medical Center
| | - David Gozal
- Department of Child Health, MU Women’s and Children’s Hospital, University of Missouri School of Medicine
| | - Alex Gileles-Hillel
- Pediatric Pulmonary & Sleep Unit, Hadassah-Hebrew University Medical Center
- The Wohl Institute for Translational Medicine, Hadassah-Hebrew University Medical Center
| | - Itai Berger
- Pediatric Neurology; Department of Pediatrics, Assuta-Ashdod University Medical Center
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva
| |
Collapse
|
14
|
Nollet M, Wisden W, Franks NP. Sleep deprivation and stress: a reciprocal relationship. Interface Focus 2020; 10:20190092. [PMID: 32382403 PMCID: PMC7202382 DOI: 10.1098/rsfs.2019.0092] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2020] [Indexed: 12/19/2022] Open
Abstract
Sleep is highly conserved across evolution, suggesting vital biological functions that are yet to be fully understood. Animals and humans experiencing partial sleep restriction usually exhibit detrimental physiological responses, while total and prolonged sleep loss could lead to death. The perturbation of sleep homeostasis is usually accompanied by an increase in hypothalamic–pituitary–adrenal (HPA) axis activity, leading to a rise in circulating levels of stress hormones (e.g. cortisol in humans, corticosterone in rodents). Such hormones follow a circadian release pattern under undisturbed conditions and participate in the regulation of sleep. The investigation of the consequences of sleep deprivation, from molecular changes to behavioural alterations, has been used to study the fundamental functions of sleep. However, the reciprocal relationship between sleep and the activity of the HPA axis is problematic when investigating sleep using traditional sleep-deprivation protocols that can induce stress per se. This is especially true in studies using rodents in which sleep deprivation is achieved by exogenous, and potentially stressful, sensory–motor stimulations that can undoubtedly confuse their conclusions. While more research is needed to explore the mechanisms underlying sleep loss and health, avoiding stress as a confounding factor in sleep-deprivation studies is therefore crucial. This review examines the evidence of the intricate links between sleep and stress in the context of experimental sleep deprivation, and proposes a more sophisticated research framework for sleep-deprivation procedures that could benefit from recent progress in biotechnological tools for precise neuromodulation, such as chemogenetics and optogenetics, as well as improved automated real-time sleep-scoring algorithms.
Collapse
Affiliation(s)
- Mathieu Nollet
- Department of Life Sciences, Imperial College London, London, UK.,UK Dementia Research Institute at Imperial College London, London, UK
| | - William Wisden
- Department of Life Sciences, Imperial College London, London, UK.,UK Dementia Research Institute at Imperial College London, London, UK.,Centre for Neurotechnology, Imperial College London, London, UK
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, London, UK.,UK Dementia Research Institute at Imperial College London, London, UK.,Centre for Neurotechnology, Imperial College London, London, UK
| |
Collapse
|
15
|
Reddy N, Martinez JR, Mulligan E, Nakonezny P, Wells J. Sleep quality and nocturnal pain in patients with femoroacetabular impingement and acetabular dysplasia. BMC Musculoskelet Disord 2020; 21:134. [PMID: 32111218 PMCID: PMC7049208 DOI: 10.1186/s12891-020-3151-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Background Femoroacetabular impingement (FAI) syndrome and acetabular dysplasia (AD) are common pathologies that lead to pain in the young adult hip. Nocturnal pain in these patients is often reported, yet little is known regarding the effect of these hip pathologies on overall sleep quality. The purpose of this study was to evaluate sleep quality in patients with AD and FAI syndrome. Methods This cross-sectional study consisted of 115 patients who complained of hip pain secondary to either FAI syndrome or AD. One hundred fifteen patients with hip pain secondary to FAI syndrome and AD were assessed using the Hip Outcome Score (HOS), Modified Harris Hip Score (mHHS), and then Hip disability and Osteoarthritis Outcome Score (HOOS). Sleep quality was assessed using the Pittsburgh Sleep Quality Index (PSQI). Multiple linear regression, with adaptive LASSO variable selection, was used to assess factors associated with sleep quality. Results Of the 115 patients, 62 had a diagnosis of FAI syndrome and 53 with AD. The mean age was 34.55 ± 11.66 (age range: 14 to 58 years), 76.52% had an ASA classification of 1 (ASA range: 1 to 3), and all Tonnis grades were either 0 or 1. The mean PSQI global score for all patients was 8.46 ± 4.35 (PSQI range: 0 to 21), indicating poor sleep quality. The adaptive LASSO-penalized least squares multiple linear regression revealed that HOOS Pain, SF-12 Role Emotional, and SF-12 Mental Health significantly predicted Sleep Quality (Adjusted R2 = 0.4041). Sleep quality improved as pain, emotional problems, and mental health improved. Conclusion Patients with symptomatic FAI syndrome and AD have poor sleep quality. Worsening pain from a patient’s hip pathology is associated with poor sleep, even prior to the onset of osteoarthrosis of the hip. Patients presenting with hip pain from FAI syndrome and AD should be screened for sleep disturbance and may benefit from a multidisciplinary treatment approach.
Collapse
Affiliation(s)
- Nisha Reddy
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Rd 1st floor, Dallas, TX, 75390, USA
| | - J Riley Martinez
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Rd 1st floor, Dallas, TX, 75390, USA
| | - Edward Mulligan
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Rd 1st floor, Dallas, TX, 75390, USA
| | - Paul Nakonezny
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Rd 1st floor, Dallas, TX, 75390, USA.,Department of Population and Data Sciences (Division of Biostatistics), University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Joel Wells
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Rd 1st floor, Dallas, TX, 75390, USA.
| |
Collapse
|
16
|
Bowers SJ, Vargas F, González A, He S, Jiang P, Dorrestein PC, Knight R, Wright KP, Lowry CA, Fleshner M, Vitaterna MH, Turek FW. Repeated sleep disruption in mice leads to persistent shifts in the fecal microbiome and metabolome. PLoS One 2020; 15:e0229001. [PMID: 32078624 PMCID: PMC7032712 DOI: 10.1371/journal.pone.0229001] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/27/2020] [Indexed: 02/07/2023] Open
Abstract
It has been established in recent years that the gut microbiome plays a role in health and disease, potentially via alterations in metabolites that influence host physiology. Although sleep disruption and gut dysbiosis have been associated with many of the same diseases, studies investigating the gut microbiome in the context of sleep disruption have yielded inconsistent results, and have not assessed the fecal metabolome. We exposed mice to five days of sleep disruption followed by four days of ad libitum recovery sleep, and assessed the fecal microbiome and fecal metabolome at multiple timepoints using 16S rRNA gene amplicons and untargeted LC-MS/MS mass spectrometry. We found global shifts in both the microbiome and metabolome in the sleep-disrupted group on the second day of recovery sleep, when most sleep parameters had recovered to baseline levels. We observed an increase in the Firmicutes:Bacteroidetes ratio, along with decreases in the genus Lactobacillus, phylum Actinobacteria, and genus Bifidobacterium in sleep-disrupted mice compared to control mice. The latter two taxa remained low at the fourth day post-sleep disruption. We also identified multiple classes of fecal metabolites that were differentially abundant in sleep-disrupted mice, some of which are physiologically relevant and commonly influenced by the microbiome. This included bile acids, and inference of microbial functional gene content suggested reduced levels of the microbial bile salt hydrolase gene in sleep-disrupted mice. Overall, this study adds to the evidence base linking disrupted sleep to the gut microbiome and expands it to the fecal metabolome, identifying sleep disruption-sensitive bacterial taxa and classes of metabolites that may serve as therapeutic targets to improve health after poor sleep.
Collapse
Affiliation(s)
- Samuel J. Bowers
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| | - Fernando Vargas
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Antonio González
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Shannon He
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Peng Jiang
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Pieter C. Dorrestein
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, United States of America
| | - Rob Knight
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, United States of America
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
| | - Kenneth P. Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
- Sleep and Chronobiology Laboratory, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Martha H. Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Fred W. Turek
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
17
|
Frau R, Traccis F, Bortolato M. Neurobehavioural complications of sleep deprivation: Shedding light on the emerging role of neuroactive steroids. J Neuroendocrinol 2020; 32:e12792. [PMID: 31505075 PMCID: PMC6982588 DOI: 10.1111/jne.12792] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/06/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
Sleep deprivation (SD) is associated with a broad spectrum of cognitive and behavioural complications, including emotional lability and enhanced stress reactivity, as well as deficits in executive functions, decision making and impulse control. These impairments, which have profound negative consequences on the health and productivity of many individuals, reflect alterations of the prefrontal cortex (PFC) and its connectivity with subcortical regions. However, the molecular underpinnings of these alterations remain elusive. Our group and others have begun examining how the neurobehavioural outcomes of SD may be influenced by neuroactive steroids, a family of molecules deeply implicated in sleep regulation and the stress response. These studies have revealed that, similar to other stressors, acute SD leads to increased synthesis of the neurosteroid allopregnanolone in the PFC. Whereas this up-regulation is likely aimed at counterbalancing the detrimental impact of oxidative stress induced by SD, the increase in prefrontal allopregnanolone levels contributes to deficits in sensorimotor gating and impulse control, signalling a functional impairment of PFC. This scenario suggests that the synthesis of neuroactive steroids during acute SD may be enacted as a neuroprotective response in the PFC; however, such compensation may in turn set off neurobehavioural complications by interfering with the corticolimbic connections responsible for executive functions and emotional regulation.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
- National Institute of Neuroscience (INN), University of Cagliari, Monserrato (CA), Italy
| | - Francesco Traccis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA
| |
Collapse
|
18
|
Duque-Díaz E, Alvarez-Ojeda O, Coveñas R. Enkephalins and ACTH in the mammalian nervous system. VITAMINS AND HORMONES 2019; 111:147-193. [PMID: 31421699 DOI: 10.1016/bs.vh.2019.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The pentapeptides methionine-enkephalin and leucine-enkephalin belong to the opioid family of peptides, and the non-opiate peptide adrenocorticotropin hormone (ACTH) to the melanocortin peptide family. Enkephalins/ACTH are derived from pro-enkephalin, pro-dynorphin or pro-opiomelanocortin precursors and, via opioid and melanocortin receptors, are responsible for many biological activities. Enkephalins exhibit the highest affinity for the δ receptor, followed by the μ and κ receptors, whereas ACTH binds to the five subtypes of melanocortin receptor, and is the only member of the melanocortin family of peptides that binds to the melanocortin-receptor 2 (ACTH receptor). Enkephalins/ACTH and their receptors exhibit a widespread anatomical distribution. Enkephalins are involved in analgesia, angiogenesis, blood pressure, embryonic development, emotional behavior, feeding, hypoxia, limbic system modulation, neuroprotection, peristalsis, and wound repair; as well as in hepatoprotective, motor, neuroendocrine and respiratory mechanisms. ACTH plays a role in acetylcholine release, aggressive behavior, blood pressure, bone maintenance, hyperalgesia, feeding, fever, grooming, learning, lipolysis, memory, nerve injury repair, neuroprotection, sexual behavior, sleep, social behavior, tissue growth and stimulates the synthesis and secretion of glucocorticoids. Enkephalins/ACTH are also involved in many pathologies. Enkephalins are implicated in alcoholism, cancer, colitis, depression, heart failure, Huntington's disease, influenza A virus infection, ischemia, multiple sclerosis, and stress. ACTH plays a role in Addison's disease, alcoholism, cancer, Cushing's disease, dermatitis, encephalitis, epilepsy, Graves' disease, Guillain-Barré syndrome, multiple sclerosis, podocytopathies, and stress. In this review, we provide an updated description of the enkephalinergic and ACTH systems.
Collapse
Affiliation(s)
- Ewing Duque-Díaz
- Universidad de Santander UDES, Laboratory of Neurosciences, School of Medicine, Bucaramanga, Colombia.
| | - Olga Alvarez-Ojeda
- Universidad Industrial de Santander, Department of Pathology, School of Medicine, Bucaramanga, Colombia
| | - Rafael Coveñas
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| |
Collapse
|
19
|
Tapp ZM, Godbout JP, Kokiko-Cochran ON. A Tilted Axis: Maladaptive Inflammation and HPA Axis Dysfunction Contribute to Consequences of TBI. Front Neurol 2019; 10:345. [PMID: 31068886 PMCID: PMC6491704 DOI: 10.3389/fneur.2019.00345] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 03/20/2019] [Indexed: 12/16/2022] Open
Abstract
Each year approximately 1.7 million people sustain a traumatic brain injury (TBI) in the US alone. Associated with these head injuries is a high prevalence of neuropsychiatric symptoms including irritability, depression, and anxiety. Neuroinflammation, due in part to microglia, can worsen or even cause neuropsychiatric disorders after TBI. For example, mounting evidence demonstrates that microglia become “primed” or hyper-reactive with an exaggerated pro-inflammatory phenotype following multiple immune challenges. Microglial priming occurs after experimental TBI and correlates with the emergence of depressive-like behavior as well as cognitive dysfunction. Critically, immune challenges are various and include illness, aging, and stress. The collective influence of any combination of these immune challenges shapes the neuroimmune environment and the response to TBI. For example, stress reliably induces inflammation and could therefore be a gateway to altered neuropathology and behavioral decline following TBI. Given the increasing incidence of stress-related psychiatric disorders after TBI, the degree in which stress affects outcome is of particular interest. This review aims to highlight the role of the hypothalamic-pituitary-adrenal (HPA) axis as a key mediator of stress-immune pathway communication following TBI. We will first describe maladaptive neuroinflammation after TBI and how stress contributes to inflammation through both anti- and pro-inflammatory mechanisms. Clinical and experimental data describing HPA-axis dysfunction and consequences of altered stress responses after TBI will be discussed. Lastly, we will review common stress models used after TBI that could better elucidate the relationship between HPA axis dysfunction and maladaptive inflammation following TBI. Together, the studies described in this review suggest that HPA axis dysfunction after brain injury is prevalent and contributes to the dynamic nature of the neuroinflammatory response to brain injury. Experimental stressors that directly engage the HPA axis represent important areas for future research to better define the role of stress-immune pathways in mediating outcome following TBI.
Collapse
Affiliation(s)
- Zoe M Tapp
- Department of Neuroscience, Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Jonathan P Godbout
- Department of Neuroscience, Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
20
|
Kunze KN, Leong NL, Beck EC, Bush-Joseph CA, Nho SJ. Hip Arthroscopy for Femoroacetabular Impingement Improves Sleep Quality Postoperatively. Arthroscopy 2019; 35:461-469. [PMID: 30612761 DOI: 10.1016/j.arthro.2018.09.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 09/18/2018] [Accepted: 09/26/2018] [Indexed: 02/02/2023]
Abstract
PURPOSE To describe the prevalence of abnormal sleep quality in patients with femoroacetabular impingement syndrome and to determine whether arthroscopic hip preservation surgery with cam/pincer correction, labral preservation, and capsular plication can improve sleep quality postoperatively. METHODS All patients undergoing primary hip arthroscopy for cam/pincer correction who failed nonoperative management between March 1, 2017, and July 1, 2017, were administered a validated sleep quality questionnaire-the Pittsburgh Sleep Quality Index (PSQI)-preoperatively and at 3, 6, 12, and 24 weeks postoperatively. Exclusion criteria included patients undergoing revision arthroscopy, gluteus medius repair, or a contralateral procedure during the follow-up period and those with known sleep disorders. A global (total) PSQI score >5 indicates poor sleep quality. The Hip Outcome Score-Activities of Daily Living, Hip Outcome Score-Sports Specific Subscale, modified Harris Hip Score, and International Hip Outcome Tool-12 were used to assess functional outcomes. A repeated measures analysis of variance with post hoc Greenhouse-Geisser and Bonferroni corrections was conducted to determine statistically significant changes in sleep patterns. RESULTS A total of 52 patients (94.6%) were included in the final analysis. The mean (± standard error) patient age was 37.8 ± 1.9 years, and body mass index was 27.6 ± 0.7. Preoperatively, 49 (94.2%) of patients experienced poor sleep quality, defined as a global PSQI score >5, with a mean PSQI score of 9.8 ± 0.6. At 24 weeks postoperatively, 10 (21.7%) of patients experienced poor sleep quality with a mean PSQI score of 2.2 ± 0.2. All patients had significant improvements in all hip outcome instruments at 24 weeks postoperatively (P < .001). CONCLUSIONS Preoperatively, patients with femoroacetabular impingement syndrome have a high prevalence of sleep disturbance. These patients experience subsequent improvement in sleep disturbance after arthroscopic hip surgery early in the postoperative period. LEVEL OF EVIDENCE Level IV, case series.
Collapse
Affiliation(s)
- Kyle N Kunze
- Section of Young Adult Hip Surgery, Division of Sports Medicine, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois, U.S.A
| | - Natalie L Leong
- Section of Young Adult Hip Surgery, Division of Sports Medicine, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois, U.S.A
| | - Edward C Beck
- Section of Young Adult Hip Surgery, Division of Sports Medicine, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois, U.S.A
| | - Charles A Bush-Joseph
- Section of Young Adult Hip Surgery, Division of Sports Medicine, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois, U.S.A
| | - Shane J Nho
- Section of Young Adult Hip Surgery, Division of Sports Medicine, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois, U.S.A..
| |
Collapse
|
21
|
Shigiyama F, Kumashiro N, Tsuneoka Y, Igarashi H, Yoshikawa F, Kakehi S, Funato H, Hirose T. Mechanisms of sleep deprivation-induced hepatic steatosis and insulin resistance in mice. Am J Physiol Endocrinol Metab 2018; 315:E848-E858. [PMID: 29989853 DOI: 10.1152/ajpendo.00072.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sleep deprivation is associated with increased risk for type 2 diabetes mellitus. However, the underlying mechanisms of sleep deprivation-induced glucose intolerance remain elusive. The aim of this study was to investigate the mechanisms of sleep deprivation-induced glucose intolerance in mice with a special focus on the liver. We established a mouse model of sleep deprivation-induced glucose intolerance using C57BL/6J male mice. A single 6-h sleep deprivation by the gentle handling method under fasting condition induced glucose intolerance. Hepatic glucose production assessed by a pyruvate challenge test was significantly increased, as was hepatic triglyceride content (by 67.9%) in the sleep deprivation group, compared with freely sleeping control mice. Metabolome and microarray analyses were used to evaluate hepatic metabolites and gene expression levels and to determine the molecular mechanisms of sleep deprivation-induced hepatic steatosis. Hepatic metabolites, such as acetyl coenzyme A, 3β-hydroxybutyric acid, and certain acylcarnitines, were significantly increased in the sleep deprivation group, suggesting increased lipid oxidation in the liver. In contrast, fasted sleep-deprived mice showed that hepatic gene expression levels of elongation of very long chain fatty acids-like 3, lipin 1, perilipin 4, perilipin 5, and acyl-CoA thioesterase 1, which are known to play lipogenic roles, were 2.7, 4.5, 3.7, 2.9, and 2.8 times, respectively, those of the fasted sleeping control group, as assessed by quantitative RT-PCR. Sleep deprivation-induced hepatic steatosis and hepatic insulin resistance seem to be mediated through upregulation of hepatic lipogenic enzymes.
Collapse
Affiliation(s)
- Fumika Shigiyama
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Naoki Kumashiro
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Hiroyuki Igarashi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Fukumi Yoshikawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Saori Kakehi
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine , Tokyo , Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiromasa Funato
- Department of Anatomy, Toho University Graduate School of Medicine , Tokyo , Japan
| | - Takahisa Hirose
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Toho University Graduate School of Medicine , Tokyo , Japan
| |
Collapse
|