1
|
Wang W, Wang Z, Cao J, Dong Y, Chen Y. Melatonin ameliorates chronic sleep deprivation against memory encoding vulnerability: Involvement of synapse regulation via the mitochondrial-dependent redox homeostasis-induced autophagy inhibition. Free Radic Biol Med 2024; 225:398-414. [PMID: 39396581 DOI: 10.1016/j.freeradbiomed.2024.10.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/13/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Voluntary sleep curtailment is increasingly more rampant in modern society and compromises healthy cognition, including memory, to varying degrees. However, whether memory encoding is impaired after chronic sleep deprivation (CSD) and the underlying molecular mechanisms involved remain unclear. Here, using the mice, we tested the impact of CSD on the encoding abilities of social recognition-dependent memory and object recognition-dependent memory. We found that memory encoding was indeed vulnerable to CSD, while memory retrieval remained unaffected. The hippocampal neurons of mice with memory encoding deficits exhibited significant synapse damage and hyperactive autophagy, which dissipates during regular sleep cycles. This excessive autophagy appeared to be triggered by damage to mitochondrial DNA (mtDNA), resulting from oxidative stress within the mitochondria. The relief at the behavioral and molecular biological levels can be achieved with intraperitoneal injections of the antioxidant compound melatonin. Moreover, our in vitro experiments using HT-22 cells demonstrated that oxidative stress induced by hydrogen peroxide led to oxidative damage, including mtDNA damage, and activation of autophagy. Melatonin treatment effectively countered these effects, restoring redox homeostasis and reducing excessive autophagic activity. Notably, this protective effect was not observed when melatonin was administered as a pre-treatment. Together, our findings reveal the vulnerability of memory encoding during chronic sleep curtailment, which is caused by oxidative stress and consequent enhancement of autophagy, suggest a potential therapeutic strategy for addressing these effects following prolonged wakefulness through melatonin intervention, and reiterate the significance of adequate sleep for memory formation and retention.
Collapse
Affiliation(s)
- Wei Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Zixu Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Jing Cao
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Yulan Dong
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Yaoxing Chen
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
2
|
Zhang Y, Zhang C, Dai Q, Ma R. Continuous Theta Burst Stimulation Inhibits Oxidative Stress-Induced Inflammation and Autophagy in Hippocampal Neurons by Activating Glutathione Synthesis Pathway, Improving Cognitive Impairment in Sleep-Deprived Mice. Neuromolecular Med 2024; 26:40. [PMID: 39388015 DOI: 10.1007/s12017-024-08807-z] [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: 05/08/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
Sleep deprivation (SD) has been reported to have a negative impact on cognitive function. Continuous theta burst stimulation (cTBS) shows certain effects in improving sleep and neurological diseases, and its molecular or cellular role in SD-induced cognition impairment still need further exploration. In this study, C57BL/6 mice were subjected to 48 h of SD and cTBS treatment, and cTBS treatment significantly improved SD-triggered impairment of spatial learning and memory abilities in mice. Additionally, cTBS reduced malondialdehyde levels, increased superoxide dismutase activities, and inhibited the production of inflammatory cytokines, alleviating oxidative stress and inflammation levels in hippocampal tissues of SD model mice. cTBS decreased LC3II/LC3I ratio, Beclin1 protein levels, and LC3B puncta intensity, and elevated p62 protein levels to suppress excessive autophagy in hippocampal tissues of SD-stimulated mice. Then, we proved that inhibiting oxidative stress alleviated inflammation, autophagy, and death of hippocampal neuron cells through an in vitro cellular model for oxidative stress, and cTBS treatment promoted the production of glutathione (GSH), the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and the mRNA expression of GSH synthesis-related genes to enhance antioxidant capacity in hippocampal tissues of SD mice. An Nrf2 inhibitor ML385 or a GSH synthesis inhibitor BSO reversed the alleviating effects of cTBS treatment on oxidative stress-associated damage of hippocampal tissues and cognitive impairment in SD model mice. Altogether, our study demonstrated that cTBS mitigates oxidative stress-associated inflammation and autophagy through activating the Nrf2-mediated GSH synthesis pathway, improving cognitive impairment in SD mice.
Collapse
Affiliation(s)
- Yi Zhang
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China
| | - Cheng Zhang
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China
| | - Qing Dai
- Anesthesiology Department, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Rui Ma
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China.
| |
Collapse
|
3
|
Wei SM, Huang YM. Effect of sulforaphane on testicular ischemia-reperfusion injury induced by testicular torsion-detorsion in rats. Sci Rep 2024; 14:23420. [PMID: 39379457 PMCID: PMC11461801 DOI: 10.1038/s41598-024-74756-z] [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: 05/21/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
Testicular ischemia-reperfusion induces enhanced concentration of reactive oxygen species. The increased reactive oxygen species harm cellular lipids, nucleic acids, proteins, and carbohydrates, and ultimately cause testicular injury. Sulforaphane, a kind of natural dietary isothiocyanate, exists predominantly in some cruciferous vegetables, like broccoli and cabbage. It can protect tissues from oxidative stress-induced damage. Herein, we analyzed the effectiveness of sulforaphane in treating ischemia-reperfusion injury occurring after testicular torsion-detorsion. Male rats (n = 60) were grouped as follows: sham-operated group, unilateral testicular ischemia-reperfusion group, and unilateral testicular ischemia-reperfusion group receiving sulforaphane treatment at 5 mg/kg. No testicular torsion-detorsion was performed in the sham group. Unilateral testicular ischemia-reperfusion model was created by detorsion after 2 h of left testicular torsion. In the sulforaphane-treated group, intraperitoneal sulforaphane (5 mg/kg) was administered at left testicular detorsion. Biochemical assay, Western blot, and hematoxylin and eosin staining were used to evaluate testicular malondialdehyde content (an important marker of reactive oxygen species), protein levels of superoxide dismutase and catalase (intracellular antioxidant defense mechanism), and testicular reproductive function, respectively. In testicular tissues, malondialdehyde content was significantly promoted, while protein levels of superoxide dismutase and catalase, and testicular reproductive function were significantly reduced in ipsilateral testes by testicular ischemia-reperfusion. Nevertheless, sulforaphane administration partially reversed the effect of testicular ischemia-reperfusion on these indexes. It can be concluded that sulforaphane elevates protein levels of superoxide dismutase and catalase, and suppresses reactive oxygen species content, thereby preventing ischemia-reperfusion injury in testis.
Collapse
Affiliation(s)
- Si-Ming Wei
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou City, 310015, Zhejiang Province, China.
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou City, 310053, Zhejiang Province, China.
| | - Yu-Min Huang
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou City, 310058, Zhejiang Province, China
| |
Collapse
|
4
|
Zulkifli MF, Masimen MAA, Ridzuan PM, Wan Ismail WI. Exploring honey's potential as a functional food for natural sleep aid. Food Funct 2024; 15:9678-9689. [PMID: 39248637 DOI: 10.1039/d4fo02013h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Sleep disorders present significant challenges to public health, necessitating effective and sustainable solutions beyond conventional pharmacological interventions. This study aims to investigate the potential of honey and its compositions as a functional food for addressing sleep disorders. A comprehensive review of existing literature explores honey's intrinsic properties and impact on sleep quality. The findings suggest that honey, with its unique composition and soothing effects, offers a promising avenue for enhancing sleep patterns without relying on pharmaceutical drugs. This study also discusses the possible mechanism of action and the challenges of using honey in this field. While further research is needed to elucidate specific mechanisms and optimal utilisation, integrating honey into holistic sleep management strategies holds promise for improving overall well-being and quality of life.
Collapse
Affiliation(s)
- Muhammad Faiz Zulkifli
- Department of Research and Development, BioInnovSphere Labs, 21030 Kuala Nerus, Terengganu, Malaysia.
- Cell Signalling and Biotechnology Research Group (CeSBTech), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Mohammad Asyraf Adhwa Masimen
- Department of Research and Development, BioInnovSphere Labs, 21030 Kuala Nerus, Terengganu, Malaysia.
- Cell Signalling and Biotechnology Research Group (CeSBTech), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - P M Ridzuan
- Department of Research and Development, Dr. Ridz Research Centre, 21030, Kuala Nerus, Terengganu, Malaysia
| | - Wan Iryani Wan Ismail
- Department of Research and Development, BioInnovSphere Labs, 21030 Kuala Nerus, Terengganu, Malaysia.
- Biological Security and Sustainability Research Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| |
Collapse
|
5
|
Ye M, Jeong W, Yu HJ, Kim KR, Rhie SJ, Kim Y, Kim J, Shim I. Effect of Earthing Mats on Sleep Quality in Rats. Int J Mol Sci 2024; 25:9791. [PMID: 39337279 PMCID: PMC11432166 DOI: 10.3390/ijms25189791] [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: 08/15/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Grounding, a therapeutic technique involving direct contact with the earth, has been proposed by various studies to potentially have beneficial effects on pressure, sleep quality, stress, inflammation, and mood. However, the scientific evidence supporting its sedative effects remains incomplete. This study examined the sedative effectiveness of an earthing mat on sleep quality and investigated the underlying neural mechanisms using electroencephalography (EEG) analysis in rodents, focusing on orexin and superoxide dismutase (SOD) levels in the brain. Rats were randomly assigned to four groups: the naïve normal group (Nor), the group exposed to an earthing mat for 7 days (A-7D), the group exposed to an earthing mat for 21 days (A-21D), and the group exposed to an electronic blanket for 21 days (EM). EEG results revealed that the A-21D group exhibited significantly reduced wake time and increased rapid eye movement (REM), non-rapid eye movement (NREM), and total sleep time compared to the Nor group (p < 0.05). Moreover, the A-21D group demonstrated a significant increase in NREM sleep (p < 0.001), REM sleep (p < 0.01), and total sleep time (p < 0.001), along with a decrease in wake time compared to the EM group (p < 0.001). The orexin level in the A-21D group was significantly lower compared to the Nor group (p < 0.01), while SOD1 expression was markedly elevated in the A-21D group compared to the Nor group (p < 0.001). These results suggest that the earthing mat may represent a promising new method for promoting sleep quality and could serve as an effective therapeutic technique.
Collapse
Affiliation(s)
- Minsook Ye
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| | - Woojin Jeong
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| | - Hyo-jeong Yu
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| | - Kyu-ri Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| | - Sung Ja Rhie
- Department of Beauty Design, Halla University, Wonju 26404, Republic of Korea;
| | - Yongsuk Kim
- DF World Corporation, Royal Building, 19 Saemunan-ro 5-gil, Jongno-gu, Seoul 03173, Republic of Korea;
| | - Jiyoun Kim
- World Home Doctor Corporation, 73 Anyangcheonseo-ro, Manan-gu, Anyang-si 14087, Republic of Korea;
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| |
Collapse
|
6
|
Gonçalves LDS, Rusch G, Alves AG, Krüger LD, Paim MP, Martins CC, da Motta KP, Neto JSS, Luchese C, Wilhelm EA, Brüning CA, Bortolatto CF. Acute 2-phenyl-3-(phenylselanyl)benzofuran treatment reverses the neurobehavioral alterations induced by sleep deprivation in mice. Biochem Pharmacol 2024; 226:116339. [PMID: 38848781 DOI: 10.1016/j.bcp.2024.116339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 05/05/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Sleep is a fundamental state for maintaining the organism homeostasis. Disruptions in sleep patterns predispose to the appearance of memory impairments and mental disorders, including depression. Recent pre-clinical studies have highlighted the antidepressant-like properties of the synthetic compound 2-phenyl-3-(phenylselanyl)benzofuran (SeBZF1). To further investigate the neuromodulatory effects of SeBZF1, this study aimed to assess its therapeutic efficacy in ameliorating neurobehavioral impairments induced by sleep deprivation (SD) in mice. For this purpose, a method known as multiple platforms over water was used to induce rapid eye movement (REM) SD. Two hours after acute SD (24 h), male Swiss mice received a single treatment of SeBZF1 (5 mg/kg, intragastric route) or fluoxetine (a positive control, 20 mg/kg, intraperitoneal route). Subsequently, behavioral tests were conducted to assess spontaneous motor function (open-field test), depressive-like behavior (tail suspension test), and memory deficits (Y-maze test). Brain structures were utilized to evaluate oxidative stress markers, monoamine oxidase (MAO) and acetylcholinesterase (AChE) activities. Our findings revealed that SD animals displayed depressive-like behavior and memory impairments, which were reverted by SeBZF1 and fluoxetine treatments. SeBZF1 also reverted the increase in lipoperoxidation levels and glutathione peroxidase activity in the pre-frontal cortex in mice exposed to SD. Besides, the increase in hippocampal AChE activity induced by SD was overturned by SeBZF1. Lastly, cortical MAO-B activity was reestablished by SeBZF1 in mice that underwent SD. Based on the main findings of this study, it can be inferred that the compound SeBZF1 reverses the neurobehavioral alterations induced by sleep deprivation in male Swiss mice.
Collapse
Affiliation(s)
- Luciane da Silva Gonçalves
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | - Gabriela Rusch
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | - Amália Gonçalves Alves
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | - Letícia Devantier Krüger
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | - Mariana Parron Paim
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | - Carolina Cristóvão Martins
- Laboratório de Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | - Ketlyn Pereira da Motta
- Laboratório de Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | | | - Cristiane Luchese
- Laboratório de Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | - Ethel Antunes Wilhelm
- Laboratório de Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil
| | - César Augusto Brüning
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil.
| | - Cristiani Folharini Bortolatto
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Universidade Federal de Pelotas (UFPel), CEP 96010-900, Pelotas, RS, Brasil.
| |
Collapse
|
7
|
Davinelli S, Medoro A, Savino R, Scapagnini G. Sleep and Oxidative Stress: Current Perspectives on the Role of NRF2. Cell Mol Neurobiol 2024; 44:52. [PMID: 38916679 PMCID: PMC11199221 DOI: 10.1007/s10571-024-01487-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/15/2024] [Indexed: 06/26/2024]
Abstract
Sleep is a fundamental conserved physiological state across evolution, suggesting vital biological functions that are yet to be fully clarified. However, our understanding of the neural and molecular basis of sleep regulation has increased rapidly in recent years. Among various processes implicated in controlling sleep homeostasis, a bidirectional relationship between sleep and oxidative stress has recently emerged. One proposed function of sleep may be the mitigation of oxidative stress in both brain and peripheral tissues, contributing to the clearance of reactive species that accumulate during wakefulness. Conversely, reactive species, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS), at physiological levels, may act as signaling agents to regulate redox-sensitive transcriptional factors, enzymes, and other effectors involved in the regulation of sleep. As a primary sensor of intracellular oxidation, the transcription factor NRF2 is emerging as an indispensable component to maintain cellular redox homeostasis during sleep. Indeed, a number of studies have revealed an association between NRF2 dysfunction and the most common sleep conditions, including sleep loss, obstructive sleep apnea, and circadian sleep disturbances. This review examines the evidence of the intricate link between oxidative stress and NRF2 function in the context of sleep, and highlights the potential of NRF2 modulators to alleviate sleep disturbances.
Collapse
Affiliation(s)
- Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy.
| | - Alessandro Medoro
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy
| | - Rosa Savino
- Department of Woman and Child, Neuropsychiatry for Child and Adolescent Unit, General Hospital "Riuniti" of Foggia, Viale Pinto Luigi, 1, 71122, Foggia, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy
| |
Collapse
|
8
|
Chen C, Zhai R, Lan X, Yang S, Tang S, Xiong X, He Y, Lin J, Feng J, Chen D, Shi J. The influence of sleep disorders on perioperative neurocognitive disorders among the elderly: A narrative review. IBRAIN 2024; 10:197-216. [PMID: 38915944 PMCID: PMC11193868 DOI: 10.1002/ibra.12167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/26/2024]
Abstract
This review comprehensively assesses the epidemiology, interaction, and impact on patient outcomes of perioperative sleep disorders (SD) and perioperative neurocognitive disorders (PND) in the elderly. The incidence of SD and PND during the perioperative period in older adults is alarmingly high, with SD significantly contributing to the occurrence of postoperative delirium. However, the clinical evidence linking SD to PND remains insufficient, despite substantial preclinical data. Therefore, this study focuses on the underlying mechanisms between SD and PND, underscoring that potential mechanisms driving SD-induced PND include uncontrolled central nervous inflammation, blood-brain barrier disruption, circadian rhythm disturbances, glial cell dysfunction, neuronal and synaptic abnormalities, impaired central metabolic waste clearance, gut microbiome dysbiosis, hippocampal oxidative stress, and altered brain network connectivity. Additionally, the review also evaluates the effectiveness of various sleep interventions, both pharmacological and nonpharmacological, in mitigating PND. Strategies such as earplugs, eye masks, restoring circadian rhythms, physical exercise, noninvasive brain stimulation, dexmedetomidine, and melatonin receptor agonists have shown efficacy in reducing PND incidence. The impact of other sleep-improvement drugs (e.g., orexin receptor antagonists) and methods (e.g., cognitive-behavioral therapy for insomnia) on PND is still unclear. However, certain drugs used for treating SD (e.g., antidepressants and first-generation antihistamines) may potentially aggravate PND. By providing valuable insights and references, this review aimed to enhance the understanding and management of PND in older adults based on SD.
Collapse
Affiliation(s)
- Chao Chen
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Rui‐Xue Zhai
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xin Lan
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Sheng‐Feng Yang
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Si‐Jie Tang
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xing‐Long Xiong
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Yu‐Xin He
- Department of Gastroenterology and HepatologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jing‐Fang Lin
- Department of Anesthesiology, Fujian Provincial HospitalSheng Li Clinical Medical College of Fujian Medical UniversityFuzhouChina
| | - Jia‐Rong Feng
- Khoury College of Computer SciencesNortheastern UniversityBostonAmerica
| | - Dong‐Xu Chen
- Department of Anesthesiology, West China Second HospitalSichuan UniversityChengduChina
| | - Jing Shi
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
9
|
Erdos T, Masuda M, Venketaraman V. Glutathione in HIV-Associated Neurocognitive Disorders. Curr Issues Mol Biol 2024; 46:5530-5549. [PMID: 38921002 PMCID: PMC11202908 DOI: 10.3390/cimb46060330] [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: 03/21/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
A large portion of patients with Human Immunodeficiency Virus (HIV) have neurologic sequelae. Those with better-controlled HIV via antiretroviral therapies generally have less severe neurologic symptoms. However, for many patients, antiretrovirals do not adequately resolve symptoms. Since much of the pathogenesis of HIV/AIDS (Autoimmune Deficiency Syndrome) involves oxidative stress either directly, through viral interaction, or indirectly, through inflammatory mechanisms, we have reviewed relevant trials of glutathione supplementation in each of the HIV-associated neurocognitive diseases and have found disease-specific results. For diseases for which trials have not been completed, predicted responses to glutathione supplementation are made based on relevant mechanisms seen in the literature. It is not sufficient to conclude that all HIV-associated neurocognitive disorders (HAND) will benefit from the antioxidant effects of glutathione supplementation. The potential effects of glutathione supplementation in patients with HAND are likely to differ based on the specific HIV-associated neurocognitive disease.
Collapse
Affiliation(s)
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (T.E.); (M.M.)
| |
Collapse
|
10
|
Wang L, Tian H, Wang H, Mao X, Luo J, He Q, Wen P, Cao H, Fang L, Zhou Y, Yang J, Jiang L. Disrupting circadian control of autophagy induces podocyte injury and proteinuria. Kidney Int 2024; 105:1020-1034. [PMID: 38387504 DOI: 10.1016/j.kint.2024.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 02/24/2024]
Abstract
The circadian clock influences a wide range of biological process and controls numerous aspects of physiology to adapt to the daily environmental changes caused by Earth's rotation. The kidney clock plays an important role in maintaining tubular function, but its effect on podocytes remains unclear. Here, we found that podocytes expressed CLOCK proteins, and that 2666 glomerular gene transcripts (13.4%), including autophagy related genes, had 24-hour circadian rhythms. Deletion of Clock in podocytes resulted in 1666 gene transcripts with the loss of circadian rhythm including autophagy genes. Podocyte-specific Clock knockout mice at age three and eight months showed deficient autophagy, loss of podocytes and increased albuminuria. Chromatin immunoprecipitation (ChIP) sequence analysis indicated autophagy related genes were targets of CLOCK in podocytes. ChIP-PCR further confirmed Clock binding to the promoter regions of Becn1 and Atg12, two autophagy related genes. Furthermore, the association of CLOCK regulated autophagy with chronic sleep fragmentation and diabetic kidney disease was analyzed. Chronic sleep fragmentation resulted in the loss of glomerular Clock rhythm, inhibition of podocyte autophagy, and proteinuria. Rhythmic oscillations of Clock also disappeared in high glucose treated podocytes and in glomeruli from diabetic mice. Finally, circadian differences in podocyte autophagy were also abolished in diabetic mice. Deletion Clock in podocytes aggravated podocyte injury and proteinuria in diabetic mice. Thus, our findings demonstrate that clock-dependent regulation of autophagy may be essential for podocyte survival. Hence. loss of circadian controlled autophagy may play an important role in podocyte injury and proteinuria.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Han Tian
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haiyan Wang
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoming Mao
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Luo
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qingyun He
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Wen
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongdi Cao
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Fang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Yang Zhou
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Junwei Yang
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Lei Jiang
- Department of Nephrology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
Bonmatí-Carrión MÁ, Rol MA. Melatonin as a Mediator of the Gut Microbiota-Host Interaction: Implications for Health and Disease. Antioxidants (Basel) 2023; 13:34. [PMID: 38247459 PMCID: PMC10812647 DOI: 10.3390/antiox13010034] [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: 11/30/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
In recent years, the role played by melatonin on the gut microbiota has gained increasingly greater attention. Additionally, the gut microbiota has been proposed as an alternative source of melatonin, suggesting that this antioxidant indoleamine could act as a sort of messenger between the gut microbiota and the host. This review analyses the available scientific literature about possible mechanisms involved in this mediating role, highlighting its antioxidant effects and influence on this interaction. In addition, we also review the available knowledge on the effects of melatonin on gut microbiota composition, as well as its ability to alleviate dysbiosis related to sleep deprivation or chronodisruptive conditions. The melatonin-gut microbiota relationship has also been discussed in terms of its role in the development of different disorders, from inflammatory or metabolic disorders to psychiatric and neurological conditions, also considering oxidative stress and the reactive oxygen species-scavenging properties of melatonin as the main factors mediating this relationship.
Collapse
Affiliation(s)
- María-Ángeles Bonmatí-Carrión
- Chronobiology Laboratory, Department of Physiology, College of Biology, Mare Nostrum Campus, University of Murcia, Instituto Universitario de Investigación en Envejecimiento, Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30100 Murcia, Spain;
- Ciber Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria-Angeles Rol
- Chronobiology Laboratory, Department of Physiology, College of Biology, Mare Nostrum Campus, University of Murcia, Instituto Universitario de Investigación en Envejecimiento, Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30100 Murcia, Spain;
- Ciber Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|