1
|
Brice KN, Braden-Kuhle PN, Miller SK, Regan A, Lacy V, Chumley MJ, Boehm GW. Chronic sleep loss alters the inflammatory response and BDNF expression in C57BL/6J mice. J Neuroimmunol 2024; 396:578462. [PMID: 39378741 DOI: 10.1016/j.jneuroim.2024.578462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/30/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
Although adequate sleep is imperative for proper physiological function, over one-third of US adults obtain insufficient sleep. The current research investigated the impact of chronic sleep restriction (CSR) on inflammatory markers and hippocampal BDNF mRNA, following an immune insult in both male and female mice. Patterns of cytokine expression were different when the study was done in males vs. females, indicating potential sex differences in the inflammatory response following CSR. Further, CSR led to suppressed hippocampal BDNF expression in males, an effect not observed in females. These data suggest a complex interaction between chronic sleep loss, inflammation, and sex that warrants further exploration.
Collapse
Affiliation(s)
- Kelly N Brice
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, United States of America.
| | - Paige N Braden-Kuhle
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Shelby K Miller
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Allison Regan
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Vivienne Lacy
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Michael J Chumley
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| | - Gary W Boehm
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, United States of America
| |
Collapse
|
2
|
Yin C, Zhang M, Jin S, Zhou Y, Ding L, Lv Q, Huang Z, Zhou J, Chen J, Wang P, Zhang S, You Q. Mechanism of Salvia miltiorrhiza Bunge extract to alleviate Chronic Sleep Deprivation-Induced cognitive dysfunction in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155725. [PMID: 38772181 DOI: 10.1016/j.phymed.2024.155725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/16/2024] [Accepted: 05/07/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Bidirectional communication between the gut microbiota and the brain may play an essential role in the cognitive dysfunction associated with chronic sleep deprivation(CSD). Salvia miltiorrhiza Bunge (Danshen, DS), a famous Chinese medicine and functional tea, is extensively used to protect learning and memory capacities, although the mechanism of action remains unknown. PURPOSE The purpose of this research was to explore the efficacy and the underlying mechanism of DS in cognitive dysfunction caused by CSD. METHODS DS chemical composition was analyzed by UPLC-QTOF-MS/MS. Forty rats were randomly assigned to five groups (n = 8): control (CON), model (MOD), low- (1.35 g/kg, DSL), high-dose (2.70 g/kg, DSH) DS group, and Melatonin(100 mg/kg, MT) group. A CSD rat model was established over 21 days. DS's effects and the underlying mechanism were explored using the open-field test(OFT), Morris water-maze(MWM), tissue staining(Hematoxylin and Eosin Staining, Nissl staining, Alcian blue-periodic acid SCHIFF staining, and Immunofluorescence), enzyme-linked immunosorbent assay, Western blot, quantitative real-time polymerase chain reaction(qPCR), and 16S rRNA sequencing. RESULTS We demonstrated that CSD caused gut dysbiosis and cognitive dysfunction. Furthermore, 16S rRNA sequencing demonstrated that Firmicutes and Proteobacteria were more in fecal samples from model group rats, whereas Bacteroidota and Spirochaetota were less. DS therapy, on the contrary hand, greatly restored the gut microbial community, consequently alleviating cognitive impairment in rats. Further research revealed that DS administration reduced systemic inflammation via lowering intestinal inflammation and barrier disruption. Following that, DS therapy reduced Blood Brain Barrier(BBB) and neuronal damage, further decreasing neuroinflammation in the hippocampus(HP). Mechanistic studies revealed that DS therapy lowered lipopolysaccharide (LPS) levels in the HP, serum, and colon, consequently blocking the TLR4/MyD88/NF-κB signaling pathway and its downstream pro-inflammatory products(IL-1β, IL-6, TNF-α, iNOS, and COX2) in the HP and colon. CONCLUSION DS treatment dramatically improved spatial learning and memory impairments in rats with CSD by regulating the composition of the intestinal flora, preserving gut and brain barrier function, and reducing inflammation mediated by the LPS-TLR4 signaling pathway. Our findings provide novel insight into the mechanisms by which DS treats cognitive dysfunction caused by CSD.
Collapse
Affiliation(s)
- Chao Yin
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Meiya Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Shuna Jin
- Hubei Shizhen Laboratory, Wuhan 430065, PR China; School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuan Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Ding
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Qing Lv
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Zixuan Huang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Jiaqi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Jianmei Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Ping Wang
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China.
| | - Shunbo Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Qiuyun You
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China.
| |
Collapse
|
3
|
Kim DY, Kim SM, Han IO. Chronic rapid eye movement sleep deprivation aggravates the pathogenesis of Alzheimer's disease by decreasing brain O-GlcNAc cycling in mice. J Neuroinflammation 2024; 21:180. [PMID: 39044290 PMCID: PMC11264383 DOI: 10.1186/s12974-024-03179-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
This study investigated the role of O-GlcNAc cycling in Alzheimer's disease-related changes in brain pathophysiology induced by chronic REM sleep deprivation (CSD) in mice. CSD increased amyloid beta (Aβ) and p-Tau accumulation and impaired learning and memory (L/M) function. CSD decreased dendritic length and spine density. CSD also increased the intensity of postsynaptic density protein-95 (PSD-95) staining. All of these Alzheimer's disease (AD) pathogenic changes were effectively reversed through glucosamine (GlcN) treatment by enhancing O-GlcNAcylation. Interestingly, the lelvel of O-GlcNAcylated-Tau (O-Tau) exhibited an opposite trend compared to p-Tau, as it was elevated by CSD and suppressed by GlcN treatment. CSD increased neuroinflammation, as indicated by elevated levels of glial fibrillary acidic protein and IBA-1-positive glial cells in the brain, which were suppressed by GlcN treatment. CSD promoted the phosphorylation of GSK3β and led to an upregulation in the expression of endoplasmic reticulum (ER) stress regulatory proteins and genes. These alterations were effectively suppressed by GlcN treatment. Minocycline not only suppressed neuroinflammation induced by CSD, but it also rescued the decrease in O-GlcNAc levels caused by CSD. Minocycline also reduced AD neuropathy without affecting CSD-induced ER stress. Notably, overexpressing O-GlcNAc transferase in the dentate gyrus region of the mouse brain rescued CSD-induced cognitive dysfunction, neuropathy, neuroinflammation, and ER stress responses. Collectively, our findings reveal that dysregulation of O-GlcNAc cycling underlies CSD-induced AD pathology and demonstrate that restoration of OGlcNAcylation protects against CSD-induced neurodegeneration.
Collapse
Affiliation(s)
- Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea
| | - Sang-Min Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea.
| |
Collapse
|
4
|
Moon C, Schneider A, Cho YE, Zhang M, Dang H, Vu K. Sleep duration, sleep efficiency, and amyloid β among cognitively healthy later-life adults: a systematic review and meta-analysis. BMC Geriatr 2024; 24:408. [PMID: 38714912 PMCID: PMC11076214 DOI: 10.1186/s12877-024-05010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Abnormal amyloid β (Aβ) deposits in the brain are a hallmark of Alzheimer's disease (AD). Insufficient sleep duration and poor sleep quality are risk factors for developing AD. Sleep may play a role in Aβ regulation, but the magnitude of the relationship between sleep and Aβ deposition remains unclear. This systematic review examines the relationship between sleep (i.e., duration and efficiency) with Aβ deposition in later-life adults. METHODS A search of PubMed, CINAHL, Embase, and PsycINFO generated 5,005 published articles. Fifteen studies met the inclusion criteria for qualitative syntheses; thirteen studies for quantitative syntheses related to sleep duration and Aβ; and nine studies for quantitative syntheses related to sleep efficiency and Aβ. RESULTS Mean ages of the samples ranged from 63 to 76 years. Studies measured Aβ using cerebrospinal fluid, serum, and positron emission tomography scans with two tracers: Carbone 11-labeled Pittsburgh compound B or fluorine 18-labeled. Sleep duration was measured subjectively using interviews or questionnaires, or objectively using polysomnography or actigraphy. Study analyses accounted for demographic and lifestyle factors. Based on 13 eligible articles, our synthesis demonstrated that the average association between sleep duration and Aβ was not statistically significant (Fisher's Z = -0.055, 95% CI = -0.117 ~ 0.008). We found that longer self-report sleep duration is associated with lower Aβ (Fisher's Z = -0.062, 95% CI = -0.119 ~ -0.005), whereas the objectively measured sleep duration was not associated with Aβ (Fisher's Z = 0.002, 95% CI = -0.108 ~ 0.113). Based on 9 eligible articles for sleep efficiency, our synthesis also demonstrated that the average association between sleep efficiency and Aβ was not statistically significant (Fisher's Z = 0.048, 95% CI = -0.066 ~ 0.161). CONCLUSION The findings from this review suggest that shorter self-reported sleep duration is associated with higher Aβ levels. Given the heterogeneous nature of the sleep measures and outcomes, it is still difficult to determine the exact relationship between sleep and Aβ. Future studies with larger sample sizes should focus on comprehensive sleep characteristics and use longitudinal designs to better understand the relationship between sleep and AD.
Collapse
Affiliation(s)
- Chooza Moon
- University of Iowa College of Nursing, 50 Newton Rd, Iowa City, IA, 52242, USA.
| | - Aaron Schneider
- University of Iowa College of Liberal Arts and Sciences Department of Health and Human Physiology, 225 S. Grand Ave., Iowa City, IA, 52240, USA
| | - Young-Eun Cho
- University of Iowa College of Nursing, 50 Newton Rd, Iowa City, IA, 52242, USA
| | - Meina Zhang
- University of Iowa College of Nursing, 50 Newton Rd, Iowa City, IA, 52242, USA
| | - Hellen Dang
- University of Iowa College of Liberal Arts and Sciences Department of Health and Human Physiology, 225 S. Grand Ave., Iowa City, IA, 52240, USA
| | - Kelly Vu
- University of Iowa College of Pharmacy, 180 S. Grand Avenue, Iowa City, IA, 52242, USA
| |
Collapse
|
5
|
Torraville SE, Flynn CM, Kendall TL, Yuan Q. Life Experience Matters: Enrichment and Stress Can Influence the Likelihood of Developing Alzheimer's Disease via Gut Microbiome. Biomedicines 2023; 11:1884. [PMID: 37509523 PMCID: PMC10377385 DOI: 10.3390/biomedicines11071884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease, characterized by the presence of β-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs) formed from abnormally phosphorylated tau proteins (ptau). To date, there is no cure for AD. Earlier therapeutic efforts have focused on the clinical stages of AD. Despite paramount efforts and costs, pharmaceutical interventions including antibody therapies targeting Aβ have largely failed. This highlights the need to alternate treatment strategies and a shift of focus to early pre-clinical stages. Approximately 25-40% of AD cases can be attributed to environmental factors including chronic stress. Gut dysbiosis has been associated with stress and the pathogenesis of AD and can increase both Aβ and NFTs in animal models of the disease. Both stress and enrichment have been shown to alter AD progression and gut health. Targeting stress-induced gut dysbiosis through probiotic supplementation could provide a promising intervention to delay disease progression. In this review, we discuss the effects of stress, enrichment, and gut dysbiosis in AD models and the promising evidence from probiotic intervention studies.
Collapse
Affiliation(s)
- Sarah E Torraville
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Cassandra M Flynn
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Tori L Kendall
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Qi Yuan
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
6
|
Shixing X, Xueyan H, Yuan R, Wei T, Wei W. Enriched environment can reverse chronic sleep deprivation-induced damage to cellular plasticity in the dentate gyrus of the hippocampus. Transl Neurosci 2023; 14:20220280. [PMID: 36969794 PMCID: PMC10031502 DOI: 10.1515/tnsci-2022-0280] [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: 01/15/2023] [Revised: 02/15/2023] [Accepted: 02/24/2023] [Indexed: 03/24/2023] Open
Abstract
Objective We studied whether enriched environment (EE), a classic epigenetics paradigm, can prevent cellular plasticity damage caused by chronic sleep deprivation (SD). Methods We performed SD in mice by a modified multi-platform method (MMPM). Mice in the SD group were deprived of sleep for 18 h a day. In addition, half of the mice in the chronic SD group were exposed to EE stimuli for 6 h per day. Immunostaining analyzed neurogenesis and neural progenitor cell-differentiated phenotypes in the hippocampal dentate gyrus (DG) region. Result At 13 weeks, compared with the control group, SD severely impaired the proliferation and differentiation of neural stem cells, and EE completely reversed the process. SD can induce gliosis in the mouse hippocampus, and EE can delay the process. Conclusion: Our results suggest that chronic SD may damage the neurogenesis in the DG of the hippocampus. However, enrichment stimulation can reverse the processing by promoting neuronal repair related to neuronal plasticity.
Collapse
Affiliation(s)
- Xue Shixing
- Department of Neurology, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Hou Xueyan
- Department of Medical Imaging, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Ren Yuan
- Department of Neurology, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Tang Wei
- Department of Neurology, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Wang Wei
- Department of Rehabilitation Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning Province, China
| |
Collapse
|
7
|
Wang M, Zhao H, Zhang Z, Zhao Z, Wu H. Down-regulating insulin-like growth factor-1 receptor reduces amyloid-β deposition in mice cortex induced by chronic sleep restriction. Neurosci Lett 2023; 808:137189. [PMID: 36921668 DOI: 10.1016/j.neulet.2023.137189] [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: 08/09/2022] [Revised: 02/27/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023]
Abstract
OBJECTIVE Insufficient sleep affects cognitive function, but the underlying mechanism and potential protective ways are yet to be fully understood. This study aimed to explore the influence of chronic sleep restriction (CSR) on the insulin-like growth factor-1 (IGF-1) signaling pathway, and whether down-regulating IGF-1 signaling pathway would modulate amyloid-β (Aβ) peptides metabolism and its cortical deposition after CSR. Methods 8-week IGF-1R+/- mice and wild-type (WT) C57BL/6 (C57) mice were divided into four groups: IGF-1R+/- CSR (MUSR), IGF-1R+/- control (MUCO), C57 CSR (C57SR) and C57 control (C57CO). CSR model was established by application of slowly rotating drum for 2 months. Body weight and Lee's index were measured. The level of IGF-1 in plasma was measured by enzyme linked immunosorbent assay (ELISA). Aβ accumulation was detected by immunofluorescence. The expressions of amyloid precursor protein (APP), β-site amyloid precursor protein-cleaving enzyme-1 (BACE-1) and C99 were detected using western-blot (WB). Results Two-way ANOVA showed genotypic effect was significant on body weight and Lee's index. Neither treatment effect nor interaction reached significant difference on body weight and Lee's index. The level of IGF-1 in plasma was significantly decreased in C57SR compared with C57CO. Besides, compared with C57CO, Aβ was markedly accumulated in frontal cortex, in parallel with increased expressions of BACE-1 and C99, and with no difference of APP in C57SR group. Further, no significant changes of Aβ, BACE-1, C99 and APP were detected in MUSR compared with MUCO. Conclusions This study showed that CSR could induce the decrease of circulating IGF-1 in mice. By using the IGF-1R+/- mice, we found that down-regulating IGF-1R could reduce Aβ deposition in mice frontal cortex after CSR via inhibiting BACE-1 protein expression and activity, which were independent of the changes of body weight and Lee's index. These findings indicate that the blockage of IGF-1 signaling pathway might be a protection mechanism for alleviating the impact of CSR.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China; Department of hyperbaric oxygen and Neurology, Naval Medical Center, Shanghai 200052, China
| | - Hongyi Zhao
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China; Department of Neurology, NO 984 Hospital of PLA, Beijing 100094, China
| | - Zhaohuai Zhang
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Zhongxin Zhao
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Huijuan Wu
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
8
|
Disturbance of REM sleep exacerbates microglial activation in APP/PS1 mice. Neurobiol Learn Mem 2023; 200:107737. [PMID: 36813079 DOI: 10.1016/j.nlm.2023.107737] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 01/17/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
Although both nonrapid eye movement (NREM) sleep loss and rapid eye movement (REM) sleep loss exacerbate Alzheimer's disease (AD) progression, they exert different effects. Microglial activation can be beneficial or detrimental to AD patients under different conditions. However, few studies have investigated which sleep stage is the main regulator of microglial activation or the downstream effects of this activation. We aimed to explore the roles of different sleep phases in microglial activation and to investigate the possible effect of microglial activation on AD pathology. In this study, thirty-six 6-month-old APP/PS1 mice were equally divided into 3 groups: the stress control (SC), total sleep deprivation (TSD), and REM deprivation (RD) groups. All mice underwent a 48-hour intervention before their spatial memory was assessed using a Morris water maze (MWM). Then, microglial morphology, activation- and synapse-related protein expression, and inflammatory cytokine and amyloid β (Aβ) levels in hippocampal tissues were measured. We found that the RD and TSD groups exhibited worse spatial memory in the MWM tests. In addition, the RD and TSD groups showed greater microglial activation, higher inflammatory cytokine levels, lower synapse-related protein expression and more severe Aβ accumulation than the SC group, but there were no significant differences between the RD and TSD groups. This study demonstrates that disturbance of REM sleep may activate microglia in APP/PS1 mice. These activated microglia may promote neuroinflammation and engulf synapses but show a weakened ability to clear plaques.
Collapse
|
9
|
Vierra J, Boonla O, Prasertsri P. Effects of sleep deprivation and 4-7-8 breathing control on heart rate variability, blood pressure, blood glucose, and endothelial function in healthy young adults. Physiol Rep 2022; 10:e15389. [PMID: 35822447 PMCID: PMC9277512 DOI: 10.14814/phy2.15389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/17/2022] [Accepted: 06/25/2022] [Indexed: 11/24/2022] Open
Abstract
This study investigated the effects of sleep deprivation on heart rate variability (HRV), blood pressure (BP), fasting blood glucose (FBG), and endothelial function as well as the immediate effects of 4-7-8 breathing control on HRV and BP. In total, 43 healthy participants aged 19-25 years were classified into two groups: Twenty two in the with sleep deprivation group and 21 in the without sleep deprivation (control) group. Resting heart rate (HR), BP, HRV, FBG, and endothelial function were examined. Subsequently, participants practiced 4-7-8 breathing control for six cycles/set for three sets interspersed between each set by 1-min normal breathing. Thereafter, the HR, BP, and HRV were immediately examined. The HRV, HR, and BP variables and FBG were not significantly different between the two groups. However, endothelial function was significantly lower in the sleep deprivation group than that in the control group (p < 0.05). In response to 4-7-8 breathing control, low- and very-low-frequency powers significantly decreased (p < 0.05), whereas high-frequency power significantly increased (p < 0.05) in the control group. Moreover, time domain, total power, and very-low-frequency power significantly decreased (p < 0.05) in the sleep deprivation group. Both groups had significantly decreased HR and systolic BP (p < 0.05). HRV, HR, and BP variables showed no significant differences between the groups. Healthy young adults with and without sleep deprivation may have similar HRV, BP, and FBG values. However, sleep deprivation may cause decreased endothelial function. Furthermore, 4-7-8 breathing control can help participants improve their HRV and BP, particularly in those without sleep deprivation.
Collapse
Affiliation(s)
- Jaruwan Vierra
- Faculty of Allied Health SciencesBurapha UniversityChonburiThailand
| | - Orachorn Boonla
- Faculty of Allied Health SciencesBurapha UniversityChonburiThailand
- Exercise and Nutrition Innovation and Sciences Research UnitBurapha UniversityChonburiThailand
| | - Piyapong Prasertsri
- Faculty of Allied Health SciencesBurapha UniversityChonburiThailand
- Exercise and Nutrition Innovation and Sciences Research UnitBurapha UniversityChonburiThailand
| |
Collapse
|
10
|
Roflumilast, a Phosphodiesterase-4 Inhibitor, Ameliorates Sleep Deprivation-Induced Cognitive Dysfunction in C57BL/6J Mice. ACS Chem Neurosci 2022; 13:1938-1947. [PMID: 35736514 DOI: 10.1021/acschemneuro.2c00127] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Sleep deprivation (SD) interferes with long-term memory and cognitive functions by overactivation of phosphodiesterase (PDEs) enzymes. PDE4, a nonredundant regulator of the cyclic nucleotides (cAMP), is densely expressed in the hippocampus and is involved in learning and memory processes. In the present study, we investigated the effects of Roflumilast (ROF), a PDE4B inhibitor, on sleep deprivation-induced cognitive dysfunction in a mouse model. Memory assessment was performed using a novel object recognition task, and the hippocampal cAMP level was estimated by the ELISA method. The alterations in the expressions of PDE4B, amyloid-β (Aβ), CREB, BDNF, and synaptic proteins (Synapsin I, SAP 97, PSD 95) were assessed to gain insights into the possible mechanisms of action of ROF using the Western blot technique. Results show that ROF reversed SD-induced cognitive decline in mice. ROF downregulated PDE4B and Aβ expressions in the brain. Additionally, ROF improved the cAMP level and the protein expressions of synapsin I, SAP 97, and PSD 95 in the hippocampal region of SD mice. Taken together, these results suggest that ROF can suppress the deleterious effects of SD-induced cognitive dysfunction via the PDE4B-mediated cAMP/CREB/BDNF signaling cascade.
Collapse
|
11
|
García-Aviles JE, Méndez-Hernández R, Guzmán-Ruiz MA, Cruz M, Guerrero-Vargas NN, Velázquez-Moctezuma J, Hurtado-Alvarado G. Metabolic Disturbances Induced by Sleep Restriction as Potential Triggers for Alzheimer's Disease. Front Integr Neurosci 2021; 15:722523. [PMID: 34539357 PMCID: PMC8447653 DOI: 10.3389/fnint.2021.722523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/26/2021] [Indexed: 01/15/2023] Open
Abstract
Sleep has a major role in learning, memory consolidation, and metabolic function. Although it is known that sleep restriction increases the accumulation of amyloid β peptide (Aβ) and the risk to develop Alzheimer's disease (AD), the mechanism behind these effects remains unknown. In this review, we discuss how chronic sleep restriction induces metabolic and cognitive impairments that could result in the development of AD in late life. Here, we integrate evidence regarding mechanisms whereby metabolic signaling becomes disturbed after short or chronic sleep restriction in the context of cognitive impairment, particularly in the accumulation of Aβ in the brain. We also discuss the role of the blood-brain barrier in sleep restriction with an emphasis on the transport of metabolic signals into the brain and Aβ clearance. This review presents the unexplored possibility that the alteration of peripheral metabolic signals induced by sleep restriction, especially insulin resistance, is responsible for cognitive deficit and, subsequently, implicated in AD development.
Collapse
Affiliation(s)
- Jesús Enrique García-Aviles
- Area of Neurosciences, Biology of Reproduction Department, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Rebeca Méndez-Hernández
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Mara A Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel Cruz
- Instituto Mexicano del Seguro Social, Centro Médico Nacional Siglo XXI, Hospital de Especialidades, Unidad de Investigación Médica en Bioquímica, Mexico City, Mexico
| | - Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Javier Velázquez-Moctezuma
- Area of Neurosciences, Biology of Reproduction Department, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Gabriela Hurtado-Alvarado
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|