1
|
Zhang B, Tang M, Li X. A narrative review of sleep and breast cancer: from epidemiology to mechanisms. Cancer Causes Control 2025; 36:457-472. [PMID: 39731679 DOI: 10.1007/s10552-024-01951-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
Breast cancer is the leading cause of cancer-related death and the most common cancer among women worldwide. It is crucial to identify potentially modifiable risk factors to intervene and prevent breast cancer effectively. Sleep factors have emerged as a potentially novel risk factor for female breast cancer. Current epidemiologic studies suggest a significant impact of sleep factors on breast cancer. Exposure to abnormal sleep duration, poor sleep quality, sleep disorders, sleep medication use, or night shift work can increase the risk of breast cancer by decreasing melatonin secretion, disrupting circadian rhythm, compromising immune function, or altering hormone levels. However, there are still controversies regarding the epidemiologic association, and the underlying mechanisms have yet to be fully elucidated. This paper summarizes the epidemiologic evidence on the associations between sleep factors, including sleep duration, sleep quality, sleep disorders, sleep medication use, sleep habits, and night shift work, and the development of breast cancer. The potential mechanisms underlying these associations were also reviewed.
Collapse
Affiliation(s)
- Bao Zhang
- Department of Clinical Nutrition, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Mengsha Tang
- School of Humanity and Management, Wannan Medical College, Wuhu, 241002, Anhui, China
| | - Xiude Li
- Department of Clinical Nutrition, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
2
|
Yi ZY, Liang QX, Zhou Q, Yang L, Meng QR, Li J, Lin YH, Cao YP, Zhang CH, Schatten H, Qiao J, Sun QY. Maternal total sleep deprivation causes oxidative stress and mitochondrial dysfunction in oocytes associated with fertility decline in mice. PLoS One 2024; 19:e0306152. [PMID: 39413105 PMCID: PMC11482706 DOI: 10.1371/journal.pone.0306152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/11/2024] [Indexed: 10/18/2024] Open
Abstract
Previous studies have shown sleep deprivation is increasingly reported as one of the causes of female infertility. However, how and by what relevant mechanisms it affects female fertility remains unclear. In this study, female mice underwent 72 hours of total sleep deprivation (TSD) caused by rotating wheel or 2 different controls: a stationary wheel, or forced movement at night. Even though, there was no significant difference in the number of eggs ovulated by the TSD mice compared to the control groups. Overall levels of estrogen and FSH were lower throughout the estrus cycle. A total of 42 genes showed significant differential expression in GV oocytes after TSD by RNA sequencing (RNA-Seq). These included genes were enriched in gene ontology terms of mitochondrial protein complex, oxidoreductase activity, cell division, cell cycle G1/S phase transition, as well as others. The increased concentrations of reactive oxygen species (ROS) in germinal vesicle (GV) and metaphase II (MII) oocytes from TSD mice were observed, which might be induced by impaired mitochondrial function caused by TSD. The GV oocytes displayed increased mitochondrial DNA (mtDNA) copy number and a significant transient increase in inner mitochondrial membrane potential (Δψm) from the TSD mice probably due to compensatory effect. In contrast, MII oocytes in the TSD group showed a decrease in the mtDNA copy number and a lower Δψm compared with the controls. Furthermore, abnormal distribution of mitochondria in the GV and MII oocytes was also observed in TSD mice, suggesting mitochondrial dysfunction. In addition, abnormal spindle and abnormal arrangement of chromosomes in MII oocytes were markedly increased in the TSD mice compared with the control mice. In conclusion, our results suggest that TSD significantly alters the oocyte transcriptome, contributing to oxidative stress and disrupted mitochondrial function, which then resulted in oocyte defects and impaired early embryo development in female mice.
Collapse
Affiliation(s)
- Zi-Yun Yi
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Qiu-Xia Liang
- College of Life Science, Sichuan Agricultural University, Ya’an, China
| | - Qian Zhou
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China
| | - Lin Yang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences Beijing, Beijing, China
| | - Qing-Ren Meng
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jian Li
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yi-hua Lin
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yan-pei Cao
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Chun-Hui Zhang
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Jie Qiao
- Reproductive Medical Center, Peking University Third Hospital, Beijing, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
3
|
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
|
4
|
Tóth A, Traub M, Bencsik N, Détári L, Hajnik T, Dobolyi A. Sleep- and sleep deprivation-related changes of vertex auditory evoked potentials during the estrus cycle in female rats. Sci Rep 2024; 14:5784. [PMID: 38461157 PMCID: PMC10924932 DOI: 10.1038/s41598-024-56392-9] [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: 09/28/2023] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
The estrus cycle in female rodents has been shown to affect a variety of physiological functions. However, little is known about its presumably thorough effect on auditory processing during the sleep-wake cycle and sleep deprivation. Vertex auditory evoked potentials (vAEPs) were evoked by single click tone stimulation and recorded during different stages of the estrus cycle and sleep deprivation performed in metestrus and proestrus in female rats. vAEPs showed a strong sleep-dependency, with the largest amplitudes present during slow wave sleep while the smallest ones during wakefulness. Higher amplitudes and longer latencies were seen in the light phase during all vigilance stages. The largest amplitudes were found during proestrus (light phase) while the shortest latencies were seen during estrus (dark phase) compared to the 2nd day diestrus baseline. High-amplitude responses without latency changes were also seen during metestrus with increased homeostatic sleep drive. More intense and faster processing of auditory information during proestrus and estrus suggesting a more effective perception of relevant environmental cues presumably in preparation for sexual receptivity. A 4-h sleep deprivation resulted in more pronounced sleep recovery in metestrus compared to proestrus without difference in delta power replacement suggesting a better tolerance of sleep deprivation in proestrus. Sleep deprivation decreased neuronal excitability and responsiveness in a similar manner both during metestrus and proestrus, suggesting that the negative consequences of sleep deprivation on auditory processing may have a limited correlation with the estrus cycle stage.
Collapse
Affiliation(s)
- Attila Tóth
- In Vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary.
| | - Máté Traub
- In Vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
| | - Norbert Bencsik
- Cellular Neurobiology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - László Détári
- In Vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
| | - Tünde Hajnik
- In Vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
| | - Arpád Dobolyi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
5
|
Ishikura IA, Hachul H, Tufik S, Andersen ML. Dysmenorrhea and Sleep: A Review. Sleep Med Clin 2023; 18:449-461. [PMID: 38501517 DOI: 10.1016/j.jsmc.2023.06.006] [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] [Indexed: 03/20/2024]
Abstract
This review encompasses the clinical features and relevance to investigate sleep in women with dysmenorrhea. Dysmenorrhea is a prevalent gynecologic dysfunction that affects the social and professional lives of women. It can occur at every menstrual cycle, depending on the cause and psychologic factors. Studies have reported poor sleep and insomnia symptoms in dysmenorrhea condition, which may intensify the dysmenorrhea manifestation and interfere negatively to its treatment. There is an urgent need to identify the main cause of this dysfunction and provide efficient treatments to minimize the detrimental effects of dysmenorrhea in quality of life of these women.
Collapse
Affiliation(s)
- Isabela A Ishikura
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862 - Vila Clementino - 04023062 - São Paulo - SP - Brazil
| | - Helena Hachul
- Departamento de Ginecologia, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, n° 740 - Vila Clementino - 04023-062 - São Paulo - SP - Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862 - Vila Clementino - 04023062 - São Paulo - SP - Brazil
| | - Monica L Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862 - Vila Clementino - 04023062 - São Paulo - SP - Brazil.
| |
Collapse
|
6
|
Kim LJ, Pho H, Anokye-Danso F, Ahima RS, Pham LV, Polotsky VY. The effect of diet-induced obesity on sleep and breathing in female mice. Sleep 2023; 46:zsad158. [PMID: 37262435 PMCID: PMC10424169 DOI: 10.1093/sleep/zsad158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/24/2023] [Indexed: 06/03/2023] Open
Abstract
Obesity and male sex are main risk factors for sleep-disordered breathing (SDB). We have shown that male diet-induced obesity (DIO) mice develop hypoventilation, sleep apnea, and sleep fragmentation. The effects of DIO on breathing and sleep architecture in females have not been investigated. We hypothesized that female mice are less susceptible to the detrimental effects of DIO on sleep and SDB compared to males. Female DIO-C57BL/6J and lean C57BL/6J mice underwent 24-hour metabolic studies and were exposed to 8% CO2 to measure the hypercapnic ventilatory response (HCVR), and sleep studies. Ventilatory response to arousals was calculated as ratio of the average and peak minute ventilation (VE) during each arousal relative to the baseline VE. Breathing stability was measured with Poincaré plots of VE. Female obesity was associated with decreased metabolism, indicated by reduced oxygen consumption (VO2) and CO2 production (VCO2). VE in 8% CO2 and HCVR were significantly attenuated during wakefulness. NREM sleep duration was reduced in DIO mice, but REM sleep was preserved. Ventilation during NREM and REM sleep was augmented compared to lean mice. Arousal frequency was similar between groups. Obesity increased the frequency of spontaneous arousals, whereas the apnea index was 4-fold reduced in DIO compared to lean mice. Obesity decreased pre- and post-apnea arousals. Obese mice had more stable breathing with reduced ventilatory response to arousals, compared to lean females. We conclude that obese female mice are protected against SDB, which appears to be related to an attenuated CO2 responsiveness, compared to the lean state.
Collapse
Affiliation(s)
- Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frederick Anokye-Danso
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luu V Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| |
Collapse
|
7
|
Wright CJ, Milosavljevic S, Pocivavsek A. The stress of losing sleep: Sex-specific neurobiological outcomes. Neurobiol Stress 2023; 24:100543. [PMID: 37252645 PMCID: PMC10209346 DOI: 10.1016/j.ynstr.2023.100543] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Sleep is a vital and evolutionarily conserved process, critical to daily functioning and homeostatic balance. Losing sleep is inherently stressful and leads to numerous detrimental physiological outcomes. Despite sleep disturbances affecting everyone, women and female rodents are often excluded or underrepresented in clinical and pre-clinical studies. Advancing our understanding of the role of biological sex in the responses to sleep loss stands to greatly improve our ability to understand and treat health consequences of insufficient sleep. As such, this review discusses sex differences in response to sleep deprivation, with a focus on the sympathetic nervous system stress response and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We review sex differences in several stress-related consequences of sleep loss, including inflammation, learning and memory deficits, and mood related changes. Focusing on women's health, we discuss the effects of sleep deprivation during the peripartum period. In closing, we present neurobiological mechanisms, including the contribution of sex hormones, orexins, circadian timing systems, and astrocytic neuromodulation, that may underlie potential sex differences in sleep deprivation responses.
Collapse
Affiliation(s)
- Courtney J. Wright
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
8
|
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
|
9
|
Hachul H, Castro LS, Bezerra AG, Pires GN, Poyares D, Andersen ML, Bittencourt LR, Tufik S. Hot flashes, insomnia, and the reproductive stages: a cross-sectional observation of women from the EPISONO study. J Clin Sleep Med 2021; 17:2257-2267. [PMID: 34170233 PMCID: PMC8636363 DOI: 10.5664/jcsm.9432] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 01/04/2023]
Abstract
STUDY OBJECTIVES To investigate the association of hot flashes and insomnia in women in premenopause and postmenopause. METHODS The study was performed using data from the São Paulo Epidemiological Sleep Study. Women in premenopause were classified as having regular menstrual cycles, being anovulatory, or using hormonal contraceptives. Women in menopause were classified as being in perimenopause, early postmenopause, or late postmenopause. Women reporting frequent insomnia symptoms and relevant daytime complaints were classified as having insomnia disorder. Polysomnography alterations suggestive of insomnia were also identified. RESULTS The frequency of hot flashes was 42% among women in postmenopause (mainly in early postmenopause) and 9% among women in premenopause (mainly anovulatory; P < .01). Approximately 18.7% had insomnia disorder, 48% had isolated insomnia symptoms, and 32.4% had polysomnography alterations. Comparing women in menopause with those in premenopause, the diagnosis of insomnia was similar (premenopause: 18.9% vs menopause: 17.5%), but women in menopause had more frequent isolated insomnia symptoms (premenopause: 43.9% vs menopause: 55.9%; P = .02) and polysomnography correlates of insomnia (premenopause: 26.5% vs menopause: 42.6%; P < .01). Hot flashes were more frequent among women with insomnia disorders (25.5%) and with isolated insomnia symptoms (23.0%) when compared with good sleepers (12.6%) in the entire sample (P = .01). Among women in late menopause, the prevalence of hot flashes was higher in both women with insomnia disorders (42.1%) and with isolated insomnia symptoms (37.5%) when compared with women who were good sleepers (14.3%; P = .05). CONCLUSIONS Hot flashes are associated with insomnia and polysomnography alterations suggestive of insomnia. The prevalence of hot flashes among women with insomnia disorder is especially high among women in late postmenopause. CITATION Hachul H, Castro LS, Bezerra AG, et al. Hot flashes, insomnia, and the reproductive stages: a cross-sectional observation of women from the EPISONO study. J Clin Sleep Med. 2021;17(11):2257-2267.
Collapse
Affiliation(s)
- Helena Hachul
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
- Departamento de Ginecologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Gabriel Natan Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Dalva Poyares
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica Levy Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lia Rita Bittencourt
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Murack M, Chandrasegaram R, Smith KB, Ah-Yen EG, Rheaume É, Malette-Guyon É, Nanji Z, Semchishen SN, Latus O, Messier C, Ismail N. Chronic sleep disruption induces depression-like behavior in adolescent male and female mice and sensitization of the hypothalamic-pituitary-adrenal axis in adolescent female mice. Behav Brain Res 2020; 399:113001. [PMID: 33197456 DOI: 10.1016/j.bbr.2020.113001] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/01/2020] [Accepted: 11/01/2020] [Indexed: 01/05/2023]
Abstract
Depression is a prevalent mood disorder responsible for reduced quality of life for over 264 million people. Depression commonly develops during adolescence and becomes twice as prevalent in females than in males. However, the mechanisms underlying adolescent depression onset and sex differences in the prevalence rate remain unclear. Adolescent exposure to stress and subsequent sensitization of the hypothalamic-pituitary-adrenal (HPA) axis contributes to mood disorder development, and females are particularly vulnerable to HPA sensitization. Repeated exposure to stressors common to adolescent development, like sleep disruption, could partially be responsible for adolescent female susceptibility to depression. To address this possibility, 80 adolescent and adult CD-1 mice (Male, n = 40; Female, n = 40) were manually sleep disrupted for the first four hours of each rest cycle or allowed normal rest for eight consecutive days. Depression-like behavior was assessed with the forced swim test. 5-HT1A and glucocorticoid receptor expression and concurrent cellular activation via glucocorticoid receptor/c-Fos colocalization were examined in various brain regions to assess cellular correlates of depression and HPA-axis activation. Both adolescent male and female mice displayed significantly greater depression-like behavior and prelimbic c-Fos expression after chronic sleep disruption than non-sleep disrupted adolescent and sleep disrupted adult counterparts. However, sleep disrupted adolescent females demonstrated greater dorsal raphe 5-HT1A expression than sleep disrupted adolescent males. Adolescent females and males had decreased medial prefrontal 5-HT1A expression after chronic sleep disruption, but only adolescent females expressed decreased hippocampal 5-HT1A expression compared to controls. Chronic sleep disruption significantly increased corticosterone release, glucocorticoid expression in the CA1, and activation of glucocorticoid immunoreactive cells in the prelimbic cortex of adolescent females but not in adolescent males. These findings suggest that chronic sleep disruption during adolescence could give rise to depressive symptoms in male and female adolescents through differing signaling mechanisms.
Collapse
Affiliation(s)
- Michael Murack
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Kevin B Smith
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Emily G Ah-Yen
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Étienne Rheaume
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Zahra Nanji
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Olivia Latus
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Claude Messier
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Canada
| | - Nafissa Ismail
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Canada.
| |
Collapse
|
11
|
Abstract
The reproductive function of humans is regulated by several sex hormones which are secreted in synergy with the circadian timing of the body. Sleep patterns produce generic signatures that physiologically drive the synthesis, secretion, and metabolism of hormones necessary for reproduction. Sleep deprivation among men and women is increasingly reported as one of the causes of infertility. In animal models, sleep disturbances impair the secretion of sexual hormones thereby leading to a decrease in testosterone level, reduced sperm motility and apoptosis of the Leydig cells in male rats. Sleep deprivation generates stressful stimuli intrinsically, due to circadian desynchrony and thereby increases the activation of the Hypothalamus-Pituitary Adrenal (HPA) axis, which, consequently, increases the production of corticosterone. The elevated level of corticosteroids results in a reduction in testosterone production. Sleep deprivation produces a commensurate effect on women by reducing the chances of fertility. Sleeplessness among female shift workers suppresses melatonin production as well as excessive HPA activation which results in early pregnancy loss, failed embryo implantation, anovulation and amenorrhea. Sleep deprivation in women has also be found to be associated with altered gonadotropin and sex steroid secretion which all together lead to female infertility. Poor quality of sleep is observed in middle-aged and older men and this also contributes to reduced testosterone concentrations. The influence of sleep disturbances post-menopausal is associated with irregular synthesis and secretion of female sex steroid hormones.
Collapse
|
12
|
Mishra I, Pullum KB, Thayer DC, Plummer ER, Conkright BW, Morris AJ, O'Hara BF, Demas GE, Ashley NT. Chemical sympathectomy reduces peripheral inflammatory responses to acute and chronic sleep fragmentation. Am J Physiol Regul Integr Comp Physiol 2020; 318:R781-R789. [PMID: 32130024 DOI: 10.1152/ajpregu.00358.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sleep loss contributes to the development of cardiovascular, metabolic, and neurological disorders by promoting a systemic proinflammatory phenotype. The neuroendocrine-immune mechanisms contributing to such pathologies are poorly understood. The sympathetic nervous system (SNS) regulates immunity and is often activated following sleep disturbances. The aims of this study were to determine 1) the effect of SNS inhibition on inflammatory responses to sleep fragmentation (SF) and 2) whether homeostasis can be restored after 1 wk of recovery sleep. We measured stress responses (norepinephrine and corticosterone), gene expression levels of pro- and anti-inflammatory cytokines in peripheral (heart, liver, and spleen) tissues, and protein levels of cytokines and chemokines in serum of female mice that were subjected to acute SF for 24 h, chronic SF for 8 wk, or 7 days of recovery after chronic SF. In each experiment, SF and control mice were chemically sympathectomized with 6-hydroxydopamine (6-OHDA) or injected with vehicle. Both acute and chronic SF elevated mRNA and protein levels of cytokines in peripheral tissues. Changes in inflammatory responses mirrored stress-axes activation, with increased corticosterone and norepinephrine in SF mice. 6-OHDA treatment significantly alleviated SF-induced inflammation, thus providing evidence of SNS regulation of peripheral inflammation from SF. Effects of chronic SF were more severe than acute SF, and 1 wk of recovery from SF sufficiently alleviated peripheral inflammatory responses but not NE responses.
Collapse
Affiliation(s)
- Ila Mishra
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| | - Keelee B Pullum
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| | - Domnique C Thayer
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| | - Erica R Plummer
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| | | | - Andrew J Morris
- Department of Pharmacology, University of Kentucky, Lexington, Kentucky
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, Lexington, Kentucky
| | - Gregory E Demas
- Department of Biology, Indiana University, Bloomington, Indiana
| | - Noah T Ashley
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| |
Collapse
|
13
|
Frau R, Traccis F, Bortolato M. Neurobehavioural complications of sleep deprivation: Shedding light on the emerging role of neuroactive steroids. J Neuroendocrinol 2020; 32:e12792. [PMID: 31505075 PMCID: PMC6982588 DOI: 10.1111/jne.12792] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/06/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
Sleep deprivation (SD) is associated with a broad spectrum of cognitive and behavioural complications, including emotional lability and enhanced stress reactivity, as well as deficits in executive functions, decision making and impulse control. These impairments, which have profound negative consequences on the health and productivity of many individuals, reflect alterations of the prefrontal cortex (PFC) and its connectivity with subcortical regions. However, the molecular underpinnings of these alterations remain elusive. Our group and others have begun examining how the neurobehavioural outcomes of SD may be influenced by neuroactive steroids, a family of molecules deeply implicated in sleep regulation and the stress response. These studies have revealed that, similar to other stressors, acute SD leads to increased synthesis of the neurosteroid allopregnanolone in the PFC. Whereas this up-regulation is likely aimed at counterbalancing the detrimental impact of oxidative stress induced by SD, the increase in prefrontal allopregnanolone levels contributes to deficits in sensorimotor gating and impulse control, signalling a functional impairment of PFC. This scenario suggests that the synthesis of neuroactive steroids during acute SD may be enacted as a neuroprotective response in the PFC; however, such compensation may in turn set off neurobehavioural complications by interfering with the corticolimbic connections responsible for executive functions and emotional regulation.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
- National Institute of Neuroscience (INN), University of Cagliari, Monserrato (CA), Italy
| | - Francesco Traccis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA
| |
Collapse
|
14
|
Barbosa CC, Rodrigues TC, Ataídes CFS, Santos ML, Ghedini PC, Dias Junior W, Andersen ML, Mazaro-Costa R. Protective effects of Hydrocotyle umbellata var. bonariensis Lam. (Araliaceae) on memory in sleep-impaired female mice. JOURNAL OF ETHNOPHARMACOLOGY 2019; 245:112183. [PMID: 31445134 DOI: 10.1016/j.jep.2019.112183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 06/03/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hydrocotyle umbellata var.bonariensis Lam. (Hb), popularly known in Brazil as acariçoba and outside Brazil Hb by a number of names including marsh-pennywort, and many-flower, has traditionally been used in Ayurvedic medicine in the retardation of aging (Rasayana effect). AIM OF THE STUDY The present study evaluated the effect of Hb treatment before and during paradoxical sleep deprivation (PSD) and sleep restriction (SR) on learning, memory, and acetylcholinesterase (AChE) brain activity. MATERIAL AND METHODS Adult Swiss nulliparous female mice were randomly distributed among the experimental groups. The treated groups received the aqueous solution of Hb leaves orally at concentrations of 500 and 1.000 mg/kg. PSD and SR were induced by the multiple platform method, in which the animals remained for 3-days in PSD or 15-days in SR for 22 h per day. The collection of the vaginal epithelium occurred daily to determine the estrous cycle. Body mass gain was determined. The animals were submitted to the passive avoidance test and were then euthanized for the collection of brain tissue and the determination of cerebral cholinesterase activity. RESULTS The aqueous solution of Hb was associated with a significant reduction in cholinesterase activity at both doses in the SR model, and at the dose of 1.000 mg/kg in the PSD model. Regarding the learning and memory test, the PSD group treated with 1.000 mg/kg presented significant improvement, whereas in the SR experiment none of the treated-groups showed any improvement in learning and memory. In the analysis of SR/PSD interference and/or Hb treatment on the estrous cycle, it was possible to observe that the treatment acted as a protector in the SR group, maintaining a normal cycle. CONCLUSIONS The analyses showed that Hb was safe to use during periods of SR or PSD, acting as an adaptogen for these situations, in addition to being able to reduce cholinesterase activity, which suggests its neuroprotective action. In relation to the estrous cycle, Hb can act as a protector in SR situations.
Collapse
Affiliation(s)
- C C Barbosa
- Departamento de Farmacologia - Universidade Federal de Goiás, Goiás, Brazil
| | - T C Rodrigues
- Departamento de Farmacologia - Universidade Federal de Goiás, Goiás, Brazil
| | - C F S Ataídes
- Departamento de Farmacologia - Universidade Federal de Goiás, Goiás, Brazil
| | - M L Santos
- Departamento de Farmacologia - Universidade Federal de Goiás, Goiás, Brazil
| | - P C Ghedini
- Departamento de Farmacologia - Universidade Federal de Goiás, Goiás, Brazil
| | - W Dias Junior
- Universidade Estadual de Goiás, Campus Ceres, Goiás, Brazil
| | - M L Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - R Mazaro-Costa
- Departamento de Farmacologia - Universidade Federal de Goiás, Goiás, Brazil.
| |
Collapse
|
15
|
Noise-induced sleep disruption increases weight gain and decreases energy metabolism in female rats. Int J Obes (Lond) 2018; 43:1759-1768. [PMID: 30568267 PMCID: PMC6584067 DOI: 10.1038/s41366-018-0293-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 01/28/2023]
Abstract
Background/objectives: Inadequate sleep increases obesity and environmental noise contributes to poor sleep. However, women may be more vulnerable to noise and hence more susceptible to sleep disruption-induced weight gain than men. In male rats, exposure to environmental (i.e. ambient) noise disrupts sleep and increases feeding and weight gain. However, the effects of environmental noise on sleep and weight gain in female rats are unknown. Thus, this study was designed to determine whether noise exposure would disturb sleep, increase feeding and weight gain and alter the length of the estrous cycle in female rats. Subjects/methods: Female rats (12-weeks old) were exposed to noise for 17d (8h/d during the light period) to determine the effects of noise on weight gain and food intake. In a separate set of females, estrous cycle phase and length, EEG, EMG, spontaneous physical activity and energy expenditure were recorded continuously for 27d during baseline (control, 9d), noise exposure (8h/d, 9d) and recovery (9d) from sleep disruption. Results: Noise exposure significantly increased weight gain and food intake compared to females that slept undisturbed. Noise also significantly increased wakefulness, reduced sleep and resulted in rebound sleep during the recovery period. Total energy expenditure was significantly lower during both noise exposure and recovery due to lower energy expenditure during spontaneous physical activity and sleep. Notably, noise did not alter the estrous cycle length. Conclusions: As previously observed in male rats, noise exposure disrupted sleep and increased weight gain in females but did not alter the length of the estrous cycle. This is the first demonstration of weight gain in female rats during sleep disruption. We conclude that the sleep disruption caused by exposure to environmental noise is a significant tool for determining how sleep loss contributes to obesity in females.
Collapse
|
16
|
|
17
|
Cordeira J, Kolluru SS, Rosenblatt H, Kry J, Strecker RE, McCarley RW. Learning and memory are impaired in the object recognition task during metestrus/diestrus and after sleep deprivation. Behav Brain Res 2017; 339:124-129. [PMID: 29180134 DOI: 10.1016/j.bbr.2017.11.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/23/2017] [Accepted: 11/23/2017] [Indexed: 01/25/2023]
Abstract
Females are an under-represented research model and the mechanisms through which sleep loss impairs cognition are not clear. Since levels of reproductive hormones and the estrous cycle are sensitive to sleep loss and necessary for learning and memory, we hypothesized that sleep deprivation impacts learning and memory in female mice by interfering with the estrous cycle. We used the object recognition task to assess learning and memory in female mice during separate phases of the estrous cycle and after sleep loss. Mice in metestrus/diestrus attended to sample objects less than mice in proestrus/estrus during object acquisition, the first phase of the object recognition task. Subsequently, during the recognition phase of the task, only mice in proestrus/estrus displayed a preference for the novel object. Sleep deprivation for 12h immediately before the object recognition task reduced time attending to sample objects and novel object preference for mice in proestrus/estrus, without changing length of the estrous cycle. These results show that sleep deprived mice in proestrus/estrus had learning deficits and memory impairments, like mice in metestrus/diestrus. Since sleep deprivation did not disrupt the estrous cycle, however, results did not support the hypothesis. Cognitive impairments due to acute sleep loss were not due to alterations to the estrous cycle.
Collapse
Affiliation(s)
- Joshua Cordeira
- Department of Biological & EnvironmentalSciences, Western Connecticut State University, Danbury, CT, USA.
| | - Sai Saroja Kolluru
- Department of Biological & EnvironmentalSciences, Western Connecticut State University, Danbury, CT, USA
| | - Heather Rosenblatt
- Department of Biological & EnvironmentalSciences, Western Connecticut State University, Danbury, CT, USA
| | - Jenny Kry
- Department of Biological & EnvironmentalSciences, Western Connecticut State University, Danbury, CT, USA
| | - Robert E Strecker
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA
| | - Robert W McCarley
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, Brockton, MA 02301, USA
| |
Collapse
|
18
|
Samuelsson LB, Bovbjerg DH, Roecklein KA, Hall MH. Sleep and circadian disruption and incident breast cancer risk: An evidence-based and theoretical review. Neurosci Biobehav Rev 2017; 84:35-48. [PMID: 29032088 DOI: 10.1016/j.neubiorev.2017.10.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 09/26/2017] [Accepted: 10/11/2017] [Indexed: 02/08/2023]
Abstract
Opportunities for restorative sleep and optimal sleep-wake schedules are becoming luxuries in industrialized cultures, yet accumulating research has revealed multiple adverse health effects of disruptions in sleep and circadian rhythms, including increased risk of breast cancer. The literature on breast cancer risk has focused largely on adverse effects of night shift work and exposure to light at night (LAN), without considering potential effects of associated sleep disruptions. As it stands, studies on breast cancer risk have not considered the impact of both sleep and circadian disruption, and the possible interaction of the two through bidirectional pathways, on breast cancer risk in the population at large. We review and synthesize this literature, including: 1) studies of circadian disruption and incident breast cancer; 2) evidence for bidirectional interactions between sleep and circadian systems; 3) studies of sleep and incident breast cancer; and 4) potential mechanistic pathways by which interrelated sleep and circadian disruption may contribute to the etiology of breast cancer.
Collapse
Affiliation(s)
- Laura B Samuelsson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dana H Bovbjerg
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Department of Behavioral & Community Health Sciences, University of Pittsburgh, Pittsburgh, PA, United States; Biobehavioral Oncology Program, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kathryn A Roecklein
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Martica H Hall
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
19
|
Sleep influences the immune response and the rejection process alters sleep pattern: Evidence from a skin allograft model in mice. Brain Behav Immun 2017; 61:274-288. [PMID: 28069386 DOI: 10.1016/j.bbi.2016.12.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/24/2016] [Accepted: 12/31/2016] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Sleep generally regulates immune functions in a supportive manner and can affect parameters that are directly involved in the rejection process. STUDY OBJECTIVES The first objective was to assess whether sleep deprivation (SD) or sleep restriction (SR) affects the allograft rejection process in mice. The second objective was to investigate whether the rejection process itself modulates the sleep pattern of allografted mice. DESIGN Adult BALB/c and C57BL/6J male mice were used as the donors and recipients, respectively, except for the syngeneic group (ISOTX), which received skin from mice of the same strain (C57BL/6J). The recipients were randomly assigned to either one of two control groups - TX (allogenic) or ISOTX (syngeneic) - which underwent stereotaxic surgery to enable sleep recording prior to the allograft but were not sleep deprived; one of two paradoxical sleep deprived groups - SDTX and TXSD - which underwent 72h of continuous SD either before or after the allograft respectively, and one of two sleep restricted groups - SRTX and TXSR - which underwent 21h of SD and 3h of sleep for 15days either before or after the allograft respectively. INTERVENTIONS The skin allograft was inspected daily to determine the survival time, expected as 8.0±0.4days in this transplant model under no treatment. The sleep pattern was controlled throughout the rejection process in the SD and SR groups. Draining lymph nodes, spleen, blood and skin grafts were harvested on the 5th day after transplantation for evaluation of the immune parameters related to allograft rejection. MEASUREMENTS AND RESULTS In the control groups, we observed a reduction in paradoxical sleep throughout the entire allograft rejection process. Acute and chronic experimental sleep loss in the SD and SR groups produced marked alterations in the immune response. Both SD and SR prolonged allograft survival compared to the non-sleep-deprived group. There were reductions in the following parameters involved in the allograft rejection under sleep loss: CD4+ and CD8+ T cell subpopulations in the peripheral lymph organs and spleen, circulating sIL-2R levels, graft-infiltrating CD4+ T cells and skin allograft global gene expression. CONCLUSIONS We provide, as far as we are aware, the first evidence in vivo that the immune response can alter the normal sleep pattern, and that sleep loss can conversely affect the immune response related to graft rejection.
Collapse
|
20
|
Xu X, Wang L, Chen L, Su T, Zhang Y, Wang T, Ma W, Yang F, Zhai W, Xie Y, Li D, Chen Q, Fu X, Ma Y, Zhang Y. Effects of chronic sleep deprivation on bone mass and bone metabolism in rats. J Orthop Surg Res 2016; 11:87. [PMID: 27485745 PMCID: PMC4970273 DOI: 10.1186/s13018-016-0418-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 07/13/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND This study aimed to assess the effects of chronic sleep deprivation (CSD) on bone mass and bone metabolism in rats. METHODS Twenty-four rats were randomly divided into CSD and control (CON) groups. Rats were subjected to CSD by using the modified multiple platform method (MMPM) to establish an animal model of CSD. Biochemical parameters such as levels of serum N-terminal propeptide of type I procollagen (PINP), N-terminal cross-linking telopeptide of type I collagen (NTX), growth hormone (GH), estradiol (E2), serum 25(OH)D, and calcium (Ca) were evaluated at 0, 1, 2, and 3 months. After 3 months, each fourth lumbar vertebra and the distal femoral metaphysis of the left extremity of rats were harvested for micro-computed tomography scans and histological analysis, respectively, after the rats were sacrificed under an overdose of pentobarbital sodium. RESULTS Compared with rats from the CON group, rats from the CSD group showed significant decreases in bone mineral density (BMD), bone volume over total volume, trabecular bone thickness, and trabecular bone number and significant increases in bone surface area over bone volume and trabecular bone separations (P < 0.05). Bone histomorphology studies showed that rats in the CSD group had decreased osteogenesis, impaired mineralization of newly formed bones, and deteriorative trabecular bone in the secondary spongiosa zone. In addition, they showed significantly decreased levels of serum PINP (1 month later) and NTX (3 months later) (P < 0.05). The serum 25(OH)D level of rats from the CSD group was lower than that of rats from the CON group after 1 month (P < 0.05). CONCLUSIONS CSD markedly affects bone health by decreasing BMD and 25(OH)D, deteriorating the bone microarchitecture, and decreasing bone formation and bone resorption markers.
Collapse
Affiliation(s)
- Xiaowen Xu
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Liang Wang
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Liying Chen
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Tianjiao Su
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Yan Zhang
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Tiantian Wang
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Weifeng Ma
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Fan Yang
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Wujie Zhai
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Yuanyuan Xie
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Dan Li
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Qiong Chen
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Xuemei Fu
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China
| | - Yuanzheng Ma
- Center of Orthopedics, The 309th Hospital of PLA, Beijing, 100091, China.
| | - Yan Zhang
- Center for Systems Biomedical Sciences, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| |
Collapse
|
21
|
Wen X, Chen X, Chen S, Tan Y, Rong F, Zhu J, Ma W. Influence of SKF38393 on changes of gene profile in rat prefrontal cortex during chronic paradoxical sleep deprivation. Behav Brain Res 2016; 304:60-6. [PMID: 26851556 DOI: 10.1016/j.bbr.2016.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 01/27/2016] [Accepted: 02/01/2016] [Indexed: 11/17/2022]
Abstract
Chronic paradoxical sleep deprivation (CSD) can induce dramatic physiological and neurofunctional changes in rats, including decreased body weight, reduced learning and memory, and declined locomotor function. SKF38393, a dopamine D1 receptor agonist, can reverse the above damages. However, the mechanism of CSD syndrome and reversal role of SKF38393 remains largely unexplained. To preliminarily elucidate the mechanism of the neural dysfunction caused by CSD, in the present study we use gene chips to examine the expression profile of more than 28,000 transcripts in the prefrontal cortex (PFC). Rats were sleep deprived by modified multi-platform method for 3 weeks. Totally 59 transcripts showed differential expressions in CSD group in contrast to controls; they included transcripts coding for caffeine metabolism, circadian rhythm, drug metabolism and some amino acid metabolism pathway. Among the 59 transcripts, 39 increased their expression and 20 decreased. Two transcripts can be specifically reversed with SKF38393, one of them is Homer1, which is related to 20 functional classifications and coding for Glutamatergic synapse pathway. Our findings in the present study indicate that long-term sleep deprivation may trigger the changes of some certain functions and pathways in the PFC, and lead to the dysfunction of this advanced neuron, and the activation of D1 receptor by SKF38393 might ameliorate these changes via modulation of some transcripts such as Homer1, which is involved in the Ca(2+) pathway and MAPK pathway related to Glutamatergic synapse pathway.
Collapse
Affiliation(s)
- Xiaosa Wen
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China; Minhang District Center for Disease Control and Prevention, Shanghai 201101, China
| | - Xinmin Chen
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China
| | - Si Chen
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China
| | - Yue Tan
- Surgical Department, Tangshan Gongren Hospital, Tangshan 063000, China
| | - Fei Rong
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China
| | - Jiangbo Zhu
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China.
| | - Wenling Ma
- Department of Enviromental Hygiene, Faculty of Tropical Medicine and Public Health, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
22
|
Yang CK, Wu RSC, Wu CH, Lin TRY, Tsai HD. Sleep deprivation enhances peripheral serotonin secretion to regulate the large follicle steroidogenesis of rats. Taiwan J Obstet Gynecol 2016; 54:260-5. [PMID: 26166337 DOI: 10.1016/j.tjog.2014.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2014] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Sleep deprivation (SD) leads to the disturbance of the estrous cycle. Serotonin, the levels of which increase with SD, has been shown to inhibit luteinizing hormone production and the receptor has been found in the follicles. In this study, the serotonin effect on preovulatory follicular steroidogenesis is investigated and the underlying mechanisms are elucidated. MATERIALS AND METHODS Female rats were subjected to SD for a time span of 1-4 days using the dish-over-water-method with a Rechtschaffen apparatus. Serum estradiol and serotonin concentrations were assessed; thereafter, they were evaluated with the effect of serotonin on the estradiol production and steroidogenic acute regulatory (StAR) protein expression in a serum-free culture system. Preovulatory follicles were dissected mechanically from the ovaries of 21-day-old rats, which induced follicle growth, and cultured for 24 hours with or without recombinant human follicle-stimulating hormone (FSH) in the presence or absence of serotonin. RESULTS SD, led to a significant decrease in serum estradiol concentrations, while serotonin concentrations were significantly elevated (all p < 0.05). Follicles were cultured with a constant dose of FSH (50 mIU/mL) and increasing doses of serotonin, estradiol production was reduced by 20%. The inhibitory effect of serotonin was concentration dependent. The addition of serotonin (0.1 μg/mL) decreased FSH-induced estradiol production and attenuated FSH-stimulated follicular StAR protein expression. The inhibitory effects of serotonin could be reduced by the serotonin receptor antagonist ketanserin. CONCLUSION These findings suggest that decreased serum estradiol concentrations in SD rats may be the result of serotonin-related inhibition of estradiol production and decreased large follicle expression of StAR protein.
Collapse
Affiliation(s)
- Chueh-Ko Yang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC; Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua, Taiwan, ROC
| | - Rick Sai-Chuen Wu
- Department of Anesthesiology, China Medical University and Hospital, Taichung, Taiwan, ROC
| | - Cheng-Hsuan Wu
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua, Taiwan, ROC
| | | | - Horng-Der Tsai
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua, Taiwan, ROC.
| |
Collapse
|
23
|
Bollinger JL, Bergeon Burns CM, Wellman CL. Differential effects of stress on microglial cell activation in male and female medial prefrontal cortex. Brain Behav Immun 2016; 52:88-97. [PMID: 26441134 PMCID: PMC4909118 DOI: 10.1016/j.bbi.2015.10.003] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 09/25/2015] [Accepted: 10/02/2015] [Indexed: 12/19/2022] Open
Abstract
Susceptibility to stress-linked psychological disorders, including post-traumatic stress disorder and depression, differs between men and women. Dysfunction of medial prefrontal cortex (mPFC) has been implicated in many of these disorders. Chronic stress affects mPFC in a sex-dependent manner, differentially remodeling dendritic morphology and disrupting prefrontally mediated behaviors in males and females. Chronic restraint stress induces microglial activation, reflected in altered microglial morphology and immune factor expression, in mPFC in male rats. Unstressed females exhibit increased microglial ramification in several brain regions compared to males, suggesting both heightened basal activation and a potential for sex-dependent effects of stress on microglial activation. Therefore, we assessed microglial density and ramification in the prelimbic region of mPFC, and immune-associated genes in dorsal mPFC in male and female rats following acute or chronic restraint stress. Control rats were left unstressed. On the final day of restraint, brains were collected for either qPCR or visualization of microglia using Iba-1 immunohistochemistry. Microglia in mPFC were classified as ramified, primed, reactive, or amoeboid, and counted stereologically. Expression of microglia-associated genes (MHCII, CD40, IL6, CX3CL1, and CX3CR1) was also assessed using qPCR. Unstressed females showed a greater proportion of primed to ramified microglia relative to males, alongside heightened CX3CL1-CX3CR1 expression. Acute and chronic restraint stress reduced the proportion of primed to ramified microglia and microglial CD40 expression in females, but did not significantly alter microglial activation in males. This sex difference in microglial activation could contribute to the differential effects of stress on mPFC structure and function in males versus females.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, IN 47405, United States; Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, United States
| | - Christine M Bergeon Burns
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, United States
| | - Cara L Wellman
- Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, IN 47405, United States; Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, United States.
| |
Collapse
|
24
|
Spaeth AM, Dinges DF, Goel N. Sex and race differences in caloric intake during sleep restriction in healthy adults. Am J Clin Nutr 2014; 100:559-66. [PMID: 24965304 PMCID: PMC4095659 DOI: 10.3945/ajcn.114.086579] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Evidence indicates that men and African Americans may be more susceptible to weight gain resulting from sleep loss than women and whites, respectively. Increased daily caloric intake is a major behavioral mechanism that underlies the relation between sleep loss and weight gain. OBJECTIVE We sought to assess sex and race differences in caloric intake, macronutrient intake, and meal timing during sleep restriction. DESIGN Forty-four healthy adults aged 21-50 y (mean ± SD: 32.7 ± 8.7 y; n = 21 women, n = 16 whites) completed an in-laboratory protocol that included 2 consecutive baseline nights [10 or 12 h time in bed (TIB)/night; 2200-0800 or 2200-1000] followed by 5 consecutive sleep-restriction nights (4 h TIB/night; 0400-0800). Caloric intake and meal-timing data were collected during the 2 d after baseline sleep and the first 3 d after sleep restriction. RESULTS During sleep restriction, subjects increased daily caloric intake (P < 0.001) and fat intake (P = 0.024), including obtaining more calories from condiments, desserts, and salty snacks (Ps < 0.05) and consumed 532.6 ± 295.6 cal during late-night hours (2200-0359). Relative to women, men consumed more daily calories during baseline and sleep restriction, exhibited a greater increase in caloric intake during sleep restriction (d = 0.62), and consumed a higher percentage of daily calories during late-night hours (d = 0.78, Ps < 0.05). African Americans and whites did not significantly differ in daily caloric intake, increased caloric intake during sleep restriction, or meal timing. However, African Americans consumed more carbohydrates, less protein, and more caffeine-free soda and juice than whites did during the study (Ps < 0.05). CONCLUSIONS Men may be more susceptible to weight gain during sleep loss than women due to a larger increase in daily caloric intake, particularly during late-night hours. These findings are relevant to the promotion of public health awareness by highlighting nutritional risk factors and modifiable behaviors for weight gain related to sleep-wake timing.
Collapse
Affiliation(s)
- Andrea M Spaeth
- From the Department of Psychology, School of Arts and Sciences (AMS) and the Division of Sleep and Chronobiology, Department of Psychiatry, Perelman School of Medicine (DFD and NG), University of Pennsylvania, Philadelphia, PA
| | - David F Dinges
- From the Department of Psychology, School of Arts and Sciences (AMS) and the Division of Sleep and Chronobiology, Department of Psychiatry, Perelman School of Medicine (DFD and NG), University of Pennsylvania, Philadelphia, PA
| | - Namni Goel
- From the Department of Psychology, School of Arts and Sciences (AMS) and the Division of Sleep and Chronobiology, Department of Psychiatry, Perelman School of Medicine (DFD and NG), University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
25
|
Alvarenga TA, Aguiar MF, Mazaro-Costa R, Tufik S, Andersen ML. Effects of sleep deprivation during pregnancy on the reproductive capability of the offspring. Fertil Steril 2013; 100:1752-7. [DOI: 10.1016/j.fertnstert.2013.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/01/2013] [Accepted: 08/01/2013] [Indexed: 11/30/2022]
|
26
|
More than hormones: sex differences in cardiovascular parameters after sleep loss in rats. Prog Neuropsychopharmacol Biol Psychiatry 2013; 44:34-8. [PMID: 23337035 DOI: 10.1016/j.pnpbp.2013.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 12/21/2012] [Accepted: 01/11/2013] [Indexed: 11/24/2022]
Abstract
Although the influence of sex on sleep pattern and cardiovascular parameters is well known, knowledge regarding the effects of sleep loss on heart responses in both sexes is scarce. The present study investigated the effects of paradoxical sleep deprivation (PSD) and chronic sleep restriction (SR) on cardiovascular parameters and adrenocorticotropic hormone (ACTH) levels in male and female rats. Both groups were randomly assigned to PSD for 96 h, SR for 21 days or home-cage control. Mean arterial pressure (MAP), heart rate (HR), baroreflex sensitivity (bradycardia and tachycardia responses) and ACTH levels were evaluated. The results showed that PSD induced a significant increase in HR and ACTH levels in both sexes, although male rats presented higher levels of ACTH hormone compared to females. In addition to sex-specific responses, PSD decreased the tachycardia only in male rats. SR, induced a significant increase in MAP and decrease in bradycardia in both sexes. Male rats were more affected by sleep deprivation protocols than females for MAP, bradycardia response, and ACTH levels. The results showed that the effects of sleep loss on cardiovascular parameters are associated with the protocol of sleep deprivation and that sex can modulate these effects. We suggested this experimental model as a suitable tool for further investigations of the relationship between cardiovascular parameters and sleep.
Collapse
|
27
|
Matos G, Tufik S, Scorza FA, Cavalheiro EA, Andersen ML. Sleep and epilepsy: exploring an intriguing relationship with a translational approach. Epilepsy Behav 2013; 26:405-9. [PMID: 23394796 DOI: 10.1016/j.yebeh.2012.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 12/08/2012] [Indexed: 01/16/2023]
Abstract
The relationship between sleep and epilepsy has been well established. There is a high prevalence of sleep disturbances in epilepsy, which are associated with a decreased quality of life of individuals with epilepsy. In view of this fact, preclinical research is necessary to address many gaps in knowledge. For instance, it is well known that sleep deprivation can trigger seizures; however, this is a complex pathophysiological event. In this context, there are many valuable animal models of epilepsy that reproduce clinical symptoms and can be used. Investigations using animal models that simulate clinical epilepsy are imperative. Furthermore, preclinical studies that reveal mechanisms related to sleep-epilepsy interactions are very important. Results of such studies can, in turn, improve the understanding of epilepsy itself and can be useful in developing new antiepileptic drugs and preventive measures to control seizures. Preclinical research should be performed using a translational framework with experimental designs that can lead to advances in the quality of life of individuals with epilepsy. In view of the fact that more than 50 million of people are affected by epilepsy around the world, understanding the relationship between sleep and epilepsy is imperative.
Collapse
Affiliation(s)
- Gabriela Matos
- Departamento de Psicobiologia, Universidade Federal de São Paulo, SP, Brazil
| | | | | | | | | |
Collapse
|
28
|
Tenório NM, Tufik S, Bergamaschi CT, Campos RR, Cintra F, Alvarenga TA, Andersen ML. Influence of acute sleep deprivation on cardiovascular parameters in female Zucker obese and lean rats. Obesity (Silver Spring) 2013; 21:510-5. [PMID: 23404731 DOI: 10.1002/oby.20063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Accepted: 08/10/2012] [Indexed: 11/05/2022]
Abstract
OBJECTIVE There is a reciprocal relationship between sleep duration and weight gain. However, the consequences of this relationship on the cardiovascular system over an entire life span are still not fully elucidated. We examined the effect of acute sleep deprivation (SD) on baroreflex sensitivity and blood pressure in Zucker rats of different ages. DESIGN AND METHODS Female lean and obese Zucker rats at 3, 6 and 15 months of age were assigned to SD or control (CTRL) groups. After a 6 h period of the SD procedure (6 h of gentle handling) or CTRL procedure (an equivalent period without handling), the animals were anesthetized for surgical catheterization of the femoral artery and vein. To evaluate the baroreflex sensitivity index, bolus infusions of phenylephrine (bradycardia response) and sodium nitroprusside (tachycardia response) were administered. RESULTS Obesity resulted in dysfunctional tachycardia responses at 3 months of age. At 6 and 15 months of age, both bradycardia and tachycardia responses were significantly lower in obese animals than those in lean animals. At 15 months of age, interactions among obesity, SD and aging produced the most marked effects on the cardiovascular system (increased mean arterial pressure and heart rate and decreased baroreflex sensitivity). CONCLUSIONS Therefore, these results suggest that there is no direct relationship between baroreflex imbalance and an increase in arterial pressure.
Collapse
Affiliation(s)
- Neuli M Tenório
- Departamento de Psicobiologia, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
29
|
Hachul H, Andersen ML, Bittencourt L, Santos-Silva R, Tufik S. A population-based survey on the influence of the menstrual cycle and the use of hormonal contraceptives on sleep patterns in São Paulo, Brazil. Int J Gynaecol Obstet 2012. [DOI: 10.1016/j.ijgo.2012.08.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Fernandes-Santos L, Patti CL, Zanin KA, Fernandes HA, Tufik S, Andersen ML, Frussa-Filho R. Sleep deprivation impairs emotional memory retrieval in mice: influence of sex. Prog Neuropsychopharmacol Biol Psychiatry 2012; 38:216-22. [PMID: 22521334 DOI: 10.1016/j.pnpbp.2012.03.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 03/28/2012] [Accepted: 03/29/2012] [Indexed: 11/24/2022]
Abstract
The deleterious effects of paradoxical sleep deprivation on memory processes are well documented. However, non-selective sleep deprivation occurs more commonly in modern society and thus represents a better translational model. We have recently reported that acute total sleep deprivation (TSD) for 6 h immediately before testing impaired performance of male mice in the plus-maze discriminative avoidance task (PM-DAT) and in the passive avoidance task (PAT). In order to extend these findings to females, we examined the effect of (pre-test) TSD on the retrieval of different memory tasks in both male and female mice. Animals were tested using 3 distinct memory models: 1) conditioning fear context (CFC), 2) PAT and 3) PM-DAT. In all experiments, animals were totally sleep-deprived by the gentle interference method for 6h immediately before being tested. In the CFC task and the PAT, TSD induced memory impairment regardless of sex. In PM-DAT, the memory impairing effects of TSD were greater in females. Collectively, our results confirm the impairing effect of TSD on emotional memory retrieval and demonstrate that it can be higher in female mice depending on the memory task evaluated.
Collapse
Affiliation(s)
- Luciano Fernandes-Santos
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Napoleão de Barros, 925, São Paulo, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
31
|
Gaskins GT, Moenter SM. Orexin a suppresses gonadotropin-releasing hormone (GnRH) neuron activity in the mouse. Endocrinology 2012; 153:3850-60. [PMID: 22673226 PMCID: PMC3404355 DOI: 10.1210/en.2012-1300] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GnRH neurons are critical for the central regulation of fertility, integrating steroidal, metabolic and other cues. GnRH neurons appear to lack receptors for many of these cues, suggesting involvement of afferent systems to convey information. Orexin A (orexin) is of interest in this regard as a neuromodulator that up-regulates metabolic activity, increases wakefulness, and affects GnRH/LH release. We examined the electrophysiological response of GnRH neurons to orexin application and how this response changes with estradiol and time of day in a defined animal model. Mice were either ovariectomized (OVX) or OVX and implanted with estradiol capsules (OVX+E). GnRH neurons from OVX+E mice exhibit low firing rates in the morning, due to estradiol-negative feedback, and high firing rates in the evening, due to positive feedback. Orexin inhibited activity of GnRH neurons from OVX mice independent of time of day. In GnRH neurons from OVX+E mice, orexin was inhibitory during the evening, suggesting orexin inhibition is not altered by estradiol. No effect of orexin was observed in OVX+E morning recordings, due to low basal GnRH activity. Inhibitory effects of orexin were mediated by the type 1 orexin receptor, but antagonism of this receptor did not increase GnRH neuron activity during estradiol-negative feedback. Spike pattern analysis revealed orexin increases interevent interval by reducing the number of single spikes and bursts. Orexin reduced spikes/burst and burst duration but did not affect intraburst interval. This suggests orexin may reduce overall firing rate by suppressing spike initiation and burst maintenance in GnRH neurons.
Collapse
Affiliation(s)
- Garrett T Gaskins
- Department of Molecular and Integrative Physiology, University of Michigan, 7725 Medical Science II, 1137 East Catherine Street, Ann Arbor Michigan 48109-5622, USA
| | | |
Collapse
|
32
|
O'connor EA, Parker MO, Davey EL, Grist H, Owen RC, Szladovits B, Demmers TGM, Wathes CM, Abeyesinghe SM. Effect of low light and high noise on behavioural activity, physiological indicators of stress and production in laying hens. Br Poult Sci 2012; 52:666-74. [PMID: 22221232 DOI: 10.1080/00071668.2011.639342] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
1. Commercial laying hens are commonly housed in noisy and dim environments, yet relatively little is known about whether these conditions, particularly in combination, have any effect on welfare or egg production. 2. The study was designed to investigate whether chronic exposure to continuous noise (60 dB(A) vs. 80 dB(A)) and/or light intensity (150 lux vs. 5 lux) during the critical period of coming into lay (16-24 weeks of age) influenced behaviour (activity, resting and feather maintenance), physiological stress (plasma corticosterone and heterophil to lymphocyte ratio) and production (number and weight of eggs laid) in laying hens. 3. Hens in the low light pens were less active and preened and dust-bathed more than those housed in 150 lux; hens in the high noise pens rested more frequently than those in quieter pens. 4. There was no evidence that chronic exposure to low light or high noise caused appreciable physiological stress but egg production was affected by these conditions. Hens kept in pens with low light or high noise laid fewer eggs per day than those kept in high light or low noise pens. These effects were additive, so that the fewest eggs were laid by hens subject to both low light and high noise. 5. These results show that low light intensity and continual high background noise have a detrimental effect on egg production in the early laying phase as well as influencing the time allocated to different behaviours. However there was no strong evidence for a physiological stress response to either of these conditions or their combination.
Collapse
Affiliation(s)
- E A O'connor
- Centre for Animal Welfare, Department of Veterinary Clinical Sciences, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire, AL7 9TA, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Sex differences in sleep pattern of rats in an experimental model of osteoarthritis. Eur J Pain 2012; 15:545-53. [DOI: 10.1016/j.ejpain.2010.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Revised: 09/20/2010] [Accepted: 10/15/2010] [Indexed: 01/22/2023]
|
34
|
Matos G, Tufik S, Scorza FA, Cavalheiro EA, Andersen ML. Sleep, epilepsy and translational research: What can we learn from the laboratory bench? Prog Neurobiol 2011; 95:396-405. [DOI: 10.1016/j.pneurobio.2011.09.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 08/19/2011] [Accepted: 09/18/2011] [Indexed: 12/20/2022]
|
35
|
Maia LO, Júnior WD, Carvalho LS, Jesus LR, Paiva GD, Araujo P, Costa MFO, Andersen ML, Tufik S, Mazaro-Costa R. Association of methamidophos and sleep loss on reproductive toxicity of male mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2011; 32:155-161. [PMID: 21843794 DOI: 10.1016/j.etap.2011.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/13/2011] [Accepted: 04/26/2011] [Indexed: 05/31/2023]
Abstract
This study investigated the effects of organophosphate exposure on the male reproductive system of mice submitted to chronic sleep loss condition. Adult Swiss mice were distributed into 4 groups: control; methamidophos (MTP); sleep restriction (SR); and MTP+SR. The dose of methamidophos was 0.002 mgkg(-1)day(-1) (half of the Acceptable Daily Intake). Sleep restriction condition was 21 h day(-1) during 15 days. In relation to control group, MTP treatment induced a significant reduction of 12% on morphologically normal spermatozoa in both MTP and MTP+SR groups. In addition, the absolute and relative weights of the seminal vesicles were decreased (MTP, -34%; MTP+SR, -45%). Epididymal fat was reduced in SR groups (SR, -64%; MTP+SR, -58%). Plasma testosterone levels were significantly decreased in MTP and SR groups, and progesterone levels were increased 8 times in MTP+SR in comparison with the control group. The corticosterone levels were unaffected by MTP or SR conditions. Thus, low dose MTP exposure resulted in deleterious effects on the male reproductive system. Sleep loss associated with MTP potentiated the effect on steroidogenesis, mainly in terms of progesterone levels.
Collapse
Affiliation(s)
- Lucas O Maia
- Departamento de Ciências Fisiológicas, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Andersen ML, Alvarenga TF, Mazaro-Costa R, Hachul HC, Tufik S. The association of testosterone, sleep, and sexual function in men and women. Brain Res 2011; 1416:80-104. [PMID: 21890115 DOI: 10.1016/j.brainres.2011.07.060] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 07/29/2011] [Accepted: 07/30/2011] [Indexed: 11/29/2022]
Abstract
Testosterone has been the focus of several investigations and review studies in males, but few have addressed its effects on sleep and sexual function, despite evidence of its androgenic effects on circadian activity in both sexes. Studies have been conducted to understand how sleeping increases (and how waking decreases) testosterone levels and how this rhythm can be related to sexual function. This review addresses the inter-relationships among testosterone, sexual function and sleep, including sleep-disordered breathing in both sexes, specifically its effects related to sleep deprivation. In addition, hormonal changes in testosterone that occur in the gonadal and adrenal axis with obstructive sleep apnea and other conditions of chronic sleep deprivation, and which consequently affect sexual life, have also been explored. Nevertheless, hormone-associated sleep disruptions occur across a lifetime, particularly in women. The association between endogenous testosterone and sex, sleep and sleep disturbances is discussed, including the results of clinical trials as well as animal model studies. Evidence of possible pathophysiological mechanisms underlying this relationship is also described. Unraveling the associations of sex steroid hormone concentrations with sleep and sexual function may have clinical implications, as sleep loss reduces testosterone levels in males, and low sex steroid hormone concentrations have been associated with sexual dysfunction.
Collapse
Affiliation(s)
- Monica L Andersen
- Departmento de Psicobiologia, Universidade Federal de São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
37
|
Egydio F, Tomimori J, Tufik S, Andersen ML. Does sleep deprivation and morphine influence wound healing? Med Hypotheses 2011; 77:353-5. [PMID: 21652152 DOI: 10.1016/j.mehy.2011.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 05/11/2011] [Indexed: 10/18/2022]
Abstract
The contrast between present-day sleep habits and those of the pre-industrial era are quite evident. One study recent has shown that the amount of sleep has decreased 2 h per night over the past 50 years. Such sleep curtailment, ubiquitous in the modern lifestyle, inflicts adverse repercussions upon health and well being. Investigations examining the relationship between stress and the skin have shown that different types of stress affect the healing process. Morphine is an immunosuppressive drug, and when it is used chronically, it can lead to an increased incidence of infections and a delay in the healing process. Therefore, our hypothesis is that the lack of sleep associated with chronic treatment with morphine is detrimental to the healing of the skin in the animal model we have adopted. Thus, it is important that future studies consider the paradigm of sleep curtailment when investigating the mechanisms involved in the process of skin healing in individuals who are dependent on morphine.
Collapse
Affiliation(s)
- F Egydio
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | | | | |
Collapse
|
38
|
Araujo P, Mazaro-Costa R, Tufik S, Andersen ML. Impact of sex on hyperalgesia induced by sleep loss. Horm Behav 2011; 59:174-9. [PMID: 21081131 DOI: 10.1016/j.yhbeh.2010.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 11/04/2010] [Accepted: 11/07/2010] [Indexed: 12/14/2022]
Abstract
This study evaluated the impact of sex on the short term consequences of different periods of sleep deprivation and the effect of the respective sleep recovery periods on nociceptive responses. Male and female C57BL/6J mice were assigned to the following groups: paradoxical sleep deprived (PSD) for 72 h, sleep restricted (SR) for 15 days, exposed to respective recovery periods for 24 h, or untreated home-cage controls (CTRL). Mice were submitted to a noxious thermal stimulus to evaluate their nociceptive response after PSD, SR, or recovery periods. Blood was collected for hormonal analysis. The nociceptive response was significantly lower in PSD and SR mice compared to CTRL animals, regardless of the sex. However, SR females had a lower paw withdrawal threshold than males. Sleep recovery was able to restore normal nociceptive sensitivity after PSD in both sexes. The hyperalgesia induced by SR was not reversed by sleep rebound. In females, low concentrations of estradiol were found after SR, and these concentrations continued to decrease after 24 hours of sleep recovery. The PSD male mice exhibited higher concentrations of corticosterone than the CTRL and SR male mice. Corticosterone levels were not affected by SR. Our study revealed that PSD and SR induce hyperalgesia in mice. The SR groups showed marked changes in the nociceptive response, and the females were more sensitive to these alterations. This finding indicates that, although different periods of sleep deprivation change the nociceptive sensitivity in male and female mice, sex could influence hyperalgesia induced by chronic sleep loss.
Collapse
Affiliation(s)
- P Araujo
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Brazil
| | | | | | | |
Collapse
|
39
|
Pires GN, Andersen ML, Giovenardi M, Tufik S. Sleep impairment during pregnancy: possible implications on mother-infant relationship. Med Hypotheses 2010; 75:578-82. [PMID: 20800370 DOI: 10.1016/j.mehy.2010.07.036] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 07/19/2010] [Indexed: 11/25/2022]
Abstract
The modern living standard has imposed upon society a situation of chronic sleep deprivation. This chronic loss of sleep affects women more than it does men. As a result, the postponement of pregnancy has become a common choice due to the priority given to social and domestic activities. For women, pregnancy represents a condition of intense physical and physiological changes that subject the pregnant woman to a number of potentially stressful situations, ultimately interfering with their quality of sleep. Chronic sleep deprivation, along with the changes imposed on women through pregnancy, can lead to several harmful consequences for the pregnant woman and the child, and can potentially undermine the mother-infant relationship. This article discusses circumstances under which sleep deprivation and poor sleep quality during pregnancy could result in damage to the mother-infant relationship, specifically through maternal fatigue, postpartum depression and changes in pregnancy-related hormonal secretions and activity.
Collapse
Affiliation(s)
- Gabriel Natan Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
40
|
Kahan V, Ribeiro DA, Andersen ML, Alvarenga TA, Tufik S. Sleep Loss Induces Differential Response Related To Genotoxicity in Multiple Organs of Three Different Mice Strains. Basic Clin Pharmacol Toxicol 2010; 107:598-602. [DOI: 10.1111/j.1742-7843.2010.00540.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
41
|
Andersen ML, Ribeiro DA, Alvarenga TA, Silva A, Araujo P, Zager A, Tenorio NM, Tufik S. Are endogenous sex hormones related to DNA damage in paradoxically sleep-deprived female rats? Horm Behav 2010; 57:216-21. [PMID: 19958772 DOI: 10.1016/j.yhbeh.2009.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 11/18/2009] [Accepted: 11/20/2009] [Indexed: 02/06/2023]
Abstract
The aim of this investigation was to evaluate overall DNA damage induced by experimental paradoxical sleep deprivation (PSD) in estrous-cycling and ovariectomized female rats to examine possible hormonal involvement during DNA damage. Intact rats in different phases of the estrous cycle (proestrus, estrus, and diestrus) or ovariectomized female Wistar rats were subjected to PSD by the single platform technique for 96 h or were maintained for the equivalent period as controls in home-cages. After this period, peripheral blood and tissues (brain, liver, and heart) were collected to evaluate genetic damage using the single cell gel (comet) assay. The results showed that PSD caused extensive genotoxic effects in brain cells, as evident by increased DNA migration rates in rats exposed to PSD for 96 h when compared to negative control. This was observed for all phases of the estrous cycle indistinctly. In ovariectomized rats, PSD also led to DNA damage in brain cells. No significant statistically differences were detected in peripheral blood, the liver or heart for all groups analyzed. In conclusion, our data are consistent with the notion that genetic damage in the form of DNA breakage in brain cells induced by sleep deprivation overrides the effects related to endogenous female sex hormones.
Collapse
Affiliation(s)
- Monica L Andersen
- Department of Psychobiology, Universidade Federal de São Paulo, Vila Clementino, SP, São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Hachul H, Andersen ML, Bittencourt LRA, Santos-Silva R, Conway SG, Tufik S. Does the reproductive cycle influence sleep patterns in women with sleep complaints? Climacteric 2009; 13:594-603. [DOI: 10.3109/13697130903450147] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
43
|
Tufik S, Andersen ML, Bittencourt LRA, Mello MTD. Paradoxical sleep deprivation: neurochemical, hormonal and behavioral alterations. Evidence from 30 years of research. AN ACAD BRAS CIENC 2009; 81:521-38. [DOI: 10.1590/s0001-37652009000300016] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Accepted: 04/03/2009] [Indexed: 11/22/2022] Open
Abstract
Sleep comprises approximately one-third of a person's lifetime, but its impact on health and medical conditions remains partially unrecognized. The prevalence of sleep disorders is increasing in modern societies, with significant repercussions on people's well-being. This article reviews past and current literature on the paradoxical sleep deprivation method as well as data on its consequences to animals, ranging from behavioral changes to alterations in the gene expression. More specifically, we highlight relevant experimental studies and our group's contribution over the last three decades.
Collapse
|
44
|
Alvarenga TA, Andersen ML, Velázquez-Moctezuma J, Tufik S. Food restriction or sleep deprivation: Which exerts a greater influence on the sexual behaviour of male rats? Behav Brain Res 2009; 202:266-71. [DOI: 10.1016/j.bbr.2009.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Revised: 04/01/2009] [Accepted: 04/02/2009] [Indexed: 10/20/2022]
|
45
|
Andersen ML, Alvarenga TA, Guindalini C, Perry JC, Silva A, Zager A, Tufik S. Paradoxical Sleep Deprivation Influences Sexual Behavior in Female Rats. J Sex Med 2009; 6:2162-72. [DOI: 10.1111/j.1743-6109.2009.01339.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
46
|
To what extent is sleep rebound effective in reversing the effects of paradoxical sleep deprivation on gene expression in the brain? Behav Brain Res 2009; 201:53-8. [DOI: 10.1016/j.bbr.2009.01.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 01/15/2009] [Accepted: 01/19/2009] [Indexed: 11/20/2022]
|
47
|
Schütz TCB, Andersen ML, Silva A, Tufik S. Distinct gender-related sleep pattern in an acute model of TMJ pain. J Dent Res 2009; 88:471-6. [PMID: 19493893 DOI: 10.1177/0022034509334618] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Since it is recognized that acute inflammation of the temporomandibular joint results in sleep disturbances in male rats, and that the orofacial region may display a site-specific effect of ovarian hormones on nociception, we hypothesized that distinct genders would respond differently when subjected to this inflammatory acute orofacial pain. Sleep was monitored after injection of saline/Freund's adjuvant into the temporomandibular joint in male and female (proestrus and diestrus phases) rats. Progesterone and stress-related hormones were also assessed. In males, Freund's adjuvant induced a significant nociceptive response and sleep disturbances. Behavior and sleep architecture in the females remained unaffected. Our results suggest that females and males present distinct responses to an acute model of orofacial pain.
Collapse
Affiliation(s)
- T C B Schütz
- Department of Psychobiology, Universidade Federal de São Paulo, Escola Paulista de Medicina, Rua Napoleão de Barros, 925, Vila Clementino, SP-04024-002, São Paulo, SP, Brazil.
| | | | | | | |
Collapse
|
48
|
Andersen ML, Ribeiro DA, Bergamaschi CT, Alvarenga TA, Silva A, Zager A, Campos RR, Tufik S. Distinct effects of acute and chronic sleep loss on DNA damage in rats. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:562-7. [PMID: 19258023 DOI: 10.1016/j.pnpbp.2009.02.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 02/20/2009] [Accepted: 02/20/2009] [Indexed: 01/20/2023]
Abstract
The aim of this investigation was to evaluate genetic damage induced in male rats by experimental sleep loss for short-term (24 and 96 h) and long-term (21 days) intervals, as well as their respective recovery periods in peripheral blood, brain, liver and heart tissue by the single cell gel (comet) assay. Rats were paradoxically deprived of sleep (PSD) by the platform technique for 24 or 96 h, or chronically sleep-restricted (SR) for 21 days. We also sought to verify the time course of their recovery after 24 h of rebound sleep. The results showed DNA damage in blood cells of rats submitted to PSD for 96 h. Brain tissue showed extensive genotoxic damage in PSD rats (both 24 and 96 h), though the effect was more pronounced in the 96 h group. Rats allowed to recover from the PSD-96 h and SR-21 days treatments showed DNA damage as compared to negative controls. Liver and heart did not display any genotoxicity activity. Corticosterone concentrations were increased after PSD (24 and 96 h) relative to control rats, whereas these levels were unaffected in the SR group. Collectively, these findings reveal that sleep loss was able to induce genetic damage in blood and brain cells, especially following acute exposure. Since DNA damage is an important step in events leading to genomic instability, this study represents a relevant contribution to the understanding of the potential health risks associated with sleep deprivation.
Collapse
Affiliation(s)
- M L Andersen
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP), Brazil
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Silva A, Andersen M, De Mello M, Bittencourt L, Peruzzo D, Tufik S. Gender and age differences in polysomnography findings and sleep complaints of patients referred to a sleep laboratory. Braz J Med Biol Res 2008; 41:1067-75. [DOI: 10.1590/s0100-879x2008001200005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Accepted: 11/28/2008] [Indexed: 11/21/2022] Open
Affiliation(s)
- A. Silva
- Universidade Federal de São Paulo, Brasil
| | | | | | | | - D. Peruzzo
- Universidade Estadual de Campinas, Brasil
| | - S. Tufik
- Universidade Federal de São Paulo, Brasil
| |
Collapse
|
50
|
Alvarenga TA, Patti CL, Andersen ML, Silva RH, Calzavara MB, Lopez GB, Frussa-Filho R, Tufik S. Paradoxical sleep deprivation impairs acquisition, consolidation, and retrieval of a discriminative avoidance task in rats. Neurobiol Learn Mem 2008; 90:624-32. [DOI: 10.1016/j.nlm.2008.07.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 07/09/2008] [Accepted: 07/22/2008] [Indexed: 10/21/2022]
|