51
|
Hauquiert B, Drion E, Deflandre E. [The role of biomarkers in the detection of the OSA syndrome. A narrative review of the literature]. Rev Mal Respir 2021; 38:455-465. [PMID: 33958251 DOI: 10.1016/j.rmr.2021.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/20/2021] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Obstructive sleep apnoea (OSA) is a common sleep-related breath disorder associated with cardiovascular and cerebrovascular complications, such as hypertension, arrhythmia, coronary artery disease and stroke. Unfortunately, OSA is underdiagnosed. BACKGROUND Because of its clinical and therapeutic variability, OSA could benefit a personalized medicine approach. Diagnosis with polysomnography is expensive and access is limited. Clinical scoring systems allow screening of OSA, but many limitations exist. Because of this, biomarkers could be useful for the detection of OSA. OUTLOOK Biomarkers specific to OSA would allow for better mass screening and more personalized treatment of the disease. This narrative review of the literature aims to summarize the biomarkers already described for the diagnosis of OSA and clarify both their advantages and limitations in daily practice. CONCLUSIONS Our review of the literature did not actually identify an ideal biomarker even if promising research is ongoing.
Collapse
Affiliation(s)
- B Hauquiert
- Département d'anesthésie-réanimation, Université catholique de Louvain, Bruxelles, Belgique
| | - E Drion
- Département de neurologie, Université de Liège, Liège, Belgique
| | - E Deflandre
- Chef de service et maître de stages, Département d'anesthésie-réanimation, Clinique Saint-Luc-de-Bouge, Namur, Belgique.
| |
Collapse
|
52
|
Okuda M, Noda A, Mabuchi S, Iwamoto K, Banno M, Miyata S, Yasuma F, Ozaki N. Sleep fragmentation and working memory in healthy adults. Sleep Sci 2021; 14:111-117. [PMID: 35082979 PMCID: PMC8764943 DOI: 10.5935/1984-0063.20200088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/08/2020] [Indexed: 12/05/2022] Open
Abstract
Introduction Sleep is essential for performing cognitive function in humans. We have hypothesized that sleep fragmentation compared to sleep efficiency may have a negative impact on the working memory. Material and Methods Twenty-eight healthy adults (18 males and 10 females; mean age 27.8±15.5 years) were enrolled in this study. We measured the total sleep time (TST), sleep efficiency, %stage wakefulness (W), %stage rapid eye movement (REM), %stage N1, %stage N2, %stage N3, wake after sleep onset (WASO), and arousal index using polysomnography. Working memory, executive function, and sustained attention of three domains of cognitive function were evaluated with the number of back task (N-back task), Wisconsin card sorting test (WCST), and continuous performance test-identical pairs (CPT-IP), respectively. Results The percentage of correct answers on the 2-back task was significantly correlated with %stage REM, %stage N1, and %stage N2 (%stage REM: r=0.505, p=0.006; %stage N1: r=-0.637, p<0.001; %stage N2: r=0.670, p<0.001), and multiple regression analysis including the stepwise forward selection method revealed that %stage N2 was the most significant factor (%stage N2: β=0.670, p<0.001). The percentage of correct answers on the 2-back task was also significantly correlated with TST, sleep efficiency, WASO, and arousal index (TST: r=0.492, p=0.008; sleep efficiency: r=0.622, p<0.001; WASO: r=-0.721, p<0.001; arousal index: r=-0.656, p<0.001), and WASO was the significant factor (β=-2.086, p=0.007). The WCST category achievement and CPT-IP d-prime score were correlated with none of the sleep variables. Conclusion Increased WASO and a decrease in %stage N2 were associated with worse working memory.
Collapse
Affiliation(s)
- Masato Okuda
- Chubu University Graduate School of Life and Health Sciences, Department of Biomedical Sciences - Kasugai - Aichi - Japan
| | - Akiko Noda
- Chubu University Graduate School of Life and Health Sciences, Department of Biomedical Sciences - Kasugai - Aichi - Japan
| | - Sho Mabuchi
- Chubu University Collage of Life and Health Sciences, Department of Biomedical Sciences -Kasugai - Aichi- Japan
| | - Kunihiro Iwamoto
- Nagoya University Graduate School of Medicine, Department of Psychiatry - Nagoya - Aichi -Japan
| | - Masahiro Banno
- Nagoya University Graduate School of Medicine, Department of Psychiatry - Nagoya - Aichi -Japan.,Seichiryo Hospital, Department of Psychiatry - Nagoya - Aichi - Japan
| | - Seiko Miyata
- Nagoya University Graduate School of Medicine, Department of Psychiatry - Nagoya - Aichi -Japan
| | - Fumihiko Yasuma
- National Hospital Organization Suzuka Hospital, Department of Internal Medicine - Suzuka - Mie -Japan
| | - Norio Ozaki
- Nagoya University Graduate School of Medicine, Department of Psychiatry - Nagoya - Aichi -Japan
| |
Collapse
|
53
|
Britten RA, Wellman LL, Sanford LD. Progressive increase in the complexity and translatability of rodent testing to assess space-radiation induced cognitive impairment. Neurosci Biobehav Rev 2021; 126:159-174. [PMID: 33766676 DOI: 10.1016/j.neubiorev.2021.01.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/15/2020] [Accepted: 01/07/2021] [Indexed: 11/29/2022]
Abstract
Ground-based rodent models have established that space radiation doses (approximately those that astronauts will be exposed to on a mission to Mars) significantly impair performance in a wide range of cognitive tasks. Over the last 40 years there has been a progressive increase in both the complexity and the translatability (to humans) of the cognitive tasks investigated. This review outlines technical and conceptual advances in space radiation rodent testing approaches, along with the advances in analytical approaches, that will make data from ground based studies more amenable to probabilistic risk analysis. While great progress has been made in determining the impact of space radiation on many advanced cognitive processes, challenges remain that need to be addressed prior to commencing deep space missions. A summary of on-going attempts to address existing knowledge gaps and the critical role that rodent studies will have in establishing the impact of space radiation on even more complex (human) cognitive tasks are presented and discussed.
Collapse
Affiliation(s)
- Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Leroy T Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA.
| | - Laurie L Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Pathology & Anatomy, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Larry D Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Pathology & Anatomy, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| |
Collapse
|
54
|
Ji T, Li X, Qiu Y, Mei L, Jia X, Tai J, Guo Y, Zhang J, Wang S, Ni X. Disease characteristics and neuropathological changes associated with cognitive dysfunction in obstructive sleep apnea. Pediatr Investig 2021; 5:52-57. [PMID: 33778428 PMCID: PMC7984002 DOI: 10.1002/ped4.12247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a common sleep-disordered breathing disease that often leads to many comorbidities (e.g., cognitive dysfunction), which adversely affect the quality of life for patients with OSA. Thus far, the underlying mechanisms of this dysfunction remain unclear. Many studies have focused on OSA-related characteristics, including intermittent hypoxemia and sleep fragmentation. There is increasing emphasis on neuroimaging studies to explore underlying relationships between neuropathological changes and cognitive dysfunction. This article reviews recent research progress concerning cognitive dysfunction associated with OSA to reveal potential mechanisms that contribute to this dysfunction.
Collapse
Affiliation(s)
- Tingting Ji
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Xiaodan Li
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Yue Qiu
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Lin Mei
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Xinbei Jia
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Jun Tai
- Department of OtorhinolaryngologyBeijing Children’s HospitalCapital Institute of PediatricsBeijingChina
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of OtolaryngologyHead and Neck SurgeryBeijing Pediatric Research InstituteBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Shengcai Wang
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Xin Ni
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
- Beijing Key Laboratory for Pediatric Diseases of OtolaryngologyHead and Neck SurgeryBeijing Pediatric Research InstituteBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| |
Collapse
|
55
|
Arora S, Venugopalan A, Dharavath RN, Bishnoi M, Kondepudi KK, Chopra K. Naringenin Ameliorates Chronic Sleep Deprivation-Induced Pain via Sirtuin1 Inhibition. Neurochem Res 2021; 46:1177-1187. [PMID: 33599956 DOI: 10.1007/s11064-021-03254-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 12/01/2022]
Abstract
Growing experimental evidences have suggested the reciprocal correlation between sleep deprivation and pain. Inflammation and oxidative stress are among the key pathways underlying this correlation. Therefore, the present study was aimed to assess the effect of antioxidant and anti-inflammatory compound naringenin (NGN) against chronic sleep deprivation (CSD)-induced mechanical and thermal hyperalgesia in female Swiss albino mice. In this study, mice were chronically sleep-deprived for 8 h a day for five days a week with the weekend as a free sleep period and continued for nine weeks using a modified multiple platform method. The pain behavioral tests were conducted at the end of the fourth week to assess the development of hyperalgesia followed by the administration of NGN and a combination of NGN with Sirtinol (SIR, a sirtuin1 inhibitor) till the end of the study. After nine weeks, pain behavioral tests, along with oxidative stress and inflammatory parameters in cortex and striatum, were assessed. Results indicated that CSD-induced hyperalgesia in mice accompanied by increased oxidative stress and inflammatory markers in cortex and striatum of the brain. NGN combatted the hyperalgesic response and also decreased levels of oxidative stress and inflammatory markers. Furthermore, the pharmacological effect of NGN was mitigated with SIR. Thus, the findings of the present study reveal that NGN is acting via sirtuin1 to exert its antinociceptive activity against CSD-induced hyperalgesia.
Collapse
Affiliation(s)
- Shiyana Arora
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, Panjab University, Sector 14, 160014, Chandigarh, India
| | - Aishwarya Venugopalan
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, Panjab University, Sector 14, 160014, Chandigarh, India
| | - Ravinder Naik Dharavath
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, Panjab University, Sector 14, 160014, Chandigarh, India
| | - Mahendra Bishnoi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, 140306, Punjab, India
| | - Kanthi Kiran Kondepudi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, 140306, Punjab, India
| | - Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, Panjab University, Sector 14, 160014, Chandigarh, India.
| |
Collapse
|
56
|
Circadian Rhythm: Potential Therapeutic Target for Atherosclerosis and Thrombosis. Int J Mol Sci 2021; 22:ijms22020676. [PMID: 33445491 PMCID: PMC7827891 DOI: 10.3390/ijms22020676] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 02/07/2023] Open
Abstract
Every organism has an intrinsic biological rhythm that orchestrates biological processes in adjusting to daily environmental changes. Circadian rhythms are maintained by networks of molecular clocks throughout the core and peripheral tissues, including immune cells, blood vessels, and perivascular adipose tissues. Recent findings have suggested strong correlations between the circadian clock and cardiovascular diseases. Desynchronization between the circadian rhythm and body metabolism contributes to the development of cardiovascular diseases including arteriosclerosis and thrombosis. Circadian rhythms are involved in controlling inflammatory processes and metabolisms, which can influence the pathology of arteriosclerosis and thrombosis. Circadian clock genes are critical in maintaining the robust relationship between diurnal variation and the cardiovascular system. The circadian machinery in the vascular system may be a novel therapeutic target for the prevention and treatment of cardiovascular diseases. The research on circadian rhythms in cardiovascular diseases is still progressing. In this review, we briefly summarize recent studies on circadian rhythms and cardiovascular homeostasis, focusing on the circadian control of inflammatory processes and metabolisms. Based on the recent findings, we discuss the potential target molecules for future therapeutic strategies against cardiovascular diseases by targeting the circadian clock.
Collapse
|
57
|
Britten RA, Fesshaye AS, Duncan VD, Wellman LL, Sanford LD. Sleep Fragmentation Exacerbates Executive Function Impairments Induced by Low Doses of Si Ions. Radiat Res 2020; 194:116-123. [PMID: 32845991 DOI: 10.1667/rade-20-00080.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/22/2020] [Indexed: 11/03/2022]
Abstract
Astronauts on deep space missions will be required to work autonomously and thus their ability to perform executive functions could be critical to mission success. Ground-based rodent experiments have shown that low (<25 cGy) doses of several space radiation (SR) ions impair various aspects of executive function. Translating ground-based rodent studies into tangible risk estimates for astronauts remains an enormous challenge, but should similar neurocognitive impairments occur in astronauts exposed to low-SR doses, a Numbers-Needed-to-Harm analysis (of the rodent data) predicts that approximately 30% of the astronauts could develop severe cognitive flexibility decrements. In addition to the health risks associated with SR exposure, astronauts have to contend with other stressors, of which inadequate sleep quantity and quality are considered to be major concerns. We have shown that a single session of fragmented sleep uncovered latent attentional set-shifting (ATSET) performance deficits in rats exposed to protracted neutron radiation that had no obvious defects in performance under rested wakefulness conditions. It is unclear if the exacerbating effect of sleep fragmentation (SF) only occurs in rats receiving protracted low-dose-rate-neutron radiation. In this study, we assessed whether SF also unmasks latent ATSET deficits in rats exposed to 5 cGy 600 MeV/n 28Si ions. Only sham and Si-irradiated rats that had good ATSET performance (passing every stage of the test on their first attempt) were selected for study. Sleep fragmentation selectively impaired performance in the more complex IDR, EDS and EDR stages of the ATSET test in the Si-irradiated rats. Set-shifting performance has rarely been affected by SR exposure in our studies conducted with rats tested under rested wakefulness conditions. The consistent SF-related unmasking of latent set-shifting deficits in both Si- and neutron-irradiated rats suggests that there is a unique interaction between sleep fragmentation and space radiation on the functionality of the brain regions that regulate performance in the IDR, EDS and EDR stages of ATSET. The uncovering of these latent SR-induced ATSET performance deficits in both Si- and neutron-irradiated rats suggests that the true impact of SR-induced cognitive impairment may not be fully evident in normally rested rats, and thus cognitive testing needs to be conducted under both rested wakefulness and sleep fragmentation conditions.
Collapse
Affiliation(s)
- Richard A Britten
- Departments of a Radiation Oncology.,Departments of Microbiology and Molecular Cell Biology.,Center for Integrative Neuroscience and Inflammatory Diseases.,Leroy T. Canoles Jr. Cancer Center
| | | | | | - Laurie L Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases.,Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Larry D Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases.,Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
58
|
Ji T, Lu T, Qiu Y, Li X, Liu Y, Tai J, Guo Y, Zhang J, Wang S, Zhao J, Ni X, Xu Z. The efficacy and safety of montelukast in children with obstructive sleep apnea: a systematic review and meta-analysis. Sleep Med 2020; 78:193-201. [PMID: 33465554 DOI: 10.1016/j.sleep.2020.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The efficacy and safety of montelukast in children with obstructive sleep apnea (OSA) remain controversial. Therefore, the aims of this systemic review and meta-analysis are to verify this issue and further provide reference for clinical practice. METHODS Seven databases were searched for randomized controlled trials (RCTs) up to September 30, 2019. The literature screening and data extraction were performed by two independent researchers. Adverse reactions from trials were also recorded. Meta-analysis was performed and analyzed heterogeneity. Methodological and evidence quality were followed by to evaluate according to Cochrane handbook. RESULTS A total of 4 RCTs including 305 children with mild to moderate OSA were involved. Compared with placebo, we found that oral montelukast (OM) significantly improved polysomnography (PSG) monitoring parameters, typical and relevant symptoms including snoring and mouth breathing, and adenoid morphology in children with OSA. When compared with routine drugs, not only PSG monitoring parameters and adenoid morphology, but also sleep-disordered breathing (SDB)-related questionnaire scores were improved in patients with OSA treated by combination of OM and routine drugs. In addition, compared with single nasal spray of mometasone furoate, the present study also showed that OM combined with nasal spray of mometasone furoate significantly improved PSG monitoring parameters, symptoms of snoring and mouth breathing and reduced tonsil morphology in pediatric OSA. In terms of treatment safety, one study reported adverse reactions of OM such as headache, nausea and vomiting, while no adverse events were reported after OM treatment in another study. CONCLUSION As a classic leukotriene receptor antagonist, montelukast can be used to treat children with mild to moderate OSA in the short term and improve clinical characteristics. The promotion and application of OM in clinic is considered to be a noninvasive option to avoid surgical treatment.
Collapse
Affiliation(s)
- Tingting Ji
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China
| | - Tingting Lu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Yue Qiu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China
| | - Xiaodan Li
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China
| | - Yali Liu
- Center for Clinical Epidemiology and Evidence-based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China
| | - Jun Tai
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China; Children's Hospital Capital Institute of Pediatrics, Beijing, PR China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China
| | - Shengcai Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China
| | - Jing Zhao
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China
| | - Xin Ni
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China; Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China.
| | - Zhifei Xu
- Department of Respiratory Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China.
| |
Collapse
|
59
|
Schilperoort M, van den Berg R, Coomans CP, Khedoe PPSJ, Ramkisoensing A, Boekestijn S, Wang Y, Berbée JFP, Meijer JH, Biermasz NR, Rensen PCN, Kooijman S. Continuous Light Does Not Affect Atherosclerosis in APOE*3-Leiden.CETP Mice. J Biol Rhythms 2020; 35:598-611. [PMID: 32915671 PMCID: PMC7683885 DOI: 10.1177/0748730420951320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Artificial light exposure is associated with dyslipidemia in humans, which is a major risk factor for the development of atherosclerotic cardiovascular disease. However, it remains unclear whether artificial light at night can exacerbate atherosclerosis. In this study, we exposed female APOE*3-Leiden.CETP mice, a well-established model for human-like lipid metabolism and atherosclerosis, to either a regular light-dark cycle or to constant bright light for 14 weeks. Mice exposed to constant light demonstrated a minor reduction in food intake, without any effect on body weight, body composition, or the weight of metabolic organs. Constant light increased the plasma levels of proatherogenic non–high-density lipoprotein (HDL) cholesterol but did not increase the size or severity of atherosclerotic lesions in the aortic root. Mice exposed to constant light did show lower immune cell counts, which could explain the absence of an effect of atherosclerosis despite increased non–HDL cholesterol levels. Behavioral analysis demonstrated variability in the response of mice to the light intervention. Constant light completely blunted behavioral rhythms in some mice, while others extended their behavioral period. However, rhythm strength was not an important determinant of atherosclerosis. Altogether, these results demonstrate that constant bright light does not affect atherosclerosis in APOE*3-Leiden.CETP mice. Whether artificial light exposure contributes to cardiovascular disease risk in humans remains to be investigated.
Collapse
Affiliation(s)
- Maaike Schilperoort
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Rosa van den Berg
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Claudia P Coomans
- Department of Molecular Cell Biology, Division of Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Padmini P S J Khedoe
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ashna Ramkisoensing
- Department of Molecular Cell Biology, Division of Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sanne Boekestijn
- Oncode Institute, Utrecht, the Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Yanan Wang
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Department of Endocrinology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jimmy F P Berbée
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Johanna H Meijer
- Department of Molecular Cell Biology, Division of Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nienke R Biermasz
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Department of Endocrinology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
60
|
Khalyfa A, Castro-Grattoni AL, Gozal D. Cardiovascular morbidities of obstructive sleep apnea and the role of circulating extracellular vesicles. Ther Adv Respir Dis 2020; 13:1753466619895229. [PMID: 31852426 PMCID: PMC6923690 DOI: 10.1177/1753466619895229] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Obstructive sleep apnea (OSA) is characterized by recurrent upper airway collapse
during sleep resulting in impaired blood gas exchange, namely intermittent
hypoxia (IH) and hypercapnia, fragmented sleep (SF), increased oxidative stress
and systemic inflammation. Among a myriad of potential associated morbidities,
OSA has been particularly implicated as mechanistically contributing to the
prevalence and severity of cardiovascular diseases (CVD). However, the benefits
of continuous positive airway pressure (CPAP), which is generally employed in
OSA treatment, to either prevent or improve CVD outcomes remain unconvincing,
suggesting that the pathophysiological mechanisms underlying the incremental CVD
risk associated with OSA are not clearly understood. One of the challenges in
development of non-invasive diagnostic assays is the ability to identify
clinically and mechanistically relevant biomarkers. Circulating extracellular
vesicles (EVs) and their cargos reflect underlying changes in cellular
homeostasis and can provide insights into how cells and systems cope with
physiological perturbations by virtue of the identity and abundance of miRNAs,
mRNAs, proteins, and lipids that are packaged in the EVs under normal as well as
diseased states, such as OSA. EVs can not only provide unique insights into
coordinated cellular responses at the organ or systemic level, but can also
serve as reporters of the effects of OSA in CVD, either by their properties
enabling regeneration and repair of injured vascular cells or by damaging them.
Here, we highlight recent progress in the pathological CVD consequences of OSA,
and explore the putative roles of EVs in OSA-associated CVD, along with emerging
diagnostic and therapeutic opportunities. The reviews of this paper are available via the supplemental material
section.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - Anabel L Castro-Grattoni
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - David Gozal
- Department of Child Health and MU Women's and Children's Hospital, University of Missouri School of Medicine, 400 N. Keene Street, Suite 010, Columbia, MO 65201, USA
| |
Collapse
|
61
|
Bertrand SJ, Zhang Z, Patel R, O'Ferrell C, Punjabi NM, Kudchadkar SR, Kannan S. Transient neonatal sleep fragmentation results in long-term neuroinflammation and cognitive impairment in a rabbit model. Exp Neurol 2020; 327:113212. [PMID: 31987835 DOI: 10.1016/j.expneurol.2020.113212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/18/2020] [Accepted: 01/24/2020] [Indexed: 12/29/2022]
Abstract
Sleep fragmentation is an increase in sleep-wake transitions without an overall decrease in total sleep time. Sleep fragmentation is well documented during acute and chronic hospitalization and can result in delirium and memory problems in children. Sleep fragmentation is also often noted in neurodevelopmental disorders. However, it is unclear how sleep fragmentation independent of disease affects brain development and function. We hypothesized that acute sleep fragmentation during the neonatal period in otherwise healthy animals would result in neuroinflammation and would be associated with abnormalities in cognitive development. The orbital shaker method was used to fragment sleep for 72 h in postnatal day 3 New Zealand white rabbit kits (fragmentation group). To control for maternal separation, the sham group was separated from the dam and maintained in the same conditions without undergoing sleep fragmentation. A naïve control group remained with the dam. Kits underwent behavioral testing with novel object recognition and spontaneous alternation T-maze tests at 2-3 weeks post-fragmentation and were sacrificed 3-50 days after fragmentation. Sleep fragmentation resulted in acute and chronic changes in microglial morphology in the hippocampus and cortex, and regional differences in mRNA expression of pro- and anti-inflammatory cytokines at 3, 7 and 50 days post-fragmentation. Impaired novel object recognition and a longer latency in T-maze task completion were noted in the fragmented kits. This was in spite of normalization of sleep architecture noted at 2 months of age in these kits. The results indicate that transient neonatal sleep fragmentation results in short-term and long-term immune alterations in the brain, along with diminished performance in cognitive tasks long-term.
Collapse
Affiliation(s)
- Sarah J Bertrand
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Zhi Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Ruchit Patel
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Caroline O'Ferrell
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Naresh M Punjabi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America
| | - Sapna R Kudchadkar
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America; Department of Pediatrics, Johns Hopkins University School of Medicine, United States of America; Department of Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, United States of America.
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, United States of America.
| |
Collapse
|
62
|
Britten RA, Duncan VD, Fesshaye AS, Wellman LL, Fallgren CM, Sanford LD. Sleep fragmentation exacerbates executive function impairments induced by protracted low dose rate neutron exposure. Int J Radiat Biol 2019; 97:1077-1087. [PMID: 31724895 DOI: 10.1080/09553002.2019.1694190] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/07/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE Astronauts on the planned missions to Mars are expected to have to work more autonomously than on previous missions. Thus mission success may be influenced by the astronauts' ability to respond quickly to unexpected problems, processes that require several executive functions. The purpose of this study was to determine the impact that prolonged low dose and low dose rate exposure to neutrons had on two executive functions, and whether the severity and incidence of cognitive impairment was altered by sleep fragmentation. MATERIALS AND METHODS In this study we assessed the impact that prolonged (six month) low dose rate neutron exposure had on the ability of male Wistar rats to perform in two executive function tasks (i.e. attentional set shifting (ATSET) - a constrained cognitive flexibility task and the UCFlex assay - an unconstrained cognitive flexibility task). In recognition of the fact that astronauts also have to contend with inadequate sleep quantity and quality for much of their time in space, we determined the impact that relatively mild sleep disruption had on the ability to perform in the ATSET test in sham and neutron-irradiated rats. RESULTS Chronic low dose (18 cGy) and dose-rate (1 mGy/day) exposure of rats to mixed neutron and photon over the course of six months resulted in significant impairment of simple discrimination (SD) performance. Should similar effects occur in astronauts subjected to low dose rate exposure to Space Radiation, the impairment of SD performance would result in a decreased ability to identify and learn the 'rules' required to respond to a new task or situation. Analysis of the behavioral data by kernel density estimation revealed that 40% of rats had severe ATSET impairments. This value may be a best-case scenario because exposure to neutrons also adversely impacted performance in the UCFlex task. Furthermore, when the good performing rats were reevaluated after they had been subjected to sleep fragmentation, additional ATSET performance decrements were observed in the set shifting stages of the ATSET test, with only 7.4% of the neutron exposed rats able to successfully perform ATSET under normal and sleep fragmented conditions, as opposed to ∼55% of shams. CONCLUSION Protracted low dose and low dose rate neutron exposures impairs executive functions in a high percentage of rats that were normally rested, however further detriments in performance become evident when the rats are subjected to sleep fragmentation.
Collapse
Affiliation(s)
- Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, USA
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA
- Leroy T Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Vania D Duncan
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Arriyam S Fesshaye
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Laurie L Wellman
- Department of Pathology & Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Christina M Fallgren
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Larry D Sanford
- Department of Pathology & Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
63
|
Zhang ZQ, Ding JW, Wang XA, Luo CY, Yu B, Zheng XX, Zhou T, Shang BX, Tong XH, Zhang J. Abnormal circadian rhythms are associated with plaque instability in acute coronary syndrome patients. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3761-3771. [PMID: 31933764 PMCID: PMC6949736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/29/2019] [Indexed: 06/10/2023]
Abstract
AIM Acute coronary syndrome (ACS), a leading cause of morbidity and mortality worldwide, is among the most serious cardiovascular diseases. Circadian rhythms are present in almost all organisms. In clinical practice, we have found that ACS is closely related to these circadian rhythms. However, the relationship between circadian rhythms and plaque instability in ACS patients is incompletely understood. The aim of this study is to provide new insights into the relationship between circadian rhythms and plaque instability in ACS patients. METHODS We enrolled patients with ACS and individuals with normal coronary artery function in this study. The Athens Insomnia Scale (AIS), Pittsburgh Sleep Quality Index (PSQI), International Physical Activity Questionnaire (IPAQ) and Healthy Diet Score (HDS) were used to evaluate circadian rhythms. Furthermore, quantitative real-time polymerase chain reaction (qRT-PCR) was used to assess the mRNA expression levels of muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1), circadian locomotor output cycles kaput (Clock), Cryptochrome1 (Cry1), Period2 (Per2), nuclear receptor subfamily 1, group D, member 1 (Rev-erbα), and matrix metalloproteinases MMP2 and MMP9. RESULTS AIS scores and PSQI scores were significantly higher in patients with ST segment elevation myocardial infarction (STEMI), non-ST segment elevation myocardial infarction (NSTEMI), and unstable angina pectoris (UA) than in the normal controls (NCs) (P < 0.05). The IPAQ scores of the NCs and patients with UA were significantly higher than in patients with STEMI and NSTEMI (P < 0.05). Notably higher HDS scores were recorded for the NCs compared to those of patients with UA, NSTEMI, and STEMI (P < 0.05). Consistent with these findings, compared with the NCs, the lowest levels of Bmal1, Clock, Cry1, Per2 and Rev-erbα mRNAs were detected in patients with STEMI, followed by patients with NSTEMI and then patients with UA (P < 0.05). Furthermore, the levels of MMP2 and MMP9 mRNA were significantly higher in the patients with STEMI, NSTEMI, and UA than those in the NCs (P < 0.05). In addition, we found that the levels of MMP mRNA negatively correlated with the levels of clock genes mRNAs (P < 0.05, respectively). CONCLUSIONS Based on our data, the circadian rhythms and clock genes are correlatively with the occurrence of ACS, and the expression levels of clock genes are negatively correlated with plaque stability in ACS patients.
Collapse
Affiliation(s)
- Zai-Qiang Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Jia-Wang Ding
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Xin-An Wang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Cai-Yun Luo
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Bin Yu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Xia-Xia Zheng
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Tian Zhou
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Bai-Xue Shang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Xiao-Hong Tong
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
- Institute of Cardiovascular Diseases, China Three Gorges UniversityYichang 443000, Hubei Province, PR China
| |
Collapse
|
64
|
Xie Y, Ba L, Wang M, Deng SY, Chen SM, Huang LF, Zhang M, Wang W, Ding FF. Chronic sleep fragmentation shares similar pathogenesis with neurodegenerative diseases: Endosome-autophagosome-lysosome pathway dysfunction and microglia-mediated neuroinflammation. CNS Neurosci Ther 2019; 26:215-227. [PMID: 31549780 PMCID: PMC6978272 DOI: 10.1111/cns.13218] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 12/15/2022] Open
Abstract
Aims Insufficient sleep has been found to result in varying degrees of cognitive impairment and emotional changes. Sleep was reported probably responsible for cleaning metabolic wastes in brain by increasing extracellular bulk flow. Herein, we propose that chronic sleep insufficiency in young adult wild‐type mice is also linked with dysfunction of intracellular protein degradation pathways and microglia‐mediated neuroinflammation, which are potentially important mechanisms in the initiation of neurodegeneration. Methods We applied the chronic sleep fragmentation (CSF) model to induce chronic sleep insufficiency in wild‐type mice. After 2 months of CSF, cognitive function, amyloid‐β accumulation, dysfunction of endosome‐autophagosome‐lysosome pathway, and microglia activation were evaluated. Results Following CSF, impairment of spatial learning and memory, and aggravated anxiety‐like behavior in mice were identified by behavioral experiments. Increased intracellular amyloid‐β accumulation was observed in cortex and hippocampus. Mechanistically, CSF could significantly enhance the expression of Rab5 (early endosome marker), Rab7 (late endosome marker), as well as LC3B (autophagosome marker), and autophagy‐positive regulatory factors in brain detected by immunofluorescent staining and Western blot. In addition, activation of microglia was evident by enhanced CD68, CD16/32, and CD206 levels after CSF treatment. Conclusions Chronic sleep fragmentation could initiate pathogenetic processes similar to the early stage of neurodegeneration, including dysfunction of endosome‐autophagosome‐lysosome pathway and microglia‐mediated neuroinflammation. Our findings further strengthen the link between chronic sleep insufficiency and the initiation of neurodegeneration even if lack of genetic predisposition.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Ba
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sai-Yue Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Miao Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Fang Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Diseases of Chinese Ministry of Education, The School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng-Fei Ding
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
65
|
Untargeted metabolomics analysis of rat hippocampus subjected to sleep fragmentation. Brain Res Bull 2019; 153:74-83. [PMID: 31419538 DOI: 10.1016/j.brainresbull.2019.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 07/25/2019] [Accepted: 08/10/2019] [Indexed: 01/08/2023]
Abstract
Sleep fragmentation (SF) commonly occurs in several pathologic conditions and is especially associated with impairments of hippocampus-dependent neurocognitive functions. Although the effects of SF on hippocampus in terms of protein or gene levels were examined in several studies, the impact of SF at the metabolite level has not been investigated. Thus, in this study, the differentially expressed large-scale metabolite profiles of hippocampus in a rat model of SF were investigated using untargeted metabolomics approaches. Forty-eight rats were divided into the following 4 groups: 4-day SF group, 4-day exercise control (EC) group, 15-day SF group, and 15-day EC group (n = 12, each). SF was accomplished by forced exercise using a walking wheel system with 30-s on/90-s off cycles, and EC condition was set at 10-min on/30-min off. The metabolite profiles of rat hippocampi in the SF and EC groups were analyzed using liquid chromatography/mass spectrometry. Multivariate analysis revealed distinctive metabolic profiles and marker signals between the SF and corresponding EC groups. Metabolic changes were significant only in the 15-day SF group. In the 15-day SF group, L-tryptophan, myristoylcarnitine, and palmitoylcarnitine were significantly increased, while adenosine monophosphate, hypoxanthine, L-glutamate, L-aspartate, L-methionine, and glycerophosphocholine were decreased compared to the EC group. The alanine, aspartate, and glutamate metabolism pathway was observed as the common key pathway in the 15-day SF groups. The results from this untargeted metabolomics study provide a perspective on metabolic impact of SF on the hippocampus.
Collapse
|
66
|
Mohammadipoor-Ghasemabad L, Sangtarash MH, Sheibani V, Sasan HA, Esmaeili-Mahani S. Hippocampal microRNA-191a-5p Regulates BDNF Expression and Shows Correlation with Cognitive Impairment Induced by Paradoxical Sleep Deprivation. Neuroscience 2019; 414:49-59. [DOI: 10.1016/j.neuroscience.2019.06.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 12/23/2022]
|
67
|
Cuddapah VA, Zhang SL, Sehgal A. Regulation of the Blood-Brain Barrier by Circadian Rhythms and Sleep. Trends Neurosci 2019; 42:500-510. [PMID: 31253251 PMCID: PMC6602072 DOI: 10.1016/j.tins.2019.05.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 01/09/2023]
Abstract
The blood-brain barrier (BBB) is an evolutionarily conserved, structural, and functional separation between circulating blood and the central nervous system (CNS). By controlling permeability into and out of the nervous system, the BBB has a critical role in the precise regulation of neural processes. Here, we review recent studies demonstrating that permeability at the BBB is dynamically controlled by circadian rhythms and sleep. An endogenous circadian rhythm in the BBB controls transporter function, regulating permeability across the BBB. In addition, sleep promotes the clearance of metabolites along the BBB, as well as endocytosis across the BBB. Finally, we highlight the implications of this regulation for diseases, including epilepsy.
Collapse
Affiliation(s)
- Vishnu Anand Cuddapah
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shirley L Zhang
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
68
|
Xu J, Qin Z, Li W, Li X, Shen H, Wang W. Effects of somatotropic axis on cognitive dysfunction of obstructive sleep apnea. Sleep Breath 2019; 24:175-182. [PMID: 31073904 DOI: 10.1007/s11325-019-01854-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Obstructive sleep apnea (OSA) is associated with a variety of neuroendocrine disorders and may lead to many complications, including cognitive dysfunction. The aim of this study was to assess the change of somatotropic axis and to detect the relation between somatotropic axis hormone and cognitive dysfunction. METHODS Sixty-six patients with OSA and 16 healthy controls were enrolled in this cross-sectional study. Cognitive function assessment using the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) and polysomnography were performed on all individuals. Blood samples were taken the next morning following the polysomnography and the level of serum growth hormone-releasing hormone (GHRH) and growth hormone (GH) were analyzed by enzyme-linked immunosorbent assay. RESULTS Compared with the control group, OSA patients showed significantly lower serum GH level (p < 0.05), whereas no statistical significance of GHRH level was found. In addition, lower MMSE and MoCA scores were found only in the severe OSA patients when compared with the controls. Furthermore, in severe OSA patients with cognitive dysfunction (MMSE score < 27 and MoCA score < 26), serum GHRH and GH levels were significantly lower than those without cognitive dysfunction. Logistic analysis revealed that cognitive dysfunction in severe OSA patients was associated with micro-arousal index and the level of serum GHRH and GH. CONCLUSION Decreased serum GH and GHRH levels were found among severe OSA patients with cognitive dysfunction who were overweight, which might promote the occurrence of cognitive dysfunction.
Collapse
Affiliation(s)
- Jiahuan Xu
- Institute of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Zheng Qin
- Institute of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Wenyang Li
- Institute of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xiaomeng Li
- Institute of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Hui Shen
- Institute of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Wei Wang
- Institute of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
69
|
Duncan MJ, Farlow H, Tirumalaraju C, Yun DH, Wang C, Howard JA, Sanden MN, O'Hara BF, McQuerry KJ, Bachstetter AD. Effects of the dual orexin receptor antagonist DORA-22 on sleep in 5XFAD mice. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:70-80. [PMID: 30859123 PMCID: PMC6396100 DOI: 10.1016/j.trci.2019.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction Sleep disruption is a characteristic of Alzheimer's disease (AD) that may exacerbate disease progression. This study tested whether a dual orexin receptor antagonist (DORA) would enhance sleep and attenuate neuropathology, neuroinflammation, and cognitive deficits in an AD-relevant mouse model, 5XFAD. Methods Wild-type (C57Bl6/SJL) and 5XFAD mice received chronic treatment with vehicle or DORA-22. Piezoelectric recordings monitored sleep and spatial memory was assessed via spontaneous Y-maze alternations. Aβ plaques, Aβ levels, and neuroinflammatory markers were measured by immunohistochemistry, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction, respectively. Results In 5XFAD mice, DORA-22 significantly increased light-phase sleep without reducing Aβ levels, plaque density, or neuroinflammation. Effects of DORA-22 on cognitive deficits could not be determined because the 5XFAD mice did not exhibit deficits. Discussion These findings suggest that DORAs may improve sleep in AD patients. Further investigations should optimize the dose and duration of DORA-22 treatment and explore additional AD-relevant animal models and cognitive tests.
Collapse
Affiliation(s)
- Marilyn J Duncan
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Hannah Farlow
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Chairtra Tirumalaraju
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Do-Hyun Yun
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Chanung Wang
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - James A Howard
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Madison N Sanden
- Department of Statistics, University of Kentucky, Lexington, KY, USA
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | | | - Adam D Bachstetter
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
70
|
Wilson H, Pagano G, Niccolini F, Muhlert N, Mehta MA, Searle G, Gunn RN, Rabiner EA, Foltynie T, Politis M. The role of phosphodiesterase 4 in excessive daytime sleepiness in Parkinson's disease. Parkinsonism Relat Disord 2019; 77:163-169. [PMID: 30824285 DOI: 10.1016/j.parkreldis.2019.02.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/09/2019] [Accepted: 02/18/2019] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Preclinical studies suggest a link between cAMP/PKA signalling, phosphodiesterase 4 (PDE4) expression and excessive daytime sleepiness (EDS). Here, we investigated in vivo the association between PDE4 expression and EDS in Parkinson's disease (PD) patients using [11C]rolipram PET and MR imaging. METHODS Eighteen participants, 12 PD and 6 healthy controls, underwent one [11C]rolipram PET and a multi-modal MRI scan. Probabilistic tractography was performed on subjects' diffusion data to functionally parcellate the striatum according with projections to limbic cortical areas. The severity of EDS was assessed using the Epworth Sleepiness Scale (ESS). To assess PDE4 expression in PD patients with EDS, the PD cohort was divided according to the presence (n = 5) or absence (n = 7) of EDS, defined using validated cut-off of score ≥10 on the ESS as score ≥10 on the ESS. RESULTS PD patients with EDS showed significantly increased [11C]rolipram volume of distribution (VT) in the caudate (P = 0.029), hypothalamus (P = 0.013), hippocampus (P = 0.036) and limbic striatum (P = 0.030) compared to patients without EDS. Furthermore, higher ESS scores correlated with increased [11C]rolipram VT in the caudate (r = 0.77; P = 0.003), hypothalamus (r = 0.84; P = 0.001), hippocampus (r = 0.81; P = 0.001) and limbic subdivisions of the striatum (r = 0.80; P = 0.003). CONCLUSION Our findings translate into humans preclinical data indicating that EDS is associated with elevated PDE4 in regions regulating sleep. The severity of EDS in PD was associated with elevated PDE4 expression; thus, suggesting a role of PDE4 in the pathophysiology of EDS in PD.
Collapse
Affiliation(s)
- Heather Wilson
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, United Kingdom
| | - Gennaro Pagano
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, United Kingdom
| | - Flavia Niccolini
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, United Kingdom
| | - Nils Muhlert
- School of Psychology and Cardiff University Brain Research Imaging Centre, Cardiff University, United Kingdom; Division of Neuroscience & Experimental Psychology, University of Manchester, United Kingdom
| | - Mitul A Mehta
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, United Kingdom
| | - Graham Searle
- Invicro London, Hammersmith Hospital, London, United Kingdom
| | - Roger N Gunn
- Invicro London, Hammersmith Hospital, London, United Kingdom; Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Eugenii A Rabiner
- Invicro London, Hammersmith Hospital, London, United Kingdom; Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, United Kingdom
| | - Thomas Foltynie
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology, London, United Kingdom
| | - Marios Politis
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, United Kingdom.
| |
Collapse
|
71
|
Smith DF, Amin RS. OSA and Cardiovascular Risk in Pediatrics. Chest 2019; 156:402-413. [PMID: 30790552 DOI: 10.1016/j.chest.2019.02.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 01/21/2019] [Accepted: 02/08/2019] [Indexed: 02/07/2023] Open
Abstract
OSA occurs in approximately 1% to 5% of children in the United States. Long-term cardiovascular risks associated with OSA in the adult population are well documented. Although changes in BP regulation occur in children with OSA, the pathways leading to chronic cardiovascular risks of OSA in children are less clear. Risk factors associated with cardiovascular disease in adult populations could carry the same future risk for children. It is imperative to determine whether known mechanisms of cardiovascular diseases in adults are like those that lead to pediatric disease. Early pathophysiologic changes may lead to a lifetime burden of cardiovascular disease and early mortality. With this perspective in mind, our review discusses pathways leading to cardiovascular pathology in children with OSA and provides a comprehensive overview of recent research findings related to cardiovascular sequelae in the pediatric population.
Collapse
Affiliation(s)
- David F Smith
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Otolaryngology-Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Raouf S Amin
- Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| |
Collapse
|
72
|
Nox1/Ref-1-mediated activation of CREB promotes Gremlin1-driven endothelial cell proliferation and migration. Redox Biol 2019; 22:101138. [PMID: 30802716 PMCID: PMC6395885 DOI: 10.1016/j.redox.2019.101138] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 01/21/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex degenerative disorder marked by aberrant vascular remodeling associated with hyperproliferation and migration of endothelial cells (ECs). Previous reports implicated bone morphogenetic protein antagonist Gremlin 1 in this process; however, little is known of the molecular mechanisms involved. The current study was designed to test whether redox signaling initiated by NADPH oxidase 1 (Nox1) could promote transcription factor CREB activation by redox factor 1 (Ref-1), transactivation of Gremlin1 transcription, EC migration, and proliferation. Human pulmonary arterial EC (HPAECs) exposed in vitro to hypoxia to recapitulate PAH signaling displayed induced Nox1 expression, reactive oxygen species (ROS) production, PKA activity, CREB phosphorylation, and CREB:CRE motif binding. These responses were abrogated by selective Nox1 inhibitor NoxA1ds and/or siRNA Nox1. Nox1-activated CREB migrated to the nucleus and bound to Ref-1 leading to CREB:CRE binding and Gremlin1 transcription. CHiP assay and CREB gene-silencing illustrated that CREB is pivotal for hypoxia-induced Gremlin1, which, in turn, stimulates EC proliferation and migration. In vivo, participation of Nox1, CREB, and Gremlin1, as well as CREB:CRE binding was corroborated in a rat PAH model. Activation of a previously unidentified Nox1-PKA-CREB/Ref-1 signaling pathway in pulmonary endothelial cells leads to Gremlin1 transactivation, proliferation and migration. These findings reveal a new signaling pathway by which Nox1 via induction of CREB and Gremlin1 signaling contributes to vascular remodeling and provide preclinical indication of its significance in PAH.
Collapse
|
73
|
Obstructive Sleep Apnea and Inflammation: Proof of Concept Based on Two Illustrative Cytokines. Int J Mol Sci 2019; 20:ijms20030459. [PMID: 30678164 PMCID: PMC6387387 DOI: 10.3390/ijms20030459] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/12/2019] [Accepted: 01/15/2019] [Indexed: 12/19/2022] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a markedly prevalent condition across the lifespan, particularly in overweight and obese individuals, which has been associated with an independent risk for neurocognitive, behavioral, and mood problems as well as cardiovascular and metabolic morbidities, ultimately fostering increases in overall mortality rates. In adult patients, excessive daytime sleepiness (EDS) is the most frequent symptom leading to clinical referral for evaluation and treatment, but classic EDS features are less likely to be reported in children, particularly among those with normal body-mass index. The cumulative evidence collected over the last two decades supports a conceptual framework, whereby sleep-disordered breathing in general and more particularly OSAS should be viewed as low-grade chronic inflammatory diseases. Accordingly, it is assumed that a proportion of the morbid phenotypic signature in OSAS is causally explained by underlying inflammatory processes inducing end-organ dysfunction. Here, the published links between OSAS and systemic inflammation will be critically reviewed, with special focus on the pro-inflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6), since these constitute classical prototypes of the large spectrum of inflammatory molecules that have been explored in OSAS patients.
Collapse
|
74
|
McLain JM, Alami WH, Glovak ZT, Cooley CR, Burke SJ, Collier JJ, Baghdoyan HA, Karlstad MD, Lydic R. Sleep fragmentation delays wound healing in a mouse model of type 2 diabetes. Sleep 2018; 41:5070462. [PMID: 30107617 PMCID: PMC6231532 DOI: 10.1093/sleep/zsy156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/04/2018] [Indexed: 12/15/2022] Open
Abstract
Study Objectives This study tested the hypothesis that sleep fragmentation (SF) delays wound healing in obese B6.BKS(D)-Leprdb/J (db/db) mice with impaired leptin signaling and type 2 diabetes compared with wild-type C57BL/6J (B6) mice. Methods Adult male mice (n = 34) were anesthetized and bilateral full-thickness excisional wounds were created on the back of each mouse. Half of the db/db and B6 mice were housed in SF cages equipped with a bar that moved across the cage floor every 2 min, 12 hr/day for 23 days. The other half of each group of mice was housed in the same room and did not experience SF. The dependent measures were number of days required to achieve wound closure, mRNA expression of four inflammatory mediators, blood glucose, insulin, and corticosterone. Results SF in the db/db mice caused a significant delay in wound healing relative to db/db mice with no SF. Days to achieve 50 per cent wound healing were 13.3 ± 0.4 with SF compared with 10.3 ± 0.7 without SF. All B6 mice achieved 50 per cent wound healing within 6 days and complete healing after 16 days. SF caused a significant increase in wound levels of TNF-α mRNA only in the db/db mice and an increase in corticosterone only in the B6 mice. Conclusions The delayed wound healing in obese, diabetic mice caused by SF is homologous to delayed wound healing in some patients with type 2 diabetes. The results support the interpretation that altered leptinergic signaling and inflammatory proteins contribute to delayed wound healing.
Collapse
Affiliation(s)
- John Mark McLain
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Wateen H Alami
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Zachary T Glovak
- Department of Psychology, The University of Tennessee, Knoxville, TN
| | - Chris R Cooley
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Susan J Burke
- Pennington Biomedical Research Center, Baton Rouge, LA
| | | | - Helen A Baghdoyan
- Department of Psychology, The University of Tennessee, Knoxville, TN
- Department of Anesthesiology, Graduate School of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Oak Ridge National Laboratory, Oak Ridge, TN
| | - Michael D Karlstad
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Ralph Lydic
- Department of Psychology, The University of Tennessee, Knoxville, TN
- Department of Anesthesiology, Graduate School of Medicine, University of Tennessee Health Science Center, Memphis, TN
- Oak Ridge National Laboratory, Oak Ridge, TN
| |
Collapse
|
75
|
Pak VM, Mazzotti DR, Keenan BT, Hirotsu C, Gehrman P, Bittencourt L, Pack AI, Tufik S. Candidate gene analysis in the São Paulo Epidemiologic Sleep Study (EPISONO) shows an association of variant in PDE4D and sleepiness. Sleep Med 2018; 47:106-112. [DOI: 10.1016/j.sleep.2017.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/30/2017] [Indexed: 12/24/2022]
|
76
|
Trzepizur W, Cortese R, Gozal D. Murine models of sleep apnea: functional implications of altered macrophage polarity and epigenetic modifications in adipose and vascular tissues. Metabolism 2018; 84:44-55. [PMID: 29154950 PMCID: PMC5955762 DOI: 10.1016/j.metabol.2017.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
Abstract
Obstructive sleep apnea (OSA) is a highly prevalent disease across the lifespan, is characterized by chronic intermittent hypoxia and sleep fragmentation, and has been independently associated with substantial cardiometabolic morbidity. However, the reversibility of end-organ morbidity with treatment is not always apparent, suggesting that both tissue remodeling and epigenetic mechanisms may be operationally involved. Here, we review the cumulative evidence focused around murine models of OSA to illustrate the temporal dependencies of cardiometabolic dysfunction and its reversibility, and more particularly to discuss the critical contributions of tissue macrophages to adipose tissue insulin resistance and vascular atherogenesis. In addition, we describe initial findings potentially implicating epigenetic alterations in both the emergence of the cardiometabolic morbidity of OSA, and in its reversibility with treatment. We anticipate that improved understanding of macrophage biology and epigenetics in the context of intermittent hypoxia and sleep fragmentation will lead to discovery of novel therapeutic targets and improved cardiovascular and metabolic outcomes in OSA.
Collapse
Affiliation(s)
- Wojciech Trzepizur
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, United States; Département de Pneumologie, Centre de Recherche Clinique, CHU d'Angers, Université Bretagne Loire, UNIV Angers, INSERM UMR 1063, Angers, France
| | - Rene Cortese
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, United States
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, United States.
| |
Collapse
|
77
|
Sleep-disordered breathing, circulating exosomes, and insulin sensitivity in adipocytes. Int J Obes (Lond) 2018; 42:1127-1139. [PMID: 29892042 PMCID: PMC6195831 DOI: 10.1038/s41366-018-0099-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/20/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sleep-disordered-breathing (SDB), which is characterized by chronic intermittent hypoxia (IH) and sleep fragmentation (SF), is a prevalent condition that promotes metabolic dysfunction, particularly among patients suffering from obstructive hypoventilation syndrome (OHS). Exosomes are generated ubiquitously, are readily present in the circulation, and their cargo may exert substantial functional cellular alterations in both physiological and pathological conditions. However, the effects of plasma exosomes on adipocyte metabolism in patients with OHS or in mice subjected to IH or SF mimicking SDB are unclear. METHODS Exosomes from fasting morning plasma samples from obese adults with polysomnographically-confirmed OSA before and after 3 months of adherent CPAP therapy were assayed. In addition, C57BL/6 mice were randomly assigned to (1) sleep control (SC), (2) sleep fragmentation (SF), and (3) intermittent hypoxia (HI) for 6 weeks, and plasma exosomes were isolated. Equivalent exosome amounts were added to differentiated adipocytes in culture, after which insulin sensitivity was assessed using 0 nM and 5 nM insulin-induced pAKT/AKT expression changes by western blotting. RESULTS When plasma exosomes were co-cultured and internalized by human naive adipocytes, significant reductions emerged in Akt phosphorylation responses to insulin when compared to exosomes obtained after 24 months of adherent CPAP treatment (n = 24; p < 0.001), while no such changes occur in untreated patients (n = 8). In addition, OHS exosomes induced significant increases in adipocyte lipolysis that were attenuated after CPAP, but did not alter pre-adipocyte differentiation. Similarly, exosomes from SF- and IH-exposed mice induced attenuated p-AKT/total AKT responses to exogenous insulin and increased glycerol content in naive murine adipocytes, without altering pre-adipocyte differentiation. CONCLUSIONS Using in vitro adipocyte-based functional reporter assays, alterations in plasma exosomal cargo occur in SDB, and appear to contribute to adipocyte metabolic dysfunction. Further exploration of exosomal miRNA signatures in either human subjects or animal models and their putative organ and cell targets appears warranted.
Collapse
|
78
|
Hurtado-Alvarado G, Becerril-Villanueva E, Contis-Montes de Oca A, Domínguez-Salazar E, Salinas-Jazmín N, Pérez-Tapia SM, Pavon L, Velázquez-Moctezuma J, Gómez-González B. The yin/yang of inflammatory status: Blood-brain barrier regulation during sleep. Brain Behav Immun 2018; 69:154-166. [PMID: 29154957 DOI: 10.1016/j.bbi.2017.11.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022] Open
Abstract
Sleep loss induces a low-grade inflammatory status characterized by a subtle but sustained increase of pro-inflammatory mediators, which are key regulators of blood-brain barrier function. To investigate the influence of inflammatory status on blood-brain barrier dysfunction induced by sleep restriction we performed an experiment using two strains of mice with different immunological backgrounds, C57BL/6 mice that have a predominant pro-inflammatory response and BALB/c mice that have a predominant anti-inflammatory response. Mice were sleep-restricted during 10 days using the flowerpot technique during 20 h per day with 4 h of daily sleep opportunity. The systemic inflammatory status, blood-brain barrier permeability, and the hippocampal expression of neuroinflammatory markers were characterized at the 10th day. Serum levels of TNF and IFN-γ increased in sleep-restricted C57BL/6 but not in BALB/c mice; no changes in other cytokines were found. Sleep restriction increased blood-brain barrier permeability in C57BL/6 strain but not in BALB/c. The hippocampus of sleep-restricted C57BL/6 mice exhibited an increase in the expression of the neuroinflammatory markers Iba-1, A2A adenosine receptor, and MMP-9; meanwhile in sleep-restricted BALB/c mice the expression of this markers was lesser than the control group. These data suggest that cytokines may be playing a key role in modulating blood-brain barrier function during sleep restriction, and probably the effects are related to Iba-1, MMP-9 and A2A adenosine receptor overexpression.
Collapse
Affiliation(s)
- G Hurtado-Alvarado
- Area of Neurosciences, Dept. Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - E Becerril-Villanueva
- Dept. Psychoimmunology, National Institute of Psychiatry, "Ramón de la Fuente", Mexico City, Mexico
| | | | - E Domínguez-Salazar
- Area of Neurosciences, Dept. Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - N Salinas-Jazmín
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - S M Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico; Dept. Immunology, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - L Pavon
- Dept. Psychoimmunology, National Institute of Psychiatry, "Ramón de la Fuente", Mexico City, Mexico
| | - J Velázquez-Moctezuma
- Area of Neurosciences, Dept. Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - B Gómez-González
- Area of Neurosciences, Dept. Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico.
| |
Collapse
|
79
|
Fisk AS, Tam SKE, Brown LA, Vyazovskiy VV, Bannerman DM, Peirson SN. Light and Cognition: Roles for Circadian Rhythms, Sleep, and Arousal. Front Neurol 2018; 9:56. [PMID: 29479335 PMCID: PMC5811463 DOI: 10.3389/fneur.2018.00056] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/22/2018] [Indexed: 01/12/2023] Open
Abstract
Light exerts a wide range of effects on mammalian physiology and behavior. As well as synchronizing circadian rhythms to the external environment, light has been shown to modulate autonomic and neuroendocrine responses as well as regulating sleep and influencing cognitive processes such as attention, arousal, and performance. The last two decades have seen major advances in our understanding of the retinal photoreceptors that mediate these non-image forming responses to light, as well as the neural pathways and molecular mechanisms by which circadian rhythms are generated and entrained to the external light/dark (LD) cycle. By contrast, our understanding of the mechanisms by which lighting influences cognitive processes is more equivocal. The effects of light on different cognitive processes are complex. As well as the direct effects of light on alertness, indirect effects may also occur due to disrupted circadian entrainment. Despite the widespread use of disrupted LD cycles to study the role circadian rhythms on cognition, the different experimental protocols used have subtly different effects on circadian function which are not always comparable. Moreover, these protocols will also disrupt sleep and alter physiological arousal, both of which are known to modulate cognition. Studies have used different assays that are dependent on different cognitive and sensory processes, which may also contribute to their variable findings. Here, we propose that studies addressing the effects of different lighting conditions on cognitive processes must also account for their effects on circadian rhythms, sleep, and arousal if we are to fully understand the physiological basis of these responses.
Collapse
Affiliation(s)
- Angus S Fisk
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Shu K E Tam
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Laurence A Brown
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Vladyslav V Vyazovskiy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Stuart N Peirson
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
80
|
Tumor necrosis factor alpha in sleep regulation. Sleep Med Rev 2017; 40:69-78. [PMID: 29153862 DOI: 10.1016/j.smrv.2017.10.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/16/2017] [Accepted: 10/18/2017] [Indexed: 12/14/2022]
Abstract
This review details tumor necrosis factor alpha (TNF) biology and its role in sleep, and describes how TNF medications influence sleep/wake activity. Substantial evidence from healthy young animals indicates acute enhancement or inhibition of endogenous brain TNF respectively promotes and inhibits sleep. In contrast, the role of TNF in sleep in most human studies involves pathological conditions associated with chronic elevations of systemic TNF and disrupted sleep. Normalization of TNF levels in such patients improves sleep. A few studies involving normal healthy humans and their TNF levels and sleep are consistent with the animal studies but are necessarily more limited in scope. TNF can act on established sleep regulatory circuits to promote sleep and on the cortex within small networks, such as cortical columns, to induce sleep-like states. TNF affects multiple synaptic functions, e.g., its role in synaptic scaling is firmly established. The TNF-plasticity actions, like its role in sleep, can be local network events suggesting that sleep and plasticity share biochemical regulatory mechanisms and thus may be inseparable from each other. We conclude that TNF is involved in sleep regulation acting within an extensive tightly orchestrated biochemical network to niche-adapt sleep in health and disease.
Collapse
|
81
|
Wadhwa M, Prabhakar A, Ray K, Roy K, Kumari P, Jha PK, Kishore K, Kumar S, Panjwani U. Inhibiting the microglia activation improves the spatial memory and adult neurogenesis in rat hippocampus during 48 h of sleep deprivation. J Neuroinflammation 2017; 14:222. [PMID: 29141671 PMCID: PMC5688670 DOI: 10.1186/s12974-017-0998-z] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/08/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Sleep deprivation (SD) leads to cognitive impairment. Neuroinflammation could be a significant contributing factor in the same. An increase in regional brain pro-inflammatory cytokines induces cognitive deficits, however, the magnitude of the effect under SD is not apparent. It is plausible that microglia activation could be involved in the SD-induced cognitive impairment by modulation of neuronal cell proliferation, differentiation, and brain-derived neuronal factor (BDNF) level. The present study aimed to evaluate the possible beneficial effect of minocycline in amelioration of spatial memory decline during SD by its anti-inflammatory and neuroprotective actions. We scrutinized the effect of minocycline on the inflammatory cytokine levels associated with glial cells (microglia and astrocytes) activity and neurogenesis markers crucial for behavioral functions during SD. METHODS Male Sprague-Dawley rats weighing 230-250 g were sleep deprived for 48 h using automated cage shaking apparatus. The spatial memory was tested using MWM apparatus immediately after completion of SD with and without minocycline. The animals were euthanized, blood was collected, and brain was extracted for neuroinflammation and neurogenesis studies. The set of experiments were also conducted with use of temozolomide, a neurogenesis blocker. RESULTS Minocycline treatment increased the body weight, food intake, and spatial memory performance which declined during SD. It reduced the pro-inflammatory and increased the anti-inflammatory cytokine levels in hippocampus and plasma and inhibited the reactive gliosis in the hippocampus evidenced by improved cell count, morphology, and immunoreactivity. Additionally, minocycline administration promoted neurogenesis at different stages: proliferation (BrdU, Ki-67), differentiation (DCX) cells and growth factor (BDNF). However, no significant change was observed in maturation (NeuN) during SD. In addition, molecules related to behavior, inflammation, and neurogenesis were shown to be more affected after temozolomide administration during SD, and changes were restored with minocycline treatment. We observed a significant correlation of neurogenesis with microglial activation, cytokine levels, and spatial memory during SD. CONCLUSION The present study demonstrated that the SD-induced decline in spatial memory, neuronal cells proliferation, differentiation, and BDNF level could be attributed to upregulation of neuroinflammatory molecules, and minocycline may be an effective intervention to counteract these changes. Microglial activation is involved in SD-induced changes in inflammatory molecules, neurogenesis, and spatial memory.
Collapse
Affiliation(s)
- Meetu Wadhwa
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
| | - Amit Prabhakar
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
| | - Koushik Ray
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
| | - Koustav Roy
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
| | - Punita Kumari
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
| | - Prabhash Kumar Jha
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
| | - Krishna Kishore
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
| | - Sanjeev Kumar
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
| | - Usha Panjwani
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, India
- Neurophysiology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, -110 054 India
| |
Collapse
|
82
|
Wadhwa M, Kumari P, Chauhan G, Roy K, Alam S, Kishore K, Ray K, Panjwani U. Sleep deprivation induces spatial memory impairment by altered hippocampus neuroinflammatory responses and glial cells activation in rats. J Neuroimmunol 2017; 312:38-48. [DOI: 10.1016/j.jneuroim.2017.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/04/2017] [Accepted: 09/06/2017] [Indexed: 01/03/2023]
|
83
|
Nadjar A, Wigren HKM, Tremblay ME. Roles of Microglial Phagocytosis and Inflammatory Mediators in the Pathophysiology of Sleep Disorders. Front Cell Neurosci 2017; 11:250. [PMID: 28912686 PMCID: PMC5582207 DOI: 10.3389/fncel.2017.00250] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/07/2017] [Indexed: 11/13/2022] Open
Abstract
Sleep serves crucial learning and memory functions in both nervous and immune systems. Microglia are brain immune cells that actively maintain health through their crucial physiological roles exerted across the lifespan, including phagocytosis of cellular debris and orchestration of neuroinflammation. The past decade has witnessed an explosive growth of microglial research. Considering the recent developments in the field of microglia and sleep, we examine their possible impact on various pathological conditions associated with a gain, disruption, or loss of sleep in this focused mini-review. While there are extensive studies of microglial implication in a variety of neuropsychiatric and neurodegenerative diseases, less is known regarding their roles in sleep disorders. It is timely to stimulate new research in this emergent and rapidly growing field of investigation.
Collapse
Affiliation(s)
- Agnes Nadjar
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche AgronomiqueBordeaux, France.,Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux UniversityBordeaux, France.,OptiNutriBrain International Associated Laboratory (NutriNeuro France-INAF Canada)Québec, QC, Canada
| | | | - Marie-Eve Tremblay
- Axe Neurosciences, CRCHU de Québec-Université LavalQuébec, QC, Canada.,Département de médecine moléculaire, Université LavalQuébec, QC, Canada
| |
Collapse
|
84
|
Zhao Z, Zhao X, Veasey SC. Neural Consequences of Chronic Short Sleep: Reversible or Lasting? Front Neurol 2017; 8:235. [PMID: 28620347 PMCID: PMC5449441 DOI: 10.3389/fneur.2017.00235] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/12/2017] [Indexed: 12/22/2022] Open
Abstract
Approximately one-third of adolescents and adults in developed countries regularly experience insufficient sleep across the school and/or work week interspersed with weekend catch up sleep. This common practice of weekend recovery sleep reduces subjective sleepiness, yet recent studies demonstrate that one weekend of recovery sleep may not be sufficient in all persons to fully reverse all neurobehavioral impairments observed with chronic sleep loss, particularly vigilance. Moreover, recent studies in animal models demonstrate persistent injury to and loss of specific neuron types in response to chronic short sleep (CSS) with lasting effects on sleep/wake patterns. Here, we provide a comprehensive review of the effects of chronic sleep disruption on neurobehavioral performance and injury to neurons, astrocytes, microglia, and oligodendrocytes and discuss what is known and what is not yet established for reversibility of neural injury. Recent neurobehavioral findings in humans are integrated with animal model research examining long-term consequences of sleep loss on neurobehavioral performance, brain development, neurogenesis, neurodegeneration, and connectivity. While it is now clear that recovery of vigilance following short sleep requires longer than one weekend, less is known of the impact of CSS on cognitive function, mood, and brain health long term. From work performed in animal models, CSS in the young adult and short-term sleep loss in critical developmental windows can have lasting detrimental effects on neurobehavioral performance.
Collapse
Affiliation(s)
- Zhengqing Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiangxiang Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Sigrid C Veasey
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
85
|
McAlpine CS, Swirski FK. Circadian Influence on Metabolism and Inflammation in Atherosclerosis. Circ Res 2017; 119:131-41. [PMID: 27340272 DOI: 10.1161/circresaha.116.308034] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/11/2016] [Indexed: 11/16/2022]
Abstract
Many aspects of human health and disease display daily rhythmicity. The brain's suprachiasmic nucleus, which interprets recurring external stimuli, and autonomous molecular networks in peripheral cells together, set our biological circadian clock. Disrupted or misaligned circadian rhythms promote multiple pathologies including chronic inflammatory and metabolic diseases such as atherosclerosis. Here, we discuss studies suggesting that circadian fluctuations in the vessel wall and in the circulation contribute to atherogenesis. Data from humans and mice indicate that an impaired molecular clock, disturbed sleep, and shifting light-dark patterns influence leukocyte and lipid supply in the circulation and alter cellular behavior in atherosclerotic lesions. We propose that a better understanding of both local and systemic circadian rhythms in atherosclerosis will enhance clinical management, treatment, and public health policy.
Collapse
Affiliation(s)
- Cameron S McAlpine
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston.
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
86
|
Trzepizur W, Khalyfa A, Qiao Z, Popko B, Gozal D. Integrated stress response activation by sleep fragmentation during late gestation in mice leads to emergence of adverse metabolic phenotype in offspring. Metabolism 2017; 69:188-198. [PMID: 28139216 DOI: 10.1016/j.metabol.2017.01.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/16/2016] [Accepted: 01/16/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Late gestational sleep fragmentation (SF) is highly prevalent particularly in obese women, and induces metabolic dysfunction in adult offspring mice. SF induces activation of the integrated stress response (ISR), which might be involved in metabolic disorders. We hypothesized that adult offspring of double mutant mice (DM) involving the critical ISR genes CHOP and GADD34 would be protected from developing obesity and insulin resistance following SF. METHODS Time-pregnant CHOP/GADD34 DM and wild type (WT) mice were randomly assigned to sleep control (SC) or SF conditions during the last 5days of gestation. At 24-weeks of age, body weight, fat mass, and HOMA-IR were assessed in the offspring. Tregs lymphocytes, Lyc6chigh, M1 and M2 macrophages were examined in visceral white adipose tissues (vWAT) using flow cytometry. The effects of plasma exosomes on adipocyte cell line proliferation, differentiation and insulin sensitivity were also evaluated. RESULTS SF-WT male showed significant increases in body weight, vWAT mass and HOMA-IR compared to SC-WT mice, while SF had no effect in SF-DM mice. Inflammatory macrophages (Ly-6chigh) and the ratio of M1/M2 macrophages were increased while FoxP3+ Tregs counts were decreased in SF-WT but not in SF-DM mice. Exosomes from SF-WT, but not from the SF-DM offspring increased pre-adipocyte proliferation and differentiation, and decreased in vitro adipocyte insulin sensitivity. CONCLUSION Activation of the ISR during late gestation, as induced by late gestational SF, appears to underlie some of the transgenerational modifications in metabolic genes ultimately contributing to a metabolic syndrome phenotype in adult offspring.
Collapse
Affiliation(s)
- Wojciech Trzepizur
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Abdelnaby Khalyfa
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Zhuanhong Qiao
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Brian Popko
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, Chicago, IL 60637, United States
| | - David Gozal
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
87
|
Zhang K, Li YJ, Feng D, Zhang P, Wang YT, Li X, Liu SB, Wu YM, Zhao MG. Imbalance between TNFα and progranulin contributes to memory impairment and anxiety in sleep-deprived mice. Sci Rep 2017; 7:43594. [PMID: 28300056 PMCID: PMC5353617 DOI: 10.1038/srep43594] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/25/2017] [Indexed: 12/21/2022] Open
Abstract
Sleep disorder is becoming a widespread problem in current society, and is associated with impaired cognition and emotional disorders. Progranulin (PGRN), also known as granulin epithelin precursor, promotes neurite outgrowth and cell survival, and is encoded by the GRN gene. It is a tumor necrosis factor α receptor (TNFR) ligand which is implicated in many central nervous system diseases. However, the role PGRN in sleep disorder remains unclear. In the present study, we found that sleep deprivation (S-DEP) impaired the memory and produced thigmotaxis/anxiety-like behaviors in mice. S-DEP increased the levels of TNFα but decreased PGRN levels in the hippocampus. The intracerebroventricular (ICV) injection of PGRN or intraperitoneal injection of TNFα synthesis blocker thalidomide (25 mg/kg), prevented the memory impairment and anxiety behaviors induced by S-DEP. PGRN treatment also restored dendritic spine density in the hippocampus CA1 region and neurogenesis in hippocampus dentate gyrus (DG). These results indicate that an imbalance between TNFα and PGRN contributes to memory impairment and thigmotaxis/anxiety caused by sleep deprivation.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.,Precision Pharmacy &Drug Development Center, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yu-Jiao Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Dan Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.,Department of Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Peng Zhang
- Department of Neurobiology, Capital Medical University, Beijing, 100069, China
| | - Ya-Tao Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Ming-Gao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.,Precision Pharmacy &Drug Development Center, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| |
Collapse
|
88
|
Poroyko VA, Carreras A, Khalyfa A, Khalyfa AA, Leone V, Peris E, Almendros I, Gileles-Hillel A, Qiao Z, Hubert N, Farré R, Chang EB, Gozal D. Chronic Sleep Disruption Alters Gut Microbiota, Induces Systemic and Adipose Tissue Inflammation and Insulin Resistance in Mice. Sci Rep 2016; 6:35405. [PMID: 27739530 PMCID: PMC5064361 DOI: 10.1038/srep35405] [Citation(s) in RCA: 310] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/28/2016] [Indexed: 02/08/2023] Open
Abstract
Chronic sleep fragmentation (SF) commonly occurs in human populations, and although it does not involve circadian shifts or sleep deprivation, it markedly alters feeding behaviors ultimately promoting obesity and insulin resistance. These symptoms are known to be related to the host gut microbiota. Mice were exposed to SF for 4 weeks and then allowed to recover for 2 weeks. Taxonomic profiles of fecal microbiota were obtained prospectively, and conventionalization experiments were performed in germ-free mice. Adipose tissue insulin sensitivity and inflammation, as well as circulating measures of inflammation, were assayed. Effect of fecal water on colonic epithelial permeability was also examined. Chronic SF-induced increased food intake and reversible gut microbiota changes characterized by the preferential growth of highly fermentative members of Lachnospiraceae and Ruminococcaceae and a decrease of Lactobacillaceae families. These lead to systemic and visceral white adipose tissue inflammation in addition to altered insulin sensitivity in mice, most likely via enhanced colonic epithelium barrier disruption. Conventionalization of germ-free mice with SF-derived microbiota confirmed these findings. Thus, SF-induced metabolic alterations may be mediated, in part, by concurrent changes in gut microbiota, thereby opening the way for gut microbiome-targeted therapeutics aimed at reducing the major end-organ morbidities of chronic SF.
Collapse
Affiliation(s)
- Valeriy A Poroyko
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA.,Department of Medical Oncology, City of Hope, Duarte, CA, 91010, USA
| | - Alba Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Ahamed A Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Vanessa Leone
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Eduard Peris
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Isaac Almendros
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Alex Gileles-Hillel
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Zhuanhong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Nathaniel Hubert
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Ramon Farré
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, CIBER, Madrid, Spain.,Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain
| | - Eugene B Chang
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
89
|
Hurtado-Alvarado G, Domínguez-Salazar E, Pavon L, Velázquez-Moctezuma J, Gómez-González B. Blood-Brain Barrier Disruption Induced by Chronic Sleep Loss: Low-Grade Inflammation May Be the Link. J Immunol Res 2016; 2016:4576012. [PMID: 27738642 PMCID: PMC5050358 DOI: 10.1155/2016/4576012] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 08/14/2016] [Accepted: 08/24/2016] [Indexed: 12/16/2022] Open
Abstract
Sleep is a vital phenomenon related to immunomodulation at the central and peripheral level. Sleep deficient in duration and/or quality is a common problem in the modern society and is considered a risk factor to develop neurodegenerative diseases. Sleep loss in rodents induces blood-brain barrier disruption and the underlying mechanism is still unknown. Several reports indicate that sleep loss induces a systemic low-grade inflammation characterized by the release of several molecules, such as cytokines, chemokines, and acute-phase proteins; all of them may promote changes in cellular components of the blood-brain barrier, particularly on brain endothelial cells. In the present review we discuss the role of inflammatory mediators that increase during sleep loss and their association with general disturbances in peripheral endothelium and epithelium and how those inflammatory mediators may alter the blood-brain barrier. Finally, this manuscript proposes a hypothetical mechanism by which sleep loss may induce blood-brain barrier disruption, emphasizing the regulatory effect of inflammatory molecules on tight junction proteins.
Collapse
Affiliation(s)
- G. Hurtado-Alvarado
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - E. Domínguez-Salazar
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - L. Pavon
- Department of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente”, Mexico City, Mexico
| | - J. Velázquez-Moctezuma
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - B. Gómez-González
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| |
Collapse
|
90
|
Davies CR, Harrington JJ. Impact of Obstructive Sleep Apnea on Neurocognitive Function and Impact of Continuous Positive Air Pressure. Sleep Med Clin 2016; 11:287-98. [PMID: 27542875 DOI: 10.1016/j.jsmc.2016.04.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is evidence that obstructive sleep apnea (OSA) can negatively impact attention, memory, learning, executive function, and overall intellectual function in adults and children. Imaging techniques, including MRI, MR diffusion tensor imaging, MR spectroscopy, and fMRI, have provided additional insight into the anatomic and functional underpinnings of OSA-related cognitive impairment. Both animal and human studies have looked to elucidate the separate effects of oxygen desaturation and sleep fragmentation on independent aspects of cognition. Data from animal models point to neuro-inflammation and oxidative stress as driving factors of cognitive impairment.
Collapse
Affiliation(s)
- Charles R Davies
- Carle Neuroscience Institute, Carle Foundation Hospital, College of Medicine, University of Illinois at Urbana-Champaign, 602 West University Avenue, Urbana, IL 61801, USA.
| | - John J Harrington
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, 985990 Nebraska Medical Center, Omaha, NE 68159-5990, USA
| |
Collapse
|
91
|
Baud MO, Parafita J, Nguyen A, Magistretti PJ, Petit JM. Sleep fragmentation alters brain energy metabolism without modifying hippocampal electrophysiological response to novelty exposure. J Sleep Res 2016; 25:583-590. [PMID: 27136914 DOI: 10.1111/jsr.12419] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 02/13/2016] [Indexed: 02/06/2023]
Abstract
Sleep is viewed as a fundamental restorative function of the brain, but its specific role in neural energy budget remains poorly understood. Sleep deprivation dampens brain energy metabolism and impairs cognitive functions. Intriguingly, sleep fragmentation, despite normal total sleep duration, has a similar cognitive impact, and in this paper we ask the question of whether it may also impair brain energy metabolism. To this end, we used a recently developed mouse model of 2 weeks of sleep fragmentation and measured 2-deoxy-glucose uptake and glycogen, glucose and lactate concentration in different brain regions. In order to homogenize mice behaviour during metabolic measurements, we exposed them to a novel environment for 1 h. Using an intra-hippocampal electrode, we first showed that hippocampal electroencephalograph (EEG) response to exploration was unaltered by 1 or 14 days of sleep fragmentation. However, after 14 days, sleep fragmented mice exhibited a lower uptake of 2-deoxy-glucose in cortex and hippocampus and lower cortical lactate levels than control mice. Our results suggest that long-term sleep fragmentation impaired brain metabolism to a similar extent as total sleep deprivation without affecting the neuronal responsiveness of hippocampus to a novel environment.
Collapse
Affiliation(s)
- Maxime O Baud
- LNDC, Brain Mind Institute, Faculté des Sciences de la Vie, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Department of Neurology, UCSF, San Francisco, CA, USA
| | - Julia Parafita
- LNDC, Brain Mind Institute, Faculté des Sciences de la Vie, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Audrey Nguyen
- LNDC, Brain Mind Institute, Faculté des Sciences de la Vie, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pierre J Magistretti
- LNDC, Brain Mind Institute, Faculté des Sciences de la Vie, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,BESE Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.,Centre de Neurosciences Psychiatriques, Centre Hospitalier Universitaire Vaudois (CHUV), Prilly, Switzerland
| | - Jean-Marie Petit
- LNDC, Brain Mind Institute, Faculté des Sciences de la Vie, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland. .,Centre de Neurosciences Psychiatriques, Centre Hospitalier Universitaire Vaudois (CHUV), Prilly, Switzerland.
| |
Collapse
|
92
|
Gileles-Hillel A, Kheirandish-Gozal L, Gozal D. Biological plausibility linking sleep apnoea and metabolic dysfunction. Nat Rev Endocrinol 2016; 12:290-8. [PMID: 26939978 DOI: 10.1038/nrendo.2016.22] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obstructive sleep apnoea (OSA) is a very common disorder that affects 10-25% of the general population. In the past two decades, OSA has emerged as a cardiometabolic risk factor in both paediatric and adult populations. OSA-induced metabolic perturbations include dyslipidaemia, atherogenesis, liver dysfunction and abnormal glucose metabolism. The mainstay of treatment for OSA is adenotonsillectomy in children and continuous positive airway pressure therapy in adults. Although these therapies are effective at resolving the sleep-disordered breathing component of OSA, they do not always produce beneficial effects on metabolic function. Thus, a deeper understanding of the underlying mechanisms by which OSA influences metabolic dysfunction might yield improved therapeutic approaches and outcomes. In this Review, we summarize the evidence obtained from animal models and studies of patients with OSA of potential mechanistic pathways linking the hallmarks of OSA (intermittent hypoxia and sleep fragmentation) with metabolic dysfunction. Special emphasis is given to adipose tissue dysfunction induced by sleep apnoea, which bears a striking resemblance to adipose dysfunction resulting from obesity. In addition, important gaps in current knowledge and promising lines of future investigation are identified.
Collapse
Affiliation(s)
- Alex Gileles-Hillel
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| | - Leila Kheirandish-Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| | - David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| |
Collapse
|
93
|
Gozal D, Farré R, Nieto FJ. Putative Links Between Sleep Apnea and Cancer: From Hypotheses to Evolving Evidence. Chest 2016; 148:1140-1147. [PMID: 26020135 DOI: 10.1378/chest.15-0634] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In recent years, the potentially adverse role of sleep-disordered breathing in cancer incidence and outcomes has emerged. In parallel, animal models of intermittent hypoxia (IH) and sleep fragmentation (SF) emulating the two major components of OSA have lent support to the notion that OSA may enhance the proliferative and invasive properties of solid tumors. Based on several lines of evidence, we propose that OSA-induced increases in sympathetic outflow and alterations in immune function are critically involved in modifying oncologic processes including angiogenesis. In this context, we suggest that OSA, via IH (and potentially SF), promotes changes in several signaling pathways and transcription factors that coordinate malignant transformation and expansion, disrupts host immunologic surveillance, and consequently leads to increased probability of oncogenesis, accelerated tumor proliferation, and invasion, ultimately resulting in adverse outcomes.
Collapse
Affiliation(s)
- David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL.
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain; CIBER de Enfermedades Respiratorias, Bunyola, Spain
| | - F Javier Nieto
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI
| |
Collapse
|
94
|
The Interaction between Obstructive Sleep Apnea and Parkinson's Disease: Possible Mechanisms and Implications for Cognitive Function. PARKINSONS DISEASE 2015; 2015:849472. [PMID: 26509097 PMCID: PMC4609874 DOI: 10.1155/2015/849472] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/06/2015] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD) is a relentlessly progressive neurodegenerative disorder associated with hallmark motor and nonmotor symptoms (NMS) such as sleep disturbances and cognitive dysfunction. While dopaminergic treatments have improved the motor aspects of PD, progression remains inevitable. Research has recently increasingly focused on strategies to modify disease progression and on nonmotor manifestations of PD, given their impact on patients' quality of life. Obstructive sleep apnea (OSA) is a treatable sleep disorder, common in the general population, associated with excessive daytime sleepiness and neurocognitive deficits. Neuroimaging has demonstrated structural and functional changes in OSA patients; in animal models, OSA causes brain inflammation and oxidative injury, including in key areas involved in PD pathophysiology such as locus coeruleus. The prevalence of OSA in PD has been variable in studies to date, and potential consequences and interrelationship between the two disorders have not been well studied. There is however emerging evidence that OSA is associated with increased NMS in PD, particularly cognitive dysfunction. This review focuses on the possible interrelationship between OSA and PD. Mechanisms promoting OSA in PD will be reviewed, as well as mechanisms whereby OSA can affect the neurodegenerative process in PD.
Collapse
|
95
|
Zhu Y, Fenik P, Zhan G, Xin R, Veasey SC. Degeneration in Arousal Neurons in Chronic Sleep Disruption Modeling Sleep Apnea. Front Neurol 2015. [PMID: 26074865 DOI: 10.3389/fneur.2015.00109.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Chronic sleep disruption (CSD) is a cardinal feature of sleep apnea that predicts impaired wakefulness. Despite effective treatment of apneas and sleep disruption, patients with sleep apnea may have persistent somnolence. Lasting wake disturbances in treated sleep apnea raise the possibility that CSD may induce sufficient degeneration in wake-activated neurons (WAN) to cause irreversible wake impairments. Implementing a stereological approach in a murine model of CSD, we found reduced neuronal counts in representative WAN groups, locus coeruleus (LC) and orexinergic neurons, reduced by 50 and 25%, respectively. Mice exposed to CSD showed shortened sleep latencies lasting at least 4 weeks into recovery from CSD. As CSD results in frequent activation of WAN, we hypothesized that CSD promotes mitochondrial metabolic stress in WAN. In support, CSD increased lipofuscin within select WAN. Further, examining the LC as a representative WAN nucleus, we observed increased mitochondrial protein acetylation and down-regulation of anti-oxidant enzyme and brain-derived neurotrophic factor mRNA. Remarkably, CSD markedly increased tumor necrosis factor-alpha within WAN, and not in adjacent neurons or glia. Thus, CSD, as observed in sleep apnea, results in a composite of lasting wake impairments, loss of select neurons, a pro-inflammatory, pro-oxidative mitochondrial stress response in WAN, consistent with a degenerative process with behavioral consequences.
Collapse
Affiliation(s)
- Yan Zhu
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Polina Fenik
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Guanxia Zhan
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Ryan Xin
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Sigrid C Veasey
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
96
|
|
97
|
Gozal D, Farré R, Nieto FJ. Obstructive sleep apnea and cancer: Epidemiologic links and theoretical biological constructs. Sleep Med Rev 2015; 27:43-55. [PMID: 26447849 DOI: 10.1016/j.smrv.2015.05.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/21/2015] [Accepted: 05/25/2015] [Indexed: 12/19/2022]
Abstract
Sleep disorders have emerged as highly prevalent conditions in the last 50-75 y. Along with improved understanding of such disorders, the realization that perturbations in sleep architecture and continuity may initiate, exacerbate or modulate the phenotypic expression of multiple diseases including cancer has gained increased attention. Furthermore, the intermittent hypoxia that is attendant to sleep disordered breathing, has recently been implicated in increased incidence and more adverse prognosis of cancer. The unifying conceptual framework linking these associations proposes that increased sympathetic activity and/or alterations in immune function, particularly affecting innate immune cellular populations, underlie the deleterious effects of sleep disorders on tumor biology. In this review, the epidemiological evidence linking disrupted sleep and intermittent hypoxia to oncological outcomes, and the potential biological underpinnings of such associations as illustrated by experimental murine models will be critically appraised. The overarching conclusion appears supportive in the formulation of an hypothetical framework, in which fragmented sleep and intermittent hypoxia may promote changes in multiple signalosomes and transcription factors that can not only initiate malignant transformation, but will also alter the tumor microenvironment, disrupt immunosurveillance, and thus hasten tumor proliferation and increase local and metastatic invasion. Future bench-based experimental studies as well as carefully conducted and controlled clinical epidemiological studies appear justified for further exploration of these hypotheses.
Collapse
Affiliation(s)
- David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA.
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain; CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - F Javier Nieto
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
98
|
Zhu Y, Fenik P, Zhan G, Xin R, Veasey SC. Degeneration in Arousal Neurons in Chronic Sleep Disruption Modeling Sleep Apnea. Front Neurol 2015; 6:109. [PMID: 26074865 PMCID: PMC4443725 DOI: 10.3389/fneur.2015.00109] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/03/2015] [Indexed: 11/13/2022] Open
Abstract
Chronic sleep disruption (CSD) is a cardinal feature of sleep apnea that predicts impaired wakefulness. Despite effective treatment of apneas and sleep disruption, patients with sleep apnea may have persistent somnolence. Lasting wake disturbances in treated sleep apnea raise the possibility that CSD may induce sufficient degeneration in wake-activated neurons (WAN) to cause irreversible wake impairments. Implementing a stereological approach in a murine model of CSD, we found reduced neuronal counts in representative WAN groups, locus coeruleus (LC) and orexinergic neurons, reduced by 50 and 25%, respectively. Mice exposed to CSD showed shortened sleep latencies lasting at least 4 weeks into recovery from CSD. As CSD results in frequent activation of WAN, we hypothesized that CSD promotes mitochondrial metabolic stress in WAN. In support, CSD increased lipofuscin within select WAN. Further, examining the LC as a representative WAN nucleus, we observed increased mitochondrial protein acetylation and down-regulation of anti-oxidant enzyme and brain-derived neurotrophic factor mRNA. Remarkably, CSD markedly increased tumor necrosis factor-alpha within WAN, and not in adjacent neurons or glia. Thus, CSD, as observed in sleep apnea, results in a composite of lasting wake impairments, loss of select neurons, a pro-inflammatory, pro-oxidative mitochondrial stress response in WAN, consistent with a degenerative process with behavioral consequences.
Collapse
Affiliation(s)
- Yan Zhu
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Polina Fenik
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Guanxia Zhan
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Ryan Xin
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Sigrid C Veasey
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
99
|
Abstract
Diet, exercise, stress, and sleep are receiving attention as environmental modifiers of chronic inflammatory diseases, including atherosclerosis, the culprit condition of myocardial infarction and stroke. Accumulating data indicate that psychosocial stress and a high-fat, high-cholesterol diet aggravate cardiovascular disease, whereas regular physical activity and healthy sleeping habits help prevent it. Here, we raise the possibility that inflammation-associated leukocyte production plays a causal role in lifestyle effects on atherosclerosis progression. Specifically, we explore whether and how potent real-life disease modifiers influence hematopoiesis' molecular and cellular machinery. Lifestyle, we hypothesize, may rearrange hematopoietic topography, diverting production from the bone marrow to the periphery, thus propagating a quantitative and qualitative drift of the macrophage supply chain. These changes may involve progenitor-extrinsic and intrinsic communication nodes that connect organ systems along neuroimmune and immunometabolic axes, ultimately leading to an altered number and phenotype of lesional macrophages. We propose that, in conjunction with improved public health policy, future therapeutics could aim to modulate the quantitative and qualitative output, as well as the location, of the hematopoietic tree to decrease the risk of atherosclerosis complications.
Collapse
Affiliation(s)
- Matthias Nahrendorf
- From the Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston.
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston.
| |
Collapse
|
100
|
Dumaine JE, Ashley NT. Acute sleep fragmentation induces tissue-specific changes in cytokine gene expression and increases serum corticosterone concentration. Am J Physiol Regul Integr Comp Physiol 2015; 308:R1062-9. [PMID: 25876653 DOI: 10.1152/ajpregu.00049.2015] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/10/2015] [Indexed: 12/13/2022]
Abstract
Sleep deprivation induces acute inflammation and increased glucocorticosteroids in vertebrates, but effects from fragmented, or intermittent, sleep are poorly understood. Considering the latter is more representative of sleep apnea in humans, we investigated changes in proinflammatory (IL-1β, TNF-α) and anti-inflammatory (TGF-β1) cytokine gene expression in the periphery (liver, spleen, fat, and heart) and brain (hypothalamus, prefrontal cortex, and hippocampus) of a murine model exposed to varying intensities of sleep fragmentation (SF). Additionally, serum corticosterone was assessed. Sleep was disrupted in male C57BL/6J mice using an automated sleep fragmentation chamber that moves a sweeping bar at specified intervals (Lafayette Industries). Mice were exposed to bar sweeps every 20 s (high sleep fragmentation, HSF), 120 s (low sleep fragmentation, LSF), or the bar remained stationary (control). Trunk blood and tissue samples were collected after 24 h of SF. We predicted that HSF mice would exhibit increased proinflammatory expression, decreased anti-inflammatory expression, and elevated stress hormones in relation to LSF and controls. SF significantly elevated IL-1β gene expression in adipose tissue, heart (HSF only), and hypothalamus (LSF only) relative to controls. SF did not increase TNF-α expression in any of the tissues measured. HSF increased TGF-β1 expression in the hypothalamus and hippocampus relative to other groups. Serum corticosterone concentration was significantly different among groups, with HSF mice exhibiting the highest, LSF intermediate, and controls with the lowest concentration. This indicates that 24 h of SF is a potent inducer of inflammation and stress hormones in the periphery, but leads to upregulation of anti-inflammatory cytokines in the brain.
Collapse
Affiliation(s)
- Jennifer E Dumaine
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| | - Noah T Ashley
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| |
Collapse
|