1
|
Puech C, Badran M, Barrow MB, Gozal D. Cognitive Function, Sleep, and Neuroinflammatory Markers in Mice Exposed to Very Long-Term Intermittent Hypoxia. Int J Mol Sci 2025; 26:1815. [PMID: 40076441 PMCID: PMC11899729 DOI: 10.3390/ijms26051815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 03/14/2025] Open
Abstract
Chronic intermittent hypoxia (IH) is one of the hallmark features of obstructive sleep apnea (OSA) and adversely affects neurocognitive and behavioral functioning. However, how the duration of IH correlates with its deleterious effects remains unexplored. We aimed to assess the effects of IH over a prolonged period of time mimicking untreated OSA. Male C57Bl/6J mice were exposed to IH for 96 weeks. Sleep activity was acquired using a piezoelectric system. Novel object recognition (NOR) and the elevated plus maze test (EPMT) were conducted as measures of cognitive function and anxiety, respectively. Brain inflammation was evaluated by a panel of inflammation marker assays. All tests were performed after 16 and 96 weeks of IH exposure. After 96 weeks, sleep percentages during the dark phase decreased in both IH and room air (RA) compared to 16-week exposure (RA: p = 0.0214; IH: p = 0.0188). In addition to age-dependent declines in NOR performance, the mice after 96 weeks of IH exposure had lower NOR preference scores than RA controls (p = 0.0070). The time spent in open arms of the EPMT was reduced in mice exposed to IH compared to RA. Inflammatory marker expression increased in IH-exposed mice. Thus, aging and IH induce similar alterations in sleep, cognition, and neuroinflammation. However, the effects of aging are exacerbated by concurrent IH, suggesting that OSA is a disease associated with an acceleration in biological aging.
Collapse
Affiliation(s)
- Clementine Puech
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65201, USA; (C.P.); (M.B.); (M.B.B.)
| | - Mohammad Badran
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65201, USA; (C.P.); (M.B.); (M.B.B.)
| | - Max B. Barrow
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65201, USA; (C.P.); (M.B.); (M.B.B.)
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
2
|
Brice KN, Braden-Kuhle PN, Miller SK, Regan A, Lacy V, Chumley MJ, Boehm GW. Chronic sleep loss alters the inflammatory response and BDNF expression in C57BL/6J mice. J Neuroimmunol 2024; 396:578462. [PMID: 39378741 DOI: 10.1016/j.jneuroim.2024.578462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/30/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
Although adequate sleep is imperative for proper physiological function, over one-third of US adults obtain insufficient sleep. The current research investigated the impact of chronic sleep restriction (CSR) on inflammatory markers and hippocampal BDNF mRNA, following an immune insult in both male and female mice. Patterns of cytokine expression were different when the study was done in males vs. females, indicating potential sex differences in the inflammatory response following CSR. Further, CSR led to suppressed hippocampal BDNF expression in males, an effect not observed in females. These data suggest a complex interaction between chronic sleep loss, inflammation, and sex that warrants further exploration.
Collapse
Affiliation(s)
- Kelly N Brice
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, United States of America.
| | - Paige N Braden-Kuhle
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Shelby K Miller
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Allison Regan
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Vivienne Lacy
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Michael J Chumley
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Gary W Boehm
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| |
Collapse
|
3
|
Kaur M, Mehta R, Muthuswami R, Mallick BN. Noradrenaline enhances Na-K ATPase subunit expression by HuR-induced mRNA stabilization and their transportation to the cell surface through PLC and PKC mediated pathway: Implications with REMS-loss associated disorders. J Neurochem 2024; 168:2561-2576. [PMID: 38676340 DOI: 10.1111/jnc.16116] [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: 10/03/2023] [Revised: 03/08/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Rapid eye movement sleep (REMS) maintains brain excitability at least by regulating Na-K ATPase activity. Although REMS deprivation (REMSD)-associated elevated noradrenaline (NA) increases Na-K ATPase protein expression, its mRNA transcription did not increase. We hypothesized and confirmed both in vivo as well as in vitro that elevated mRNA stability explains the apparent puzzle. The mRNA stability was measured in control and REMSD rat brain with or without in vivo treatment with α1-adrenoceptor (AR) antagonist, prazosin (PRZ). Upon REMSD, Na-K ATPase α1-, and α2-mRNA stability increased significantly, which was prevented by PRZ. To decipher the molecular mechanism of action, we estimated NA-induced Na-K ATPase mRNA stability in Neuro-2a cells under controlled conditions and by transcription blockage using Actinomycin D (Act-D). NA increased Na-K ATPase mRNA stability, which was prevented by PRZ and propranolol (PRP, β-AR antagonist). The knockdown assay confirmed that the increased mRNA stabilization was induced by elevated cytoplasmic abundance of Human antigen R (HuR) and involving (Phospholipase C) PLC-mediated activation of Protein Kinase C (PKC). Additionally, using cell-impermeable Enz-link sulfo NHS-SS-Biotin, we observed that NA increased Na-K ATPase α1-subunits on the Neuro-2a cell surface. We conclude that REMSD-associated elevated NA, acting on α1- and β-AR, increases nucleocytoplasmic translocation of HuR and increases Na-K ATPase mRNA stability, resulting in increased Na-K ATPase protein expression. The latter then gets translocated to the neuronal membrane surface involving both PKC and (Protein Kinase A) PKA-mediated pathways. These findings may be exploited for the amelioration of REMSD-associated chronic disorders and symptoms.
Collapse
Affiliation(s)
- Manjeet Kaur
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rachna Mehta
- AMITY Institute of Neuropsychology and Neurosciences, AMITY University Uttar Pradesh, Noida, UP, India
| | - Rohini Muthuswami
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Birendra Nath Mallick
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
- AMITY Institute of Neuropsychology and Neurosciences, AMITY University Uttar Pradesh, Noida, UP, India
| |
Collapse
|
4
|
Wang W, Chen Z, Zhang W, Yuan R, Sun Y, Yao Q, Lu J, Zheng J. Association between obesity and sleep disorder in the elderly: evidence from NHANES 2005-2018. Front Nutr 2024; 11:1401477. [PMID: 39267860 PMCID: PMC11390407 DOI: 10.3389/fnut.2024.1401477] [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: 04/11/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024] Open
Abstract
Background The available data exhibit inconsistent findings regarding the association between obesity and sleep problems among older adults. The objective of this study was to assess the potential association between obesity and sleep disorders in the older population. Methods The data utilised in this cross-sectional investigation was obtained from the National Health and Nutritional Examination Survey (NHANES) conducted between 2005 and 2018. The study employed a multivariate logistic regression model and conducted subgroup analysis to evaluate the association between obesity and sleep disturbance. Results The investigation consisted of 2,570 older people who provided complete information, out of which 324 individuals satisfied the criteria for sleep disturbance. The findings from the multivariable adjusted logistic regression model indicate that individuals in the overweight and normal weight groups exhibited decreased odds of experiencing sleep disorder, as evidenced by the adjusted odds ratios (AOR) of 0.46 (95% confidence interval [CI] = 0.34-0.61) and 0.33 (95% CI = 0.22-0.47), respectively. These results were statistically significant (p < 0.001) when compared to individuals in the obese group. The investigation of age and gender subgroups demonstrated similar associations between various BMI categories and sleep disorders in the older population. Conclusion In summary, there exists a correlation between obesity and sleep disorders in the senior population. A significant association was observed between BMI and the likelihood of experiencing sleep disorders, indicating a dose-response relationship. Individuals with a higher BMI demonstrated a heightened likelihood of experiencing sleep disorders compared to those with a lower BMI.
Collapse
Affiliation(s)
- Weifei Wang
- Department of Gerontology, The First Affiliated Hospital of Ningbo University, Zhejiang, China
| | - Zhong Chen
- Department of Anesthesiology, Beilun District People's Hospital, Ningbo, Zhejiang, China
- Meigu County People's Hospital, Liangshan Prefecture, Sichuan, China
| | - Wenyuan Zhang
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Rui Yuan
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Zhejiang, China
| | - Yaqi Sun
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qi Yao
- Department of Gerontology, The First Affiliated Hospital of Ningbo University, Zhejiang, China
| | - Jian Lu
- Department of Ultrasound in Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jungang Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Zhejiang, China
- The First People's Hospital of Yuexi County, Liangshan Prefecture, Sichuan, China
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Barreto ACM, Oliveira JNS, Suchecki D. Chronic sleep restriction during juvenility alters hedonic and anxiety-like behaviours in a sex-dependent fashion in adolescent Wistar rats. Front Neurosci 2024; 18:1452429. [PMID: 39188806 PMCID: PMC11346248 DOI: 10.3389/fnins.2024.1452429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic reduction of sleep time in children and adolescents has been related to increased incidence of anxiety and depression. In rats, protocols of protracted sleep deprivation or chronic sleep restriction (CSR) are considered a stressor. In previous studies we showed that post-weaning CSR in male rats induces anxiety-like behaviour and changes in neurotransmission in emotion-related brain areas. In the present study we examined whether the effects of this adversity are sex-dependent. Twenty-two litters, containing four males and four females were distributed into control (CTL) and CSR groups. CSR began on postnatal day (PND) 21 and lasted for 21 days; each day the animals were placed onto small platforms immersed in water for 18 h and were allowed to sleep freely in their home-cages for the remaining 6 h. Throughout the CSR, all animals underwent the sucrose splash test once/week to assess their self-care and hedonic behaviours. Body weight was measured on PNDs 21 and 42. At the end of CSR period, the adolescents were allowed to sleep freely for 2 days, after which, behavioural tests began. Within each litter, one male and one female (pair) were not tested and provided blood and brain for determination of basal corticosterone (CORT) levels and hippocampal BDNF. One pair was tested in the sucrose preference test (SPT), one pair on the elevated plus maze (EPM) and one pair in the forced swim test (FST). CORT was measured after all conditions. CSR impaired self-care behaviour and body weight gain in males and females and increased relative adrenal weight only in males. There were no changes in sucrose intake in the SPT; CSR females displayed less immobility in the FST and CSR males displayed more anxiety-like behaviour in the EPM. CORT levels were similar between CTL and CSR males, whilst lower in CSR females than CTL ones in all experimental conditions. No changes in BDNF levels were detected in the dorsal hippocampus of CSR rats. The results indicate that CSR impaired self-care behaviour in both sexes, but only males displayed anxiety-like behaviour, whilst sleep recovery in females appeared to normalise their behaviour.
Collapse
Affiliation(s)
| | | | - Deborah Suchecki
- Group of Studies on the Neurobiology of Stress and its Disorders – GENED, Department of Psychobiology – Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Tonet NS, da Silva Marçal DF, da Silva FN, Brunetta HS, Mori MADS, Dos Santos GJ, Moreira ELG, Rafacho A. Moderate chronic sleep perturbation impairs glucose and lipid homeostasis in rats. Sleep 2024; 47:zsae118. [PMID: 38788154 DOI: 10.1093/sleep/zsae118] [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: 01/27/2024] [Revised: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
STUDY OBJECTIVES Sleep deprivation is a potential risk factor for metabolic diseases, including obesity and type 2 diabetes. We evaluated the impacts of moderate chronic sleep deprivation on glucose and lipid homeostasis in adult rats. METHODS Wistar rats (both sexes) were sleep-perturbed daily for 2 hours at the early (06:00-08:00) and the late light cycle (16:00-18:00) five days a week (except weekends) for 4 weeks. RESULTS Sleep perturbation (SP) resulted in reduced body weight gain in both sexes, associated with altered food intake and reduced adiposity. SP did not alter the short- or long-term memories or cause anxiogenic behavior. No major changes were observed in the plasma insulin, leptin, triacylglycerol, non-esterified fatty acids, and blood glucose upon SP. After SP, females exhibited a transitory glucose intolerance, while males became glucose intolerant at the end of the experimental period. Male rats also developed higher insulin sensitivity at the end of the SP protocol. Morphometric analyses revealed no changes in hepatic glycogen deposition, pancreatic islet mass, islet-cell distribution, or adrenal cortex thickness in SP rats from both sexes, except for lower adipocyte size compared with controls. We did not find homogeneous changes in the relative expression of circadian and metabolic genes in muscle or hepatic tissues from the SP rats. CONCLUSIONS Moderate chronic SP reduces visceral adiposity and causes glucose intolerance with a more pronounced impact on male rats, reinforcing the metabolic risks of exposure to sleep disturbances.
Collapse
Affiliation(s)
- Natália Stinghen Tonet
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Graduate Program in Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Danilo Francisco da Silva Marçal
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Flavia Natividade da Silva
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Henver Simionato Brunetta
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo Alves da Silva Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil
| | - Gustavo Jorge Dos Santos
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eduardo Luiz Gasnhar Moreira
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Alex Rafacho
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Graduate Program in Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
7
|
Manoharan N, Parasuraman R, Jayamurali D, Muthusamy P, Govindarajulu S. Role of Thymoquinone on sleep restriction and its mitigating effect on leptin-mediated signaling pathway in rat brain. Mol Biol Rep 2024; 51:769. [PMID: 38886257 DOI: 10.1007/s11033-024-09699-9] [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: 03/18/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Sleep and stress interact bidirectionally by acting on brain circuits that affect metabolism. Sleep and its alterations have impact on blood leptin levels, metabolic hormone that regulates appetite. Brain expresses the receptors for the peptide hormone leptin produced from adipocytes. The hypothalamic orexin neurons are low during sleep and active when awake, influenced by a complex interaction with leptin. Thymoquinone was found to be the major bioactive component of Nigella sativa. The aim of this study was to study the role of thymoquinone on sleep restriction and its mitigating effect on leptin-mediated signaling pathway in rat brain. METHODS AND RESULTS 30 adult male Wistar rats were divided into 5 groups with 6 animals in each group: Control; Thymoquinone (TQ); Corn oil; Chronic Sleep restriction (CSR); and CSR + TQ. After 30 days, behavioral analysis, antioxidant, lipid profile, glucose level, liver and kidney function test, neurotransmitters, neuropeptides, and mRNA expression in in vivo studies were also assessed and pharmacokinetic and docking were done for thymoquinone. Thymoquinone has also shown good binding affinity to the target proteins. CSR has induced oxidative stress in the discrete brain regions and plasma. Current study has shown many evidences that sleep restriction has altered the neurobehavioral, antioxidant status, lipid profile, neurotransmitters, neuropeptide levels, and feeding behavior which damage the Orexin-leptin system which regulates the sleep and feeding that leads to metabolic dysfunction. CONCLUSION The potentiality of Thymoquinone was revealed in in silico studies, and its action in in vivo studies has proved its effectiveness. The study concludes that Thymoquinone has exhibited its effect by diminishing the metabolic dysfunction by its neuroprotective, antioxidant, and hypolipidemic properties.
Collapse
Affiliation(s)
- Nivedita Manoharan
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India
| | - Rajeshwari Parasuraman
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India
| | - Dheepthi Jayamurali
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India
| | - Pazhanisankar Muthusamy
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India
| | - Sathyanarayanan Govindarajulu
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India.
| |
Collapse
|
8
|
Ferini-Strambi L, Liguori C, Lucey BP, Mander BA, Spira AP, Videnovic A, Baumann C, Franco O, Fernandes M, Gnarra O, Krack P, Manconi M, Noain D, Saxena S, Kallweit U, Randerath W, Trenkwalder C, Rosenzweig I, Iranzo A, Bradicich M, Bassetti C. Role of sleep in neurodegeneration: the consensus report of the 5th Think Tank World Sleep Forum. Neurol Sci 2024; 45:749-767. [PMID: 38087143 DOI: 10.1007/s10072-023-07232-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/26/2023] [Indexed: 01/18/2024]
Abstract
Sleep abnormalities may represent an independent risk factor for neurodegeneration. An international expert group convened in 2021 to discuss the state-of-the-science in this domain. The present article summarizes the presentations and discussions concerning the importance of a strategy for studying sleep- and circadian-related interventions for early detection and prevention of neurodegenerative diseases. An international expert group considered the current state of knowledge based on the most relevant publications in the previous 5 years; discussed the current challenges in the field of relationships among sleep, sleep disorders, and neurodegeneration; and identified future priorities. Sleep efficiency and slow wave activity during non-rapid eye movement (NREM) sleep are decreased in cognitively normal middle-aged and older adults with Alzheimer's disease (AD) pathology. Sleep deprivation increases amyloid-β (Aβ) concentrations in the interstitial fluid of experimental animal models and in cerebrospinal fluid in humans, while increased sleep decreases Aβ. Obstructive sleep apnea (OSA) is a risk factor for dementia. Studies indicate that positive airway pressure (PAP) treatment should be started in patients with mild cognitive impairment or AD and comorbid OSA. Identification of other measures of nocturnal hypoxia and sleep fragmentation could better clarify the role of OSA as a risk factor for neurodegeneration. Concerning REM sleep behavior disorder (RBD), it will be crucial to identify the subset of RBD patients who will convert to a specific neurodegenerative disorder. Circadian sleep-wake rhythm disorders (CSWRD) are strong predictors of caregiver stress and institutionalization, but the absence of recommendations or consensus statements must be considered. Future priorities include to develop and validate existing and novel comprehensive assessments of CSWRD in patients with/at risk for dementia. Strategies for studying sleep-circadian-related interventions for early detection/prevention of neurodegenerative diseases are required. CSWRD evaluation may help to identify additional biomarkers for phenotyping and personalizing treatment of neurodegeneration.
Collapse
Affiliation(s)
- Luigi Ferini-Strambi
- Sleep Disorders Center, Division of Neuroscience, Università Vita-Salute San Raffaele, Milan, Italy.
| | - Claudio Liguori
- Sleep Medicine Center, University of Rome Tor Vergata, Rome, Italy
| | - Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Aleksandar Videnovic
- Department of Neurology, Division of Sleep Medicine, Massachussets General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian Baumann
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Oscar Franco
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | | | - Oriella Gnarra
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Paul Krack
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Mauro Manconi
- Sleep Medicine Unit, Faculty of Biomedical Sciences, Neurocenter of the Southern Switzerland, Regional Hospital of Lugano, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Daniela Noain
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Smita Saxena
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Ulf Kallweit
- Clinical Sleep and Neuroimmunology, University Witten/Herdecke, Witten, Germany
| | | | - C Trenkwalder
- Department of Neurosurgery, Paracelsus-Elena Klinik, University Medical Center, KasselGoettingen, Germany
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, King's College London, London, UK
| | - Alex Iranzo
- Sleep Center, Neurology Service, Hospital Clinic de Barcelona, Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Matteo Bradicich
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
9
|
Arocha Rodulfo JI, Aure Fariñez G, Carrera F. Sleep and cardiometabolic risk. Narrative revision. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:38-49. [PMID: 37696704 DOI: 10.1016/j.arteri.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVES Sleep disturbances, including disrupted sleep and short sleep duration, are highly prevalent and are prospectively associated with an increased risk for various chronic diseases, including cardiometabolic, neurodegenerative, and autoimmune diseases. MATERIAL AND METHODS This is a narrative review of the literature based on numerous articles published in peer-reviewed journals since the beginning of this century. RESULTS The relationship between sleep disorders and metabolic dysregulation has been clearly established, mainly in the setting of modern epidemic of cardiometabolic disease, a cluster of conditions include obesity, insulin resistance, arterial hypertension, and dyslipidaemia, all of them considered as main risk factor for atherosclerotic cardiovascular disease (ACVD) and its clinical expression such as ischemic ictus, myocardial infarction and type 2 diabetes. Clinically viable tools to measure sleep duration and quality are needed for routine screening and intervention. CONCLUSIONS In view of what has been exposed in this review, it is evident that the timing, amount, and quality of sleep are critical to reduce the burden of risk factors for several chronic disease, including ACVD and type 2 diabetes, and most relevant in young people. Future research studies should elucidate the effectiveness of multimodal interventions to counteract the risk of short sleep for optimal patient outcomes across the healthcare continuum, especially in young people.
Collapse
Affiliation(s)
| | | | - Fernando Carrera
- Fellowship en Diabetes y Metabolismo, Hospital Vargas de Caracas, Caracas, Venezuela
| |
Collapse
|
10
|
Puech C, Badran M, Runion AR, Barrow MB, Cataldo K, Gozal D. Cognitive Impairments, Neuroinflammation and Blood-Brain Barrier Permeability in Mice Exposed to Chronic Sleep Fragmentation during the Daylight Period. Int J Mol Sci 2023; 24:9880. [PMID: 37373028 DOI: 10.3390/ijms24129880] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic condition characterized by intermittent hypoxia (IH) and sleep fragmentation (SF). In murine models, chronic SF can impair endothelial function and induce cognitive declines. These deficits are likely mediated, at least in part, by alterations in Blood-brain barrier (BBB) integrity. Male C57Bl/6J mice were randomly assigned to SF or sleep control (SC) conditions for 4 or 9 weeks and in a subset 2 or 6 weeks of normal sleep recovery. The presence of inflammation and microglia activation were evaluated. Explicit memory function was assessed with the novel object recognition (NOR) test, while BBB permeability was determined by systemic dextran-4kDA-FITC injection and Claudin 5 expression. SF exposures resulted in decreased NOR performance and in increased inflammatory markers and microglial activation, as well as enhanced BBB permeability. Explicit memory and BBB permeability were significantly associated. BBB permeability remained elevated after 2 weeks of sleep recovery (p < 0.01) and returned to baseline values only after 6 weeks. Chronic SF exposures mimicking the fragmentation of sleep that characterizes patients with OSA elicits evidence of inflammation in brain regions and explicit memory impairments in mice. Similarly, SF is also associated with increased BBB permeability, the magnitude of which is closely associated with cognitive functional losses. Despite the normalization of sleep patterns, BBB functional recovery is a protracted process that merits further investigation.
Collapse
Affiliation(s)
- Clementine Puech
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
| | - Mohammad Badran
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
| | - Alexandra R Runion
- Undergraduate Student Research Program, University of Missouri, Columbia, MO 65201, USA
| | - Max B Barrow
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
| | - Kylie Cataldo
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
| | - David Gozal
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
11
|
Zhang W, Shi X, Zhang Y, Liu G, Wu X, Huang H, Jiang H, Zhang X. Attenuation Effect of Recovery Sleep for Impaired Reproductive Function in Male Rats by Sleep Deprivation. World J Mens Health 2023:41.e8. [PMID: 36593710 DOI: 10.5534/wjmh.220130] [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: 07/01/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 01/03/2023] Open
Abstract
PURPOSE The aim of the present study was to test the hypothesis that recovery sleep could counteract the detrimental effects of sleep deprivation (SD) on male rats' fertility. MATERIALS AND METHODS Twenty-two rats were housed in groups of six per cage with unrestricted access to food and water in a room. The modified multiple platform method was used to induce SD in rats over a 96-hour period. We examined the effect of SD on semen quality, reproductive hormones, and testicular histology in adult male rats. Then, we investigated the effect of 7 days recovery sleep on impaired reproductive function induced by SD. RESULTS After the acclimation period, 22 rats were randomly separated into three experimental groups (SD, recovery sleep, and the control groups). Ninety-six hours of SD resulted in a significant decrease in sperm motility (24.33±10.93 vs. 48.20±8.55, p<0.001) and the number of morphologically normal sperm (9.68±2.77 vs. 26.21±14.60, p<0.01) in rats, accompanied by a decrease in testosterone levels (1.53±0.55 vs. 4.44±0.56, p<0.001) and destruction of testicular tissue structure compared with control group. After 7 days of recovery sleep, semen quality, especially sperm motility, was improved and testosterone levels were significantly higher compared to post-SD (3.70±0.53 vs. 1.53±0.55, p<0.05), but remained low compared to the control group. CONCLUSIONS In conclusion, 96 hours of SD deteriorated the parameters of sperm motility and the number of morphologically normal sperm in rats, probably due to the decrease in serum testosterone levels and the disruption of testicular tissue structure when compared to the control group. After 7 days of recovery sleep, semen parameter, especially sperm motility and testosterone levels did not return to baseline levels compared to the control group.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiao Shi
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuyang Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guodong Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xu Wu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Houbao Huang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Hui Jiang
- Andrology Center, Peking University First Hospital, Beijing, China.
| | - Xiansheng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
12
|
Duan D, Kim LJ, Jun JC, Polotsky VY. Connecting insufficient sleep and insomnia with metabolic dysfunction. Ann N Y Acad Sci 2023; 1519:94-117. [PMID: 36373239 PMCID: PMC9839511 DOI: 10.1111/nyas.14926] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The global epidemic of obesity and type 2 diabetes parallels the rampant state of sleep deprivation in our society. Epidemiological studies consistently show an association between insufficient sleep and metabolic dysfunction. Mechanistically, sleep and circadian rhythm exert considerable influences on hormones involved in appetite regulation and energy metabolism. As such, data from experimental sleep deprivation in humans demonstrate that insufficient sleep induces a positive energy balance with resultant weight gain, due to increased energy intake that far exceeds the additional energy expenditure of nocturnal wakefulness, and adversely impacts glucose metabolism. Conversely, animal models have found that sleep loss-induced energy expenditure exceeds caloric intake resulting in net weight loss. However, animal models have significant limitations, which may diminish the clinical relevance of their metabolic findings. Clinically, insomnia disorder and insomnia symptoms are associated with adverse glucose outcomes, though it remains challenging to isolate the effects of insomnia on metabolic outcomes independent of comorbidities and insufficient sleep durations. Furthermore, both pharmacological and behavioral interventions for insomnia may have direct metabolic effects. The goal of this review is to establish an updated framework for the causal links between insufficient sleep and insomnia and risks for type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Daisy Duan
- Division of Endocrinology, Diabetes, and Metabolism; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lenise J. Kim
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan C. Jun
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
Zhu Y, Chen X, Guo L, Wang L, Chen N, Xiao Y, Wang E. Acute sleep deprivation increases inflammation and aggravates heart failure after myocardial infarction. J Sleep Res 2022; 31:e13679. [PMID: 35785454 PMCID: PMC9786274 DOI: 10.1111/jsr.13679] [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: 12/17/2021] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 12/30/2022]
Abstract
Sleep disorders have been observed among patients with heart failure. The aim of this study was to investigate whether acute sleep deprivation (SD) aggravates left heart function. Male C57B/L6 mice were assigned to four experimental groups. Ligation of the left anterior descending branch (LAD) caused myocardial infarction (MI) in mice in the LAD group and the LAD+SD group, while mice in the sham and sham+SD groups underwent the same surgery without ligation. Echocardiography was performed before and 8 weeks after ligation of the LAD to evaluate the left ventricular internal diameter at diastole (LVIDd), left ventricular internal diameter at systole (LVIDs), ejection fraction (EF), and fractional shortening (FS). Seven days of sleep deprivation induced using the modified single platform method resulted in a lower EF and FS and a higher LVIDd and LVIDs, as well as increased expression of the IL-1β, IL-18, and IL-10 mRNAs in the left ventricular tissue of MI mice. ELISA also indicated higher levels of IL-1β and IL-10 in the LAD+SD group. It was concluded that acute sleep deprivation induced cardiovascular alterations in cardiac structure and function in HF mice, accompanied by increased levels of inflammatory cytokines.
Collapse
Affiliation(s)
- Yumin Zhu
- Department of Anesthesiologythe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhouChina
| | - Xian Chen
- The First Affiliated Hospital of Suzhou UniversitySuzhouChina
| | - Lizhe Guo
- Department of AnesthesiologyXiangya Hospital Central South UniversityChangshaChina
| | - Lu Wang
- Department of AnesthesiologyXiangya Hospital Central South UniversityChangshaChina
| | - Na Chen
- Department of AnesthesiologyXiangya Hospital Central South UniversityChangshaChina
| | - Yujie Xiao
- Department of AnesthesiologyXiangya Hospital Central South UniversityChangshaChina
| | - E. Wang
- Department of AnesthesiologyXiangya Hospital Central South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| |
Collapse
|
14
|
Mishra I, Pullum KB, Eads KN, Strunjas AR, Ashley NT. Peripheral Sympathectomy Alters Neuroinflammatory and Microglial Responses to Sleep Fragmentation in Female Mice. Neuroscience 2022; 505:111-124. [PMID: 36240943 PMCID: PMC9671838 DOI: 10.1016/j.neuroscience.2022.09.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/05/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Sleep loss, either induced by obstructive sleep apnea or other forms of sleep dysfunction, induces an inflammatory response, as commonly measured by increased circulating levels of pro-inflammatory cytokines. Increased catecholamines from sympathetic nervous system (SNS) activation regulates this peripheral inflammation. However, the role that catecholamines play in mediating neuroinflammation from sleep perturbations is undescribed. The aims of this study were to determine (i) the effect of peripheral SNS inhibition upon neuroinflammatory responses to sleep fragmentation (SF) and (ii) whether homeostasis can be restored after 1 week of recovery sleep. We measured gene expression levels of pro- and anti-inflammatory cytokines and microglial activity in brain (prefrontal cortex, hippocampus and hypothalamus) of female mice that were subjected to acute SF for 24 hours, chronic SF for 8 weeks, or 7 days of recovery after chronic SF. In each experiment, SF and control mice were peripherally sympathectomized with 6-hydroxydopamine (6-OHDA) or injected with vehicle. SF elevated cytokine mRNA expression in brain and increased microglial density and cell area in some regions. In addition, chronic SF promoted hyper-ramification in resting microglia upon exposure to chronic, but not acute, SF. Effects of chronic SF were more pronounced than acute SF, and 1 week of recovery was not sufficient to alleviate neuroinflammation. Importantly, 6-OHDA treatment significantly alleviated SF-induced inflammation and microglial responses. This study provides evidence of SNS regulation of neural inflammation from SF, suggesting a potential role for therapeutics that could mitigate neuroinflammatory responses to sleep dysfunction.
Collapse
Affiliation(s)
- Ila Mishra
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA; Harrington Discovery Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Keelee B Pullum
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kristen N Eads
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
| | - Anna R Strunjas
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
| | - Noah T Ashley
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA.
| |
Collapse
|
15
|
Markov DD, Novosadova EV. Chronic Unpredictable Mild Stress Model of Depression: Possible Sources of Poor Reproducibility and Latent Variables. BIOLOGY 2022; 11:1621. [PMID: 36358321 PMCID: PMC9687170 DOI: 10.3390/biology11111621] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023]
Abstract
Major depressive disorder (MDD) is one of the most common mood disorders worldwide. A lack of understanding of the exact neurobiological mechanisms of depression complicates the search for new effective drugs. Animal models are an important tool in the search for new approaches to the treatment of this disorder. All animal models of depression have certain advantages and disadvantages. We often hear that the main drawback of the chronic unpredictable mild stress (CUMS) model of depression is its poor reproducibility, but rarely does anyone try to find the real causes and sources of such poor reproducibility. Analyzing the articles available in the PubMed database, we tried to identify the factors that may be the sources of the poor reproducibility of CUMS. Among such factors, there may be chronic sleep deprivation, painful stressors, social stress, the difference in sex and age of animals, different stress susceptibility of different animal strains, handling quality, habituation to stressful factors, various combinations of physical and psychological stressors in the CUMS protocol, the influence of olfactory and auditory stimuli on animals, as well as the possible influence of various other factors that are rarely taken into account by researchers. We assume that careful inspection of these factors will increase the reproducibility of the CUMS model between laboratories and allow to make the interpretation of the obtained results and their comparison between laboratories to be more adequate.
Collapse
|
16
|
Intervention Effects of Okra Extract on Brain-Gut Peptides and Intestinal Microorganisms in Sleep Deprivation Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9855411. [PMID: 36193125 PMCID: PMC9526647 DOI: 10.1155/2022/9855411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/13/2022] [Indexed: 11/18/2022]
Abstract
Objective Okra, possessing various bioactive components, is used to treat different diseases. This study sought to estimate the intervention effects of okra extract (OE) on brain-gut peptides (BGPs) and intestinal microorganisms in sleep deprivation (SD) rats. Methods SD rat models were established using the modified multiple platform method and then treated with normal saline, diazepam tablets, or different doses of OE. Body weight and average daily water consumption of rats were recorded. Depressive behaviors of rats were assessed by the open field test and sucrose preference test. Serum levels of noradrenaline, melatonin, inflammatory factors (IL-1β/IL-6/TNF-α/IL-4/IL-10), and BGP indexes, including gastrin (GAS), motilin (MTL), 5-hydroxytryptamine (5-HT), cholecystokinin (CCK), and vasoactive intestinal peptide (VIP) were measured by ELISA. Additionally, the DNA relative contents of representative intestinal microorganisms in the collected rat feces were determined using RT-qPCR. Results SD decreased body weight and average daily water consumption and induced depressive behaviors as well as stress and inflammatory responses in rats. SD rats exhibited lowered GAS, MTL, 5-HT, and VIP but elevated CCK and showed diminished DNA relative contents of Bacteroidetes and probiotics (Bifidobacteria and Lactobacilli) but increased Clostridium perfringens. OE at different doses ameliorated the depressive behaviors and mitigated the stress and inflammatory responses in SD rats, raised the serum contents of GAS, MTL, 5-HT, and VIP, reduced CCK level, elevated the DNA relative contents of Bacteroidetes and probiotics, but diminished Clostridium perfringens. OE exhibited similar intervention effects to diazepam tablets (positive control). Conclusion OE exerts intervention effects on BGPs and intestinal microorganisms in SD rats.
Collapse
|
17
|
Kuna K, Szewczyk K, Gabryelska A, Białasiewicz P, Ditmer M, Strzelecki D, Sochal M. Potential Role of Sleep Deficiency in Inducing Immune Dysfunction. Biomedicines 2022; 10:biomedicines10092159. [PMID: 36140260 PMCID: PMC9496201 DOI: 10.3390/biomedicines10092159] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Sleep deficiency and insomnia deteriorate the quality of patients’ lives, yet the exact influence of these factors on the immune system has only begun to gain interest in recent years. Growing evidence shows that insomnia is a risk factor for numerous diseases, including common infections and autoimmune diseases. Levels of inflammatory markers also seem to be abnormal in sleep deficient individuals, which may lead to low-grade inflammation. The interpretation of studies is difficult due to the equivocal term “sleep disturbances,” as well as due to the various criteria used in studies. This narrative review aims to summarize the available knowledge regarding the bidirectional influence of the immune system and sleep disturbances.
Collapse
Affiliation(s)
- Kasper Kuna
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Krzysztof Szewczyk
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Marta Ditmer
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-213 Lodz, Poland
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
- Correspondence: ; Tel.: +48-42-678-18-00
| |
Collapse
|
18
|
Chronic rapid eye movement sleep restriction during juvenility has long-term effects on anxiety-like behaviour and neurotransmission of male Wistar rats. Pharmacol Biochem Behav 2022; 217:173410. [PMID: 35662652 DOI: 10.1016/j.pbb.2022.173410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/21/2022]
Abstract
Modernity imposes a toll on the sleep time of young population, with concomitant increase in symptoms of anxiety and depression. Whether there is a causal relationship between these events are only now being experimentally tested in humans and rodents. In a previous study, we showed that chronic sleep deprivation in juvenile-adolescent male rats led to increased anxiety-like behaviour and changes in noradrenaline and serotonin in the amygdala and hippocampus. In the present study we investigated whether early chronic sleep restriction affects emotional behaviour, stress response and neurochemistry in adulthood. From 21 to 42 days of age, Wistar male rats were submitted to sleep restriction by the multiple platform method or allowed to sleep freely. Forty-five days after this period, rats were tested in the elevated plus maze (EPM) and blood samples were collected from non-tested rats or 30 and 60 min after the EPM for determination of plasma corticosterone levels. Levels of monoamines were determined in the frontal cortex, hippocampus, amygdala and hypothalamus 60 min after the EPM. Sleep restriction resulted in increased anxiety-like behaviour, decreased noradrenaline levels in the amygdala and dopamine levels in the ventral hippocampus. Anxiety index was positively correlated with increased serotonin metabolism in the frontal cortex and greater dopamine metabolism in the ventral hippocampus, and negatively correlated with dopamine levels in the ventral hippocampus. These results suggest that sleep restriction in juvenility and adolescence induces persistent changes in emotional behaviour in adult male rats and that levels of anxiety are correlated with increased serotonin and dopamine metabolism in specific brain areas.
Collapse
|
19
|
Marques CG, Dos Santos Quaresma MVL, Nakamoto FP, Magalhães ACO, Lucin GA, Thomatieli-Santos RV. Does Modern Lifestyle Favor Neuroimmunometabolic Changes? A Path to Obesity. Front Nutr 2021; 8:705545. [PMID: 34621773 PMCID: PMC8490681 DOI: 10.3389/fnut.2021.705545] [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] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/27/2021] [Indexed: 12/19/2022] Open
Abstract
Factors linked to modern lifestyles, such as physical inactivity, Western diet, and poor sleep quality have been identified as key contributors to the positive energy balance (PEB). PEB rises adipose tissue hypertrophy and dysfunction over the years, affecting cells and tissues that are metabolically critical for energy homeostasis regulation, especially skeletal muscle, hypothalamic-pituitary-adrenal axis, and gut microbiota. It is known that the interaction among lifestyle factors and tissue metabolic dysfunction increases low-grade chronic systemic inflammation, leading to insulin resistance and other adverse metabolic disorders. Although immunometabolic mechanisms are widely discussed in obesity, neuroimmunoendocrine pathways have gained notoriety, as a link to neuroinflammation and central nervous system disorders. Hypothalamic inflammation has been associated with food intake dysregulation, which comprises homeostatic and non-homeostatic mechanisms, promoting eating behavior changes related to the obesity prevalence. The purpose of this review is to provide an updated and integrated perspective on the effects of Western diet, sleep debt, and physical exercise on the regulation of energy homeostasis and low-grade chronic systemic inflammation. Subsequently, we discuss the intersection between systemic inflammation and neuroinflammation and how it can contribute to energy imbalance, favoring obesity. Finally, we propose a model of interactions between systemic inflammation and neuroinflammation, providing new insights into preventive and therapeutic targets for obesity.
Collapse
Affiliation(s)
- Camila Guazzelli Marques
- Programa de Pós-graduação em Psicobiologia, Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Ana Carolina Oumatu Magalhães
- Programa de Pós-graduação em Psicobiologia, Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Nutrição, Centro Universitário São Camilo, São Paulo, Brazil
| | | | - Ronaldo Vagner Thomatieli-Santos
- Programa de Pós-graduação em Psicobiologia, Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Biociências, Universidade Federal de São Paulo, Santos, Brazil
| |
Collapse
|
20
|
García-Aviles JE, Méndez-Hernández R, Guzmán-Ruiz MA, Cruz M, Guerrero-Vargas NN, Velázquez-Moctezuma J, Hurtado-Alvarado G. Metabolic Disturbances Induced by Sleep Restriction as Potential Triggers for Alzheimer's Disease. Front Integr Neurosci 2021; 15:722523. [PMID: 34539357 PMCID: PMC8447653 DOI: 10.3389/fnint.2021.722523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/26/2021] [Indexed: 01/15/2023] Open
Abstract
Sleep has a major role in learning, memory consolidation, and metabolic function. Although it is known that sleep restriction increases the accumulation of amyloid β peptide (Aβ) and the risk to develop Alzheimer's disease (AD), the mechanism behind these effects remains unknown. In this review, we discuss how chronic sleep restriction induces metabolic and cognitive impairments that could result in the development of AD in late life. Here, we integrate evidence regarding mechanisms whereby metabolic signaling becomes disturbed after short or chronic sleep restriction in the context of cognitive impairment, particularly in the accumulation of Aβ in the brain. We also discuss the role of the blood-brain barrier in sleep restriction with an emphasis on the transport of metabolic signals into the brain and Aβ clearance. This review presents the unexplored possibility that the alteration of peripheral metabolic signals induced by sleep restriction, especially insulin resistance, is responsible for cognitive deficit and, subsequently, implicated in AD development.
Collapse
Affiliation(s)
- Jesús Enrique García-Aviles
- Area of Neurosciences, Biology of Reproduction Department, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Rebeca Méndez-Hernández
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Mara A Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel Cruz
- Instituto Mexicano del Seguro Social, Centro Médico Nacional Siglo XXI, Hospital de Especialidades, Unidad de Investigación Médica en Bioquímica, Mexico City, Mexico
| | - Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Javier Velázquez-Moctezuma
- Area of Neurosciences, Biology of Reproduction Department, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Gabriela Hurtado-Alvarado
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
21
|
Zou J, Liu Y, Xu H, Meng L, Zou J, Qian Y, Chen B, Yi H, Guan J, Yin S. Association of apnea-hypopnea index during rapid eye movement sleep with insulin resistance in patients with suspected obstructive sleep apnea: a cross-sectional study. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:243. [PMID: 33708870 PMCID: PMC7940895 DOI: 10.21037/atm-20-3165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Obstructive sleep apnea (OSA) is associated with insulin resistance. However, the association between special stages of OSA [rapid eye movement (REM) sleep] and insulin resistance is not clear. This study was designed to assess the association of the frequency of respiratory events during REM sleep with insulin resistance in adults with suspected OSA. Methods In this cross-sectional study, 4,062 adult participants with suspected OSA who underwent polysomnography in our sleep center between 2009 and 2016 were screened. Polysomnographic variables, biochemical indicators, and physical measurements were collected. Logistic regression analyses were conducted to determine the odds ratios (ORs) and 95% confidence intervals (95% CIs) for insulin resistance as assessed by the presence of hyperinsulinemia, the homeostasis model assessment of insulin resistance (HOMA-IR) index, the fasting insulin resistance index (FIRI), and Bennett’s insulin sensitivity index (ISI). Results The final analyses included 2,899 adults with suspected OSA. Multivariate adjustments, including the apnea-hypopnea index (AHI) during non-REM sleep (AHINREM), were performed. The AHI during REM sleep (AHIREM) was found to be independently associated with insulin resistance across increasing AHIREM quartiles. For hyperinsulinemia the ORs (95% CIs) followed the order of 1.340 (1.022, 1.757), 1.210 (0.882, 1.660), and 1.632 (1.103, 2.416). For abnormal HOMA-IR, ORs (95% CIs) were 1.287 (0.998, 1.661), 1.263 (0.933, 1.711), and 1.556 (1.056, 2.293). For abnormal FIRI, ORs (95% CIs) were 1.386 (1.048, 1.835), 1.317 (0.954, 1.818), and 1.888 (1.269, 2.807). For abnormal Bennett’s ISI, ORs (95% CIs) were 1.297 (1.003, 1.678), 1.287 (0.949, 1.747), and 1.663 (1.127, 2.452). All linear trends were statistically significant (P<0.01). Additionally, the results showed that REM sleep duration was independently associated with hyperinsulinemia (OR =0.777, 95% CI: 0.615–0.982; P<0.05). Conclusions AHIREM was independently associated with hyperinsulinemia and an abnormal HOMA-IR, FIRI, and Bennett’s ISI in adults with suspected OSA. Additionally, REM sleep duration was independently associated with hyperinsulinemia.
Collapse
Affiliation(s)
- Juanjuan Zou
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.,Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, China
| | - Yupu Liu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Huajun Xu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Lili Meng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Jianyin Zou
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Yingjun Qian
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Bin Chen
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Hongliang Yi
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Jian Guan
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Shankai Yin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| |
Collapse
|
22
|
Brice KN, Hagen CW, Peterman JL, Figg JW, Braden PN, Chumley MJ, Boehm GW. Chronic sleep restriction increases soluble hippocampal Aβ-42 and impairs cognitive performance. Physiol Behav 2020; 226:113128. [PMID: 32791178 DOI: 10.1016/j.physbeh.2020.113128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 11/17/2022]
Abstract
Currently, over 44 million people worldwide suffer from Alzheimer's disease (AD). A common feature of AD is disrupted sleep. Sleep is essential for many psychological and physiological functions, though 35.3% of adults report getting less than 7 hours per night. The present research examined whether chronic sleep restriction would elevate hippocampal amyloid-beta1-42 expression or alter cognitive ability in adult C57BL/6 mice. Chronic sleep restriction was associated with cognitive impairment and increased hippocampal amyloid-beta. Thus, chronic sleep loss may have a detrimental effect upon cognitive function, in part, via increasing amyloid-beta levels in the hippocampus, even in non-genetically modified mice.
Collapse
Affiliation(s)
- Kelly N Brice
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Christopher W Hagen
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Julia L Peterman
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - John W Figg
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Paige N Braden
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Michael J Chumley
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Gary W Boehm
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA.
| |
Collapse
|
23
|
Pandey A, Oliver R, Kar SK. Differential Gene Expression in Brain and Liver Tissue of Wistar Rats after Rapid Eye Movement Sleep Deprivation. Clocks Sleep 2020; 2:442-465. [PMID: 33114225 PMCID: PMC7711450 DOI: 10.3390/clockssleep2040033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/13/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Sleep is essential for the survival of most living beings. Numerous researchers have identified a series of genes that are thought to regulate "sleep-state" or the "deprived state". As sleep has a significant effect on physiology, we believe that lack of total sleep, or particularly rapid eye movement (REM) sleep, for a prolonged period would have a profound impact on various body tissues. Therefore, using the microarray method, we sought to determine which genes and processes are affected in the brain and liver of rats following nine days of REM sleep deprivation. Our findings showed that REM sleep deprivation affected a total of 652 genes in the brain and 426 genes in the liver. Only 23 genes were affected commonly, 10 oppositely, and 13 similarly across brain and liver tissue. Our results suggest that nine-day REM sleep deprivation differentially affects genes and processes in the brain and liver of rats.
Collapse
Affiliation(s)
- Atul Pandey
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
- Department of Ecology, Evolution, and Behavior, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel;
| | - Ryan Oliver
- Department of Ecology, Evolution, and Behavior, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel;
| | - Santosh K Kar
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
- Nano Herb Research Laboratory, Kalinga Institute of Industrial Technology (KIIT) Technology Bio Incubator, Campus-11, KIIT Deemed to be University, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
24
|
FATP2-targeted therapies - A role beyond fatty liver disease. Pharmacol Res 2020; 161:105228. [PMID: 33027714 DOI: 10.1016/j.phrs.2020.105228] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/26/2020] [Accepted: 09/27/2020] [Indexed: 12/31/2022]
Abstract
Fatty acid transport protein 2 (FATP2) is a multifunctional protein whose specific function is determined by the type of located cell, its intracellular location, or organelle-specific interactions. In the different diseases setting, a newfound appreciation for the biological function of FATP2 has come into view. Two main functions of FATP2 are to activate long-chain fatty acids (LCFAs) as a very long-chain acyl-coenzyme A (CoA) synthetase (ACSVL) and to transport LCFAs as a fatty acid transporter. FATP2 is not only involved in the occurrence of nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM), but also plays an important role in lithogenic diet-induced cholelithiasis, the formation of cancer tumor immunity, the progression of chronic kidney disease (CKD), and the regulation of zoledronate-induced nephrotoxicity. Herein, we review the updated information on the role of FATP2 in related diseases. In particular, we discuss the new functions of FATP2 and propose that FATP2 is a potential clinical biomarker and therapeutic target. In conclusion, regulatory strategies for FATP2 may bring new treatment options for cancer and lipid metabolism-related disorders.
Collapse
|
25
|
Menezes L, de Moraes DA, Ribeiro-Silva N, Silva SMA, Suchecki D, Luz J. Chronic REM sleep restriction in young rats increases energy expenditure with no change in food intake. Exp Physiol 2020; 105:1339-1348. [PMID: 32589295 DOI: 10.1113/ep088474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/23/2020] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? What are the effects of different periods of REM sleep restriction (7, 14 and 21 days) on metabolic parameters in young rats? What is the main finding and its importance? Animals submitted to each period of REM sleep deprivation showed a negative energy balance, with reduced body weight gain, body energy gain and gross food efficiency, less body fat content, and increased energy expenditure. There was no increase in food intake after any of the REM sleep restriction periods. In young rats, negative energy balance is not compensated by increased dietary intake as observed in adult rats. ABSTRACT Reduced sleep is associated with metabolic alterations, not only in adults, but also in children and adolescents. Several studies have shown that sleep restricted (SR) adult rats exhibit metabolic changes, followed by increased food intake, but few have evaluated these functions in young animals. The aim of the present study was to establish the metabolic parameters of young rats subjected to different periods of REM sleep restriction and to propose a correction factor for the correct measurement of food intake. Young male Wistar rats were distributed in control and SR groups for 7, 14 or 21 days. Sleep restriction was performed by the single platform method for 18 h. Regardless of the length of sleep restriction, all SR rats had a negative energy balance, evidenced by reduction in body weight gain, body energy gain and gross food efficiency, accompanied by increased energy expenditure. In addition, sleep restriction reduced body fat content throughout the entire period. Discounting food spillage, there was no increase in food intake by SR rats. In conclusion, the present study revealed metabolic changes in SR young rats after different lengths of REM sleep restriction and that weight loss and increased energy expenditure were not compensated by increased dietary intake as occurs in adult rats, indicating that young rats use other mechanisms to cope with the negative energy balance caused by sleep restriction. In addition, we propose a correction factor for food intake, to prevent overestimation of this parameter, due to food spillage in the water containers.
Collapse
Affiliation(s)
- Letícia Menezes
- Department of Phisiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Neila Ribeiro-Silva
- Department of Phisiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Deborah Suchecki
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jacqueline Luz
- Department of Phisiology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Chen W, Guo X, Jin Z, Li R, Shen L, Li W, Cai W, Zhang G. Transcriptional alterations of genes related to fertility decline in male rats induced by chronic sleep restriction. Syst Biol Reprod Med 2020; 66:99-111. [DOI: 10.1080/19396368.2019.1678694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Wenyang Chen
- Hebei Provincial Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou, China
- Research and Development Department, Peking Medriv Academy of Genetics and Reproduction, Peking, China
| | - Xingdao Guo
- Research and Development Department, Peking Medriv Academy of Genetics and Reproduction, Peking, China
| | - Zhiping Jin
- Research and Development Department, Peking Medriv Academy of Genetics and Reproduction, Peking, China
| | - Runan Li
- Hebei Provincial Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou, China
- Research and Development Department, Peking Medriv Academy of Genetics and Reproduction, Peking, China
| | - Lixia Shen
- Hebei Provincial Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou, China
| | - Wei Li
- Hebei Provincial Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou, China
| | - Wangting Cai
- Research and Development Department, Peking Medriv Academy of Genetics and Reproduction, Peking, China
| | - Guirong Zhang
- Hebei Provincial Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou, China
- Research and Development Department, Peking Medriv Academy of Genetics and Reproduction, Peking, China
| |
Collapse
|
27
|
Zheng PP, Zhang LN, Zhang J, Chang XM, Ding S, Xiao F, Guo LX. Evaluating the Effects of Different Sleep Supplement Modes in Attenuating Metabolic Consequences of Night Shift Work Using Rat Model. Nat Sci Sleep 2020; 12:1053-1065. [PMID: 33244284 PMCID: PMC7685379 DOI: 10.2147/nss.s271318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/04/2020] [Indexed: 01/05/2023] Open
Abstract
PURPOSE To study the effects of chronic-simulated night shift work using the rat model and examines if a particular sleep supplement mode could be better in alleviating the effects. METHODS The male Wistar rats were randomly divided into the control (CTL: 8 rats) and night shift work (NW: 24 rats) groups of rats. Based on the sleep supplement strategy, the NW group was further segregated into three subgroups (8 rats each); late sleep supplement group (LSS), early sleep supplement group (ESS), and intermittent sleep supplement group (ISS). Sleep deprivation was achieved using the standard small-platform-over water method. Parameters such as animal body weight and food intake were measured daily. The intraperitoneal glucose tolerance test, fasting plasma insulin concentration, insulin resistance index and insulin sensitivity were measured twice, in the 4th and 8th weeks of the study. Plasma corticosterone concentration and pathological changes in islets (insulitis) were measured at the end of the 8th week. RESULTS In NW group, night work resulted in a gain of body weight and albeit lower than that of the CTL group. NW rats also had higher food intake, showed impaired glucose metabolism and higher plasma corticosterone concentration. The sleep supplement experiments suggested that compared to the other modes, intermittent sleep supplement had significantly low changes in the body weight, glucose metabolism and the islet cells. CONCLUSION Similar to previous studies, we also found that night shift work adversely impacts the body weight and glucose metabolism in rats. However, upon evaluating different sleep supplement strategies, we found the intermittent sleep supplement strategy to be most effective.
Collapse
Affiliation(s)
- Pei-Pei Zheng
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Peking University Fifth School of Clinical Medicine, Beijing 100730, People's Republic of China
| | - Li-Na Zhang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China
| | - Jie Zhang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China
| | - Xin-Miao Chang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China
| | - Shan Ding
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China
| | - Fei Xiao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Bejing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Li-Xin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China
| |
Collapse
|
28
|
The associations between different sleep patterns and osteoporosis based on the Osteoporosis Self-Assessment Tool for Asians. Arch Osteoporos 2020; 15:164. [PMID: 33070238 PMCID: PMC7568695 DOI: 10.1007/s11657-020-00828-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/25/2020] [Indexed: 02/03/2023]
Abstract
UNLABELLED Based on the use of Osteoporosis Self-Assessment Tool for Asians (OSTA) to assess osteoporosis risk, we found that short sleep duration and taking a daytime nap had an increased risk of osteoporosis. PURPOSE To explore the associations between different sleep patterns with osteoporosis. METHODS 3659 postmenopausal women (average age of 60 years) were divided into low, middle, and high osteoporosis risk categories based on the Osteoporosis Self-Assessment Tool for Asians (OSTA). After having collected by a standard questionnaire, total and nocturnal sleep duration was collapsed to form categories of ≤ 6 h, > 6 h and ≤ 7 h, > 7 h and ≤ 8 h, > 8 h and ≤ 9 h, > 9 h, and daytime nap duration of 0 h and > 0 h. RESULTS As a categorical variable, the total sleep duration of ≤ 6 h per day (OR = 1.34, 95% CI 1.04-1.72), nocturnal sleep duration of ≤ 6 h per night (OR = 1.65, 95% CI 1.24-2.18), and taking a daytime nap (OR = 1.33, 95% CI 1.09-1.64) had higher osteoporosis risk after adjustment for covariates. As a continuous variable, after the adjustment for covariates, both longer total (OR = 0.86, 95% CI 0.78-0.94) and nocturnal sleep duration (OR = 0.83, 95% CI 0.76-0.91) had lower risk of osteoporosis risk while taking longer daytime nap (OR = 1.10, 95% CI 1.02-1.19) had higher osteoporosis risk. CONCLUSIONS Postmenopausal women with both short total and nocturnal sleep duration (6 h or less) and taking a daytime nap had increased osteoporosis risk as assessed by OSTA.
Collapse
|
29
|
Laudisio A, Giovannini S, Finamore P, Navarini L, Margiotta DPE, Vannetti F, Macchi C, Coraci D, Imbimbo I, Molino-Lova R, Loreti C, Antonelli Incalzi R, Zuccalà G, Padua L, the Mugello Study Working Group BonaccorsiGuglielmoBoniRobertaCastagnolIChiaraCecchiFrancescaCesariFrancescaEpifaniFrancescoFrandiRobertaGiustiBettiLuisiMaria Luisa ElianaMarcucciRossellaMolino-LovaRaffaelloPaperiniAnitaRazzoliniLorenzoSofiFrancescoTurcanNonaValecchiDebora. Metabolic syndrome is associated with better quality of sleep in the oldest old: results from the "Mugello Study". Diabetol Metab Syndr 2020; 12:46. [PMID: 32742311 PMCID: PMC7386243 DOI: 10.1186/s13098-020-00554-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/15/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND AIMS Reduced sleep quality is common in advanced age. Poor sleep quality is associated with adverse outcomes, chiefly cardiovascular, in young and middle-aged subjects, possibly because of its association with metabolic syndrome (MetS). However, the correlates of sleep quality in oldest populations are unknown. We evaluated the association of sleep quality with MetS in a cohort of subjects aged 90+. METHODS AND RESULTS We analysed data of 343 subjects aged 90+ living in the Mugello area (Tuscany, Italy). Quality of sleep was assessed using the Pittsburgh Sleep Quality Assessment Index (PSQI). Good quality of sleep was defined by a PSQI score < 5. MetS was diagnosed according to the National Cholesterol Education Program's ATP-III criteria; 83 (24%) participants reported good quality of sleep. MetS was diagnosed in 110 (24%) participants. In linear and logistic models, MetS was inversely associated with PSQI score ((B = - 1.04; 95% CI - 2.06 to - .03; P = .044), with increased probability of good sleep quality (OR = 2.52; 95% CI 1.26-5.02; P = .009), and with a PSQI below the median (OR = 2.11; 95% CI 1.11-3.40, P = .022), after adjusting. None of the single components of MetS were associated with PSQI (all P values > .050). However, an increasing number of MetS components was associated with increasing probability of good quality of sleep (P for trend = .002), and of PSQI below the median (P for trend = .007). Generalized Additive Model analysis documented no smoothing function suggestive of nonlinear association between PSQI and MetS. CONCLUSION Our results confirm a high prevalence of poor sleep quality in oldest age; however, in these subjects, MetS seems to be associated with better sleep quality. Additional larger, dedicated studies are required to confirm our results, and, if so, to identify the subsystems involved and the potential therapeutic implications of such an association.
Collapse
Affiliation(s)
- Alice Laudisio
- Unit of Geriatrics, Department of Medicine, Campus Bio-Medico di Roma University, Rome, Italy
| | - Silvia Giovannini
- Department of Geriatrics, Neurosciences and Orthopaedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Panaiotis Finamore
- Unit of Geriatrics, Department of Medicine, Campus Bio-Medico di Roma University, Rome, Italy
| | - Luca Navarini
- Unit of Allergology, Immunology and Rheumatology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | | | | | | | - Daniele Coraci
- Department of Geriatrics, Neurosciences and Orthopaedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | | | - Claudia Loreti
- Department of Geriatrics, Neurosciences and Orthopaedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Giuseppe Zuccalà
- Department of Geriatrics, Neurosciences and Orthopaedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Padua
- Department of Geriatrics, Neurosciences and Orthopaedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | | |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW This review summarizes key findings linking insomnia, short sleep duration, and cardiovascular health. RECENT FINDINGS Early studies associations between insomnia with short sleep and cardiovascular disease Recent studies have incorporated objective data to assess sleep and identify comorbid sleep disorders (e.g. sleep apnea). Use of objective metrics has facilitated understanding of the impacts of insufficient sleep on autonomic dysregulation, metabolic syndrome, coronary artery disease and overall cardiovascular mortality. Emerging research suggests treatment of insomnia (CBT-I) may be beneficial in terms of reducing cardiovascular disease risk. SUMMARY From short term effects on the autonomic nervous system to lasting effects on metabolic syndrome and coronary artery disease, there is growing evidence to support a physiologic pathway by which insomnia with short sleep contributes to cardiovascular disease. More research is needed to understand the effect of insomnia treatment on cardiovascular risk.
Collapse
Affiliation(s)
| | - Jennifer L. Martin
- VA Greater Los Angeles Healthcare System, University of California, Los Angeles, 16111 Plummer St #200, North Hills, CA 91343
| |
Collapse
|
31
|
Effects of acute lying and sleep deprivation on the behavior of lactating dairy cows. PLoS One 2019; 14:e0212823. [PMID: 31461439 PMCID: PMC6713338 DOI: 10.1371/journal.pone.0212823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 08/14/2019] [Indexed: 11/19/2022] Open
Abstract
The objective was to determine the effects of sleep or lying deprivation on the behavior of dairy cows. Data were collected from 8 multi- and 4 primiparous cows (DIM = 199 ± 44 (mean ± SD); days pregnant = 77 ± 30). Using a crossover design, each cow experienced: 1) sleep deprivation implemented by noise or physical contact when their posture suggested sleep, and 2) lying deprivation imposed by a grid placed on the pen floor. One day before treatment (baseline), and treatment day (treatment) were followed by a 12-d washout period (with the first 7 d used to evaluate recovery). Study days were organized from 2100 to 2059. During habituation (d -3 and -2 before treatment), baseline (d -1), and trt (d 0), housing was individual boxstalls (mattress with no bedding). After treatment, cows returned to sand-bedded freestalls for a 7-d recovery period (d 1 to 7) where data on lying behaviors were collected. Following the recovery period, an additional 5-d period was provided to allow the cows a 12-d period between exposures to treatments. Daily lying time, number lying bouts, bout duration, and number of steps were recorded by dataloggers attached to the hind leg of cows throughout the study period. Data were analyzed using a mixed model including fixed effects of treatment (sleep deprivation vs. sleep and lying deprivation), day, and their interaction with significant main effects separated using a PDIFF statement (P ≤ 0.05). Interactions between treatment and day were detected for daily lying time and the number of bouts. Lying time was lower for both treatments during the treatment period compared to baseline. Lying time increased during the recovery period for both lying and sleep deprived cows. However, it took 4 d for the lying deprived cows to fully recover their lying time after treatment, whereas it took the sleep deprived cows 2 d for their lying time to return to baseline levels. Results suggest that both sleep and lying deprivation can have impact cow behavior. Management factors that limit freestall access likely reduce lying time and sleep, causing negative welfare implications for dairy cows.
Collapse
|
32
|
Konkle ATM, Keith SE, McNamee JP, Michaud D. Chronic noise exposure in the spontaneously hypertensive rat. Noise Health 2019; 19:213-221. [PMID: 28937015 PMCID: PMC5644380 DOI: 10.4103/nah.nah_15_17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Introduction: Epidemiological studies have suggested an association between the relative risk for developing cardiovascular disease (CVD) and long-term exposure to elevated levels of transportation noise. The contention is that this association is largely owing to an increase in stress-related biomarkers that are thought to be associated with CVD. Animal models have demonstrated that acute noise exposure is capable of triggering a stress response; however, similar studies using chronic noise models are less common. Materials and Methods: The current study assessed the effects of intermittent daily exposure to broadband 80 kHz bandwidth noise of 87.3 dBA for a period of 21 consecutive days in spontaneously hypertensive rats. Results: Twenty-one days of exposure to noise significantly reduced body weight relative to the sham and unhandled control groups; however, noise had no statistically significant impact on plasma adrenocorticotropic hormone (or adrenal gland weights). Noise was associated with a significant, albeit modest, increase in both corticosterone and aldosterone concentrations following the 21 days of exposure. Interleukin 1 and interleukin 6 levels were unchanged in the noise group, whereas both tumour necrosis factor alpha and C-reactive protein were significantly reduced in noise exposed rats. Tail blood sampling for corticosterone throughout the exposure period showed no appreciable difference between the noise and sham exposed animals, largely due to the sizeable variation for each group as well as the observed fluctuations over time. Discussion: The current pilot study provides only modest support that chronic noise may promote stress-related biological and/or developmental effects. More research is required to verify the current findings and resolve some of the unexpected observations.
Collapse
Affiliation(s)
- Anne T M Konkle
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario; School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Stephen E Keith
- Healthy Environments and Consumer Safety Branch, Environmental and Radiation Health Sciences Directorate, Consumer and Clinical Radiation Protection Bureau, Health , Ottawa, Ontario, Canada
| | - James P McNamee
- Healthy Environments and Consumer Safety Branch, Environmental and Radiation Health Sciences Directorate, Consumer and Clinical Radiation Protection Bureau, Health , Ottawa, Ontario, Canada
| | - David Michaud
- Healthy Environments and Consumer Safety Branch, Environmental and Radiation Health Sciences Directorate, Consumer and Clinical Radiation Protection Bureau, Health , Ottawa, Ontario, Canada
| |
Collapse
|
33
|
Besedovsky L, Lange T, Haack M. The Sleep-Immune Crosstalk in Health and Disease. Physiol Rev 2019; 99:1325-1380. [PMID: 30920354 PMCID: PMC6689741 DOI: 10.1152/physrev.00010.2018] [Citation(s) in RCA: 787] [Impact Index Per Article: 131.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 02/08/2023] Open
Abstract
Sleep and immunity are bidirectionally linked. Immune system activation alters sleep, and sleep in turn affects the innate and adaptive arm of our body's defense system. Stimulation of the immune system by microbial challenges triggers an inflammatory response, which, depending on its magnitude and time course, can induce an increase in sleep duration and intensity, but also a disruption of sleep. Enhancement of sleep during an infection is assumed to feedback to the immune system to promote host defense. Indeed, sleep affects various immune parameters, is associated with a reduced infection risk, and can improve infection outcome and vaccination responses. The induction of a hormonal constellation that supports immune functions is one likely mechanism underlying the immune-supporting effects of sleep. In the absence of an infectious challenge, sleep appears to promote inflammatory homeostasis through effects on several inflammatory mediators, such as cytokines. This notion is supported by findings that prolonged sleep deficiency (e.g., short sleep duration, sleep disturbance) can lead to chronic, systemic low-grade inflammation and is associated with various diseases that have an inflammatory component, like diabetes, atherosclerosis, and neurodegeneration. Here, we review available data on this regulatory sleep-immune crosstalk, point out methodological challenges, and suggest questions open for future research.
Collapse
Affiliation(s)
- Luciana Besedovsky
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| | - Tanja Lange
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| | - Monika Haack
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
34
|
Cuddapah VA, Zhang SL, Sehgal A. Regulation of the Blood-Brain Barrier by Circadian Rhythms and Sleep. Trends Neurosci 2019; 42:500-510. [PMID: 31253251 PMCID: PMC6602072 DOI: 10.1016/j.tins.2019.05.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 01/09/2023]
Abstract
The blood-brain barrier (BBB) is an evolutionarily conserved, structural, and functional separation between circulating blood and the central nervous system (CNS). By controlling permeability into and out of the nervous system, the BBB has a critical role in the precise regulation of neural processes. Here, we review recent studies demonstrating that permeability at the BBB is dynamically controlled by circadian rhythms and sleep. An endogenous circadian rhythm in the BBB controls transporter function, regulating permeability across the BBB. In addition, sleep promotes the clearance of metabolites along the BBB, as well as endocytosis across the BBB. Finally, we highlight the implications of this regulation for diseases, including epilepsy.
Collapse
Affiliation(s)
- Vishnu Anand Cuddapah
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shirley L Zhang
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Siervo GEML, Ogo FM, Staurengo-Ferrari L, Anselmo-Franci JA, Cunha FQ, Cecchini R, Guarnier FA, Verri WA, Fernandes GSA. Sleep restriction during peripuberty unbalances sexual hormones and testicular cytokines in rats. Biol Reprod 2019; 100:112-122. [PMID: 30010983 DOI: 10.1093/biolre/ioy161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022] Open
Abstract
Spermatogenesis and steroidogenesis are not fully established during puberty. Especially during this period, children and adolescents may be chronically sleep deprived due to early school hours and constant exposure to artificial light and interactive activities. We have previously shown that sleep restriction (SR) during peripuberty impairs sperm motility and has consequences on epididymal development in rats. Thus, this study aimed to evaluate the effect of SR during peripuberty on sexual hormones and its impact on testicular tissue. Rats were subjected to 18 h of SR per day for 21 days or were maintained as controls (C) in the same room. The circulating luteinizing hormone levels were decreased in SR rats without changes in the follicle stimulating hormone levels. Plasma and intratesticular testosterone and corticosterone in the SR group were increased in relation to C group. These alterations impair testicular tissue, with decreased IL-1β, IL-6, and TNFα levels in the testis and diminished seminiferous epithelium height and Sertoli cell number. SR also increased testicular lipid peroxidation with no alteration in antioxidant profiles. There were no significant changes in sperm parameters, seminiferous tubule diameter, histopathology, spermatogenesis kinetics, neutrophil and macrophage recruitment, and IL-10 concentration. Our results show that SR unbalances sexual hormones and testicular cytokines at a critical period of sexual maturation. These changes lead to lipid peroxidation in the testes and negatively influence the testicular tissue, as evidenced by diminished seminiferous epithelium height-with apoptosis of germinative cell-and Sertoli cell number.
Collapse
Affiliation(s)
- Gláucia E M L Siervo
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil.,Department of General Biology, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Fernanda M Ogo
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil.,Department of General Biology, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Larissa Staurengo-Ferrari
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Janete A Anselmo-Franci
- Department of Morphology, Stomatology and Physiology, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rubens Cecchini
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Flávia A Guarnier
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Waldiceu A Verri
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Glaura S A Fernandes
- Department of General Biology, Biological Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| |
Collapse
|
36
|
Sever O, Kezirian EJ, Gillett E, Davidson Ward SL, Khoo M, Perez IA. Association between REM sleep and obstructive sleep apnea in obese and overweight adolescents. Sleep Breath 2018; 23:645-650. [PMID: 30554324 DOI: 10.1007/s11325-018-1768-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/27/2018] [Accepted: 12/04/2018] [Indexed: 12/30/2022]
Abstract
PURPOSE Overweight and obese children have demonstrated reduced rapid eye movement (REM) sleep, affecting energy balance regulation and predisposition to weight gain. Obstructive sleep apnea (OSA) is a known cause of decreased REM sleep. The purpose of this study is to examine the association between the percentage of REM sleep, BMI z-score, and OSA severity in overweight and obese adolescents. METHODS We performed a cross-sectional study of 92 (43% female) overweight and obese adolescents (13-17 years old) who underwent overnight polysomnography (PSG) at Children's Hospital Los Angeles between 2010 and 2017. RESULTS The average Body Mass Index (BMI) z-score was 2.27 ± 0.47, with 71% having BMI z-score ≥ 2. REM% during PSG was 15.6 ± 6.8, and obstructive apnea-hypopnea index was 17.1 ± 24.3. The distribution across categories of OSA severity was 27% none (≤ 1.5 events/h), 24% mild (> 1.5-5 events/h), 8% moderate (> 5-10 events/h), and 41% severe (> 10 events/h). REM% was not associated with BMI z-score, either on univariate or multivariate regression with adjustment for age, gender, and apnea-hypopnea index (AHI). When subdivided into OSA categories, a 1-unit increase in BMI z-score was associated with a 5.96 (p = 0.03) increase in REM% in mild OSA and an 8.86 (p = 0.02) decrease in REM% in severe OSA. There was no association between BMI z-score and REM% in none and moderate OSA. CONCLUSION Among overweight and obese adolescents, BMI z-score was associated with decreased REM% in severe OSA and unexpectedly increased REM% in mild OSA, but there was no association in none or moderate OSA.
Collapse
Affiliation(s)
- Orna Sever
- Children's Hospital Los Angeles, 4650 Sunset Blvd., #83, Los Angeles, CA, USA
- Keck School of Medicine of USC, Los Angeles, CA, USA
| | | | - Emily Gillett
- Children's Hospital Los Angeles, 4650 Sunset Blvd., #83, Los Angeles, CA, USA
- Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Sally L Davidson Ward
- Children's Hospital Los Angeles, 4650 Sunset Blvd., #83, Los Angeles, CA, USA
- Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Michael Khoo
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Iris A Perez
- Children's Hospital Los Angeles, 4650 Sunset Blvd., #83, Los Angeles, CA, USA.
- Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
37
|
|
38
|
Brianza-Padilla M, Sánchez-Muñoz F, Vázquez-Palacios G, Huang F, Almanza-Pérez JC, Bojalil R, Bonilla-Jaime H. Cytokine and microRNA levels during different periods of paradoxical sleep deprivation and sleep recovery in rats. PeerJ 2018; 6:e5567. [PMID: 30225174 PMCID: PMC6139242 DOI: 10.7717/peerj.5567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/13/2018] [Indexed: 01/19/2023] Open
Abstract
Background Sleep has a fundamental role in the regulation of homeostasis. The aim of this study was to assess the effect of different periods of paradoxical sleep deprivation (PSD) and recovery on serum levels of cytokines and miRNAs related to inflammatory responses. Methods Male Wistar rats were submitted to a PSD of 24, 96, or 192 h, or of 192 h followed by 20 days of recovery (192 h PSD+R). The concentrations of corticosterone, cytokines (IL-6, TNF, IL-10, Adiponectin) and miRNAs (miR-146a, miR-155, miR-223, miR-16, miR-126, miR-21) in serum were evaluated. Results At PSD 24 h a significant increase of IL-6 and decrease of IL-10 were observed. At PSD 96h adiponectin increased. At 192 h of PSD IL-6 increased significantly again, accompanied by a threefold increase of IL-10 and an increase of serum corticosterone. After 20 days of recovery (192 h PSD+R) corticosterone, IL-6 and TNF levels increased significantly, while IL-10 decreased also significantly. Regarding the miRNAs at 24 h of PSD serum miR-146a, miR-155, miR-223, and miR-16 levels all increased. At 96 h of PSD miR-223 decreased. At 192 h of PSD decreases in miR-16 and miR-126 were observed. After recovery serum miR-21 increased and miR-16 decreased. Conclusion PSD induces a dynamic response likely reflecting the induced cellular stress and manifested as variating hormonal and inflammatory responses. Sleep deprivation disturbed corticosterone, cytokine and miRNA levels in serum related to the duration of sleep deprivation, as short-term PSD produced effects similar to those of an acute inflammatory response and long-term PSD induced long-lasting disturbances of biological mediators.
Collapse
Affiliation(s)
- Malinalli Brianza-Padilla
- Posgrado en Biologia Experimental, División de Ciencias Biológicas y de la Salud, Universidad Autonoma Metropolitana Iztapalapa, Ciudad de Mexico, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiologia Ignacio Chavez, Ciudad de Mexico, Mexico
| | - Gonzalo Vázquez-Palacios
- Colegio de Ciencias y Humanidades, Universidad Autonoma de la Ciudad de Mexico, Ciudad de Mexico, Mexico
| | - Fengyang Huang
- Laboratorio de Investigación en Farmacología y Toxicología, Hospital Infantil de Mexico Federico Gomez, Ciudad de Mexico, Mexico
| | - Julio César Almanza-Pérez
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonoma Metropolitana Iztapalapa, Ciudad de Mexico, Mexico
| | - Rafael Bojalil
- Departamento de Inmunología, Instituto Nacional de Cardiologia Ignacio Chavez, Ciudad de Mexico, Mexico.,Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Xochimilco, Ciudad de México, México
| | - Herlinda Bonilla-Jaime
- Departamento de Biologia de la Reproducción, División de Ciencias Biológicas y de la Salud, Universidad Autonoma Metropolitana Iztapalapa, Ciudad de Mexico, Mexico
| |
Collapse
|
39
|
Palnitkar G, Phillips CL, Hoyos CM, Marren AJ, Bowman MC, Yee BJ. Linking sleep disturbance to idiopathic male infertility. Sleep Med Rev 2018; 42:149-159. [PMID: 30377037 DOI: 10.1016/j.smrv.2018.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 06/13/2018] [Accepted: 07/26/2018] [Indexed: 12/22/2022]
Abstract
Recently published data suggests that male fertility has declined over the past four decades. The reasons for the decline are unclear with up to 50% of cases of male infertility remaining unexplained (idiopathic male infertility). Whilst environmental factors and rising rates of obesity have been implicated, there is now growing evidence that sleep disturbance may be an independent causative factor. Indeed, the prevalence of sleep disturbance appears to be increasing in parallel with deterioration in population sperm quality, a commonly used surrogate marker of male fertility. Although there is some understanding of the relationship between sleep, gonadal hormone secretion and sexual function, it remains to be seen whether sleep disturbance is implicated in idiopathic male infertility. This review will detail the current evidence supporting a link between sleep disturbance and male infertility. Potential mechanistic pathways will be proposed and evidence supporting these pathways will be discussed. Further research is needed in clarifying links between sleep disturbance and idiopathic male infertility. At present the only available treatment option for men with idiopathic infertility is assisted reproductive technology. Demonstration of a causative link between sleep disturbance and idiopathic male infertility may in the future lead to additional treatment options in selected cases.
Collapse
Affiliation(s)
- Gaurie Palnitkar
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia; Discipline of Sleep Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia.
| | - Craig L Phillips
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia; Discipline of Sleep Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, St Leonards, Sydney, New South Wales, Australia
| | - Camilla M Hoyos
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia; School of Psychology, Faculty of Science, University of Sydney, New South Wales, Australia
| | - Anthony J Marren
- Department of Reproductive Endocrinology and Infertility, Royal Prince Alfred Hospital for Women and Babies, Camperdown, New South Wales, Australia; Genea Fertility, Sydney, New South Wales, Australia; Queen Elizabeth II Research Institute for Mothers and Infants, Department of Obstetrics, Gynaecology and Neonatology, The University of Sydney, New South Wales, Australia
| | - Mark C Bowman
- Department of Reproductive Endocrinology and Infertility, Royal Prince Alfred Hospital for Women and Babies, Camperdown, New South Wales, Australia; Genea Fertility, Sydney, New South Wales, Australia; Department of Obstetrics and Gynaecology, University of Sydney, New South Wales, Australia
| | - Brendon J Yee
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia; Discipline of Sleep Medicine, Sydney Medical School, University of Sydney, New South Wales, Australia; Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Lyamin OI, Kosenko PO, Korneva SM, Vyssotski AL, Mukhametov LM, Siegel JM. Fur Seals Suppress REM Sleep for Very Long Periods without Subsequent Rebound. Curr Biol 2018; 28:2000-2005.e2. [PMID: 29887309 DOI: 10.1016/j.cub.2018.05.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/26/2018] [Accepted: 05/08/2018] [Indexed: 11/17/2022]
Abstract
Virtually all land mammals and birds have two sleep states: slow-wave sleep (SWS) and rapid eye movement (REM) sleep [1, 2]. After deprivation of REM sleep by repeated awakenings, mammals increase REM sleep time [3], supporting the idea that REM sleep is homeostatically regulated. Some evidence suggests that periods of REM sleep deprivation for a week or more cause physiological dysfunction and eventual death [4, 5]. However, separating the effects of REM sleep loss from the stress of repeated awakening is difficult [2, 6]. The northern fur seal (Callorhinus ursinus) is a semiaquatic mammal [7]. It can sleep on land and in seawater. The fur seal is unique in showing both the bilateral SWS seen in most mammals and the asymmetric sleep previously reported in cetaceans [8]. Here we show that when the fur seal stays in seawater, where it spends most of its life [7], it goes without or greatly reduces REM sleep for days or weeks. After this nearly complete elimination of REM, it displays minimal or no REM rebound upon returning to baseline conditions. Our data are consistent with the hypothesis that REM sleep may serve to reverse the reduced brain temperature and metabolism effects of bilateral nonREM sleep, a state that is greatly reduced when the fur seal is in the seawater, rather than REM sleep being directly homeostatically regulated. This can explain the absence of REM sleep in the dolphin and other cetaceans and its increasing proportion as the end of the sleep period approaches in humans and other mammals.
Collapse
Affiliation(s)
- Oleg I Lyamin
- Department of Psychiatry and Biobehavioral Sciences and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA; A.N. Severtsov Institute of Ecology and Evolution, RAS, Moscow, Russia; Utrish Dolphinarium, Moscow, Russia.
| | | | | | - Alexei L Vyssotski
- Institute of Neuroinformatics, University of Zurich and Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Lev M Mukhametov
- A.N. Severtsov Institute of Ecology and Evolution, RAS, Moscow, Russia; Utrish Dolphinarium, Moscow, Russia
| | - Jerome M Siegel
- Department of Psychiatry and Biobehavioral Sciences and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Shift-work: is time of eating determining metabolic health? Evidence from animal models. Proc Nutr Soc 2018; 77:199-215. [DOI: 10.1017/s0029665117004128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The circadian disruption in shift-workers is suggested to be a risk factor to develop overweight and metabolic dysfunction. The conflicting time signals given by shifted activity, shifted food intake and exposure to light at night occurring in the shift-worker are proposed to be the cause for the loss of internal synchrony and the consequent adverse effects on body weight and metabolism. Because food elicited signals have proven to be potent entraining signals for peripheral oscillations, here we review the findings from experimental models of shift-work and verify whether they provide evidence about the causal association between shifted feeding schedules, circadian disruption and altered metabolism. We found mainly four experimental models that mimic the conditions of shift-work: protocols of forced sleep deprivation, of forced activity during the normal rest phase, exposure to light at night and shifted food timing. A big variability in the intensity and duration of the protocols was observed, which led to a diversity of effects. A common result was the disruption of temporal patterns of activity; however, not all studies explored the temporal patterns of food intake. According to studies that evaluate time of food intake as an experimental model of shift-work and studies that evaluate shifted food consumption, time of food intake may be a determining factor for the loss of balance at the circadian and metabolic level.
Collapse
|
42
|
Tumor necrosis factor alpha in sleep regulation. Sleep Med Rev 2017; 40:69-78. [PMID: 29153862 DOI: 10.1016/j.smrv.2017.10.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/16/2017] [Accepted: 10/18/2017] [Indexed: 12/14/2022]
Abstract
This review details tumor necrosis factor alpha (TNF) biology and its role in sleep, and describes how TNF medications influence sleep/wake activity. Substantial evidence from healthy young animals indicates acute enhancement or inhibition of endogenous brain TNF respectively promotes and inhibits sleep. In contrast, the role of TNF in sleep in most human studies involves pathological conditions associated with chronic elevations of systemic TNF and disrupted sleep. Normalization of TNF levels in such patients improves sleep. A few studies involving normal healthy humans and their TNF levels and sleep are consistent with the animal studies but are necessarily more limited in scope. TNF can act on established sleep regulatory circuits to promote sleep and on the cortex within small networks, such as cortical columns, to induce sleep-like states. TNF affects multiple synaptic functions, e.g., its role in synaptic scaling is firmly established. The TNF-plasticity actions, like its role in sleep, can be local network events suggesting that sleep and plasticity share biochemical regulatory mechanisms and thus may be inseparable from each other. We conclude that TNF is involved in sleep regulation acting within an extensive tightly orchestrated biochemical network to niche-adapt sleep in health and disease.
Collapse
|
43
|
Yujra VQ, Antunes HKM, Mônico‐Neto M, Pisani LP, Santamarina AB, Quintana HT, de Oliveira F, Oshima CTF, Ribeiro DA. Sleep deprivation induces pathological changes in rat masticatory muscles: Role of Toll like signaling pathway and atrophy. J Cell Biochem 2017; 119:2269-2277. [DOI: 10.1002/jcb.26389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 08/23/2017] [Indexed: 11/10/2022]
Affiliation(s)
- Veronica Q. Yujra
- Department of PathologyFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Hanna K. M. Antunes
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Marcos Mônico‐Neto
- Department of PshychobiologyFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Luciana P. Pisani
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Aline B. Santamarina
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Hananiah T. Quintana
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Flavia de Oliveira
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Celina T. F. Oshima
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Daniel A. Ribeiro
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| |
Collapse
|
44
|
Zhao Z, Zhao X, Veasey SC. Neural Consequences of Chronic Short Sleep: Reversible or Lasting? Front Neurol 2017; 8:235. [PMID: 28620347 PMCID: PMC5449441 DOI: 10.3389/fneur.2017.00235] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/12/2017] [Indexed: 12/22/2022] Open
Abstract
Approximately one-third of adolescents and adults in developed countries regularly experience insufficient sleep across the school and/or work week interspersed with weekend catch up sleep. This common practice of weekend recovery sleep reduces subjective sleepiness, yet recent studies demonstrate that one weekend of recovery sleep may not be sufficient in all persons to fully reverse all neurobehavioral impairments observed with chronic sleep loss, particularly vigilance. Moreover, recent studies in animal models demonstrate persistent injury to and loss of specific neuron types in response to chronic short sleep (CSS) with lasting effects on sleep/wake patterns. Here, we provide a comprehensive review of the effects of chronic sleep disruption on neurobehavioral performance and injury to neurons, astrocytes, microglia, and oligodendrocytes and discuss what is known and what is not yet established for reversibility of neural injury. Recent neurobehavioral findings in humans are integrated with animal model research examining long-term consequences of sleep loss on neurobehavioral performance, brain development, neurogenesis, neurodegeneration, and connectivity. While it is now clear that recovery of vigilance following short sleep requires longer than one weekend, less is known of the impact of CSS on cognitive function, mood, and brain health long term. From work performed in animal models, CSS in the young adult and short-term sleep loss in critical developmental windows can have lasting detrimental effects on neurobehavioral performance.
Collapse
Affiliation(s)
- Zhengqing Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiangxiang Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Sigrid C Veasey
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
45
|
da Silva Rocha-Lopes J, Machado RB, Suchecki D. Chronic REM Sleep Restriction in Juvenile Male Rats Induces Anxiety-Like Behavior and Alters Monoamine Systems in the Amygdala and Hippocampus. Mol Neurobiol 2017; 55:2884-2896. [DOI: 10.1007/s12035-017-0541-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/07/2017] [Indexed: 10/19/2022]
|
46
|
Yin M, Chen Y, Zheng H, Pu T, Marshall C, Wu T, Xiao M. Assessment of mouse cognitive and anxiety-like behaviors and hippocampal inflammation following a repeated and intermittent paradoxical sleep deprivation procedure. Behav Brain Res 2017; 321:69-78. [DOI: 10.1016/j.bbr.2016.12.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/21/2016] [Accepted: 12/25/2016] [Indexed: 12/12/2022]
|
47
|
Hurtado-Alvarado G, Domínguez-Salazar E, Pavon L, Velázquez-Moctezuma J, Gómez-González B. Blood-Brain Barrier Disruption Induced by Chronic Sleep Loss: Low-Grade Inflammation May Be the Link. J Immunol Res 2016; 2016:4576012. [PMID: 27738642 PMCID: PMC5050358 DOI: 10.1155/2016/4576012] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 08/14/2016] [Accepted: 08/24/2016] [Indexed: 12/16/2022] Open
Abstract
Sleep is a vital phenomenon related to immunomodulation at the central and peripheral level. Sleep deficient in duration and/or quality is a common problem in the modern society and is considered a risk factor to develop neurodegenerative diseases. Sleep loss in rodents induces blood-brain barrier disruption and the underlying mechanism is still unknown. Several reports indicate that sleep loss induces a systemic low-grade inflammation characterized by the release of several molecules, such as cytokines, chemokines, and acute-phase proteins; all of them may promote changes in cellular components of the blood-brain barrier, particularly on brain endothelial cells. In the present review we discuss the role of inflammatory mediators that increase during sleep loss and their association with general disturbances in peripheral endothelium and epithelium and how those inflammatory mediators may alter the blood-brain barrier. Finally, this manuscript proposes a hypothetical mechanism by which sleep loss may induce blood-brain barrier disruption, emphasizing the regulatory effect of inflammatory molecules on tight junction proteins.
Collapse
Affiliation(s)
- G. Hurtado-Alvarado
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - E. Domínguez-Salazar
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - L. Pavon
- Department of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente”, Mexico City, Mexico
| | - J. Velázquez-Moctezuma
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - B. Gómez-González
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| |
Collapse
|
48
|
Brianza-Padilla M, Bonilla-Jaime H, Almanza-Pérez JC, López-López AL, Sánchez-Muñoz F, Vázquez-Palacios G. Effects of different periods of paradoxical sleep deprivation and sleep recovery on lipid and glucose metabolism and appetite hormones in rats. Appl Physiol Nutr Metab 2016; 41:235-43. [DOI: 10.1139/apnm-2015-0337] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Sleep has a fundamental role in the regulation of energy balance, and it is an essential and natural process whose precise impacts on health and disease have not yet been fully elucidated. The aim of this study was to assess the consequences of different periods of paradoxical sleep deprivation (PSD) and recovery from PSD on lipid profile, oral glucose tolerance test (OGTT) results, and changes in insulin, corticosterone, ghrelin, and leptin concentrations. Three-month-old male Wistar rats weighing 250–350 g were submitted to 24, 96, or 192 h of PSD or 192 h of PSD with 480 h of recovery. The PSD was induced by the multiple platforms method. Subsequently, the animals were submitted to an OGTT. One day later, the animals were killed and the levels of triglycerides, total cholesterol, lipoproteins (low-density lipoprotein, very-low-density lipoprotein, and high-density lipoprotein), insulin, ghrelin, leptin, and corticosterone in plasma were quantified. There was a progressive decrease in body weight with increasing duration of PSD. The PSD induced basal hypoglycemia over all time periods evaluated. Evaluation of areas under the curve revealed progressive hypoglycemia only after 96 and 192 h of PSD. There was an increase in corticosterone levels after 192 h of PSD. We conclude that PSD induces alterations in metabolism that are reversed after a recovery period of 20 days.
Collapse
Affiliation(s)
| | - Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Área de Biología Conductual y Reproductiva, Universidad Autónoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco No. 186, Col. Vicentina, CP 09340, Mexico
| | - Julio César Almanza-Pérez
- Departamento de Ciencias de la Salud, Área de Investigación Médica, Universidad Autónoma Metropolitana-Iztapalapa, CP 09340, Mexico
| | - Ana Laura López-López
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, CP 09340, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiologia (Ignacio Chávez), Juan Badiano No. 1, Col. Sección XVI, Del. Tlalpan, CP 14080, Mexico
| | - Gonzalo Vázquez-Palacios
- Colegio de Ciencias y Humanidades, Universidad Autónoma de la Ciudad de México-San Lorenzo Tezonco, Av. Prolongación San Isidro No. 151, Col. San Lorenzo Tezonco, Del. Iztapalapa, CP 09790, Mexico
| |
Collapse
|
49
|
Pardo GVE, Goularte JF, Hoefel AL, de Castro AL, Kucharski LC, da Rosa Araujo AS, Lucion AB. Effects of sleep restriction during pregnancy on the mother and fetuses in rats. Physiol Behav 2016; 155:66-76. [DOI: 10.1016/j.physbeh.2015.11.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/29/2015] [Accepted: 11/30/2015] [Indexed: 01/15/2023]
|
50
|
Ribeiro-Silva N, Nejm MB, da Silva SMA, Suchecki D, Luz J. Restriction of rapid eye movement sleep during adolescence increases energy gain and metabolic efficiency in young adult rats. Exp Physiol 2016; 101:308-18. [PMID: 26663203 DOI: 10.1113/ep085323] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 12/03/2015] [Indexed: 12/15/2022]
Abstract
NEW FINDINGS What is the central question of this study? Sleep curtailment in infancy and adolescence may lead to long-term risk for obesity, but the mechanisms involved have not yet been determined. This study examined the immediate and long-term metabolic effects produced by sleep restriction in young rats. What is the main finding and its importance? Prolonged sleep restriction reduced weight gain (body fat stores) in young animals. After prolonged recovery, sleep-restricted rats tended to save more energy and to store more fat, possibly owing to increased gross food efficiency. This could be the first step to understand this association. Sleep curtailment is associated with obesity and metabolic changes in adults and children. The aim of the present study was to evaluate the immediate and long-term metabolic alterations produced by sleep restriction in pubertal male rats. Male Wistar rats (28 days old) were allocated to a control (CTL) group or a sleep-restricted (SR) group. This was accomplished by the single platform technique for 18 h per day for 21 days. These groups were subdivided into the following four time points for assessment: sleep restriction and 1, 2 and 4 months of recovery. Body weight and food intake were monitored throughout the experiment. At the end of each time period, blood was collected for metabolic profiling, and the carcasses were processed for measurement of body composition and energy balance. During the period of sleep restriction, SR animals consumed less food in the home cages. This group also displayed lower body weight, body fat, triglycerides and glucose levels than CTL rats. At the end of the first month of recovery, despite eating as much as CTL rats, SR animals showed greater energy and body weight gain, increased gross food efficiency and decreased energy expenditure. At the end of the second and fourth months of recovery, the groups were no longer different, except for energy gain and gross food efficiency, which remained higher in SR animals. In conclusion, sleep restriction affected weight gain of young animals, owing to reduction of fat stores. Two months were sufficient to recover this deficit and to reveal that SR rats tended to save more energy and to store more fat.
Collapse
Affiliation(s)
- Neila Ribeiro-Silva
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mariana Bocca Nejm
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Deborah Suchecki
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jacqueline Luz
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|