1
|
Huang L, Liu M, Li Z, Li B, Wang J, Zhang K. Systematic review of amyloid-beta clearance proteins from the brain to the periphery: implications for Alzheimer's disease diagnosis and therapeutic targets. Neural Regen Res 2025; 20:3574-3590. [PMID: 39820231 PMCID: PMC11974662 DOI: 10.4103/nrr.nrr-d-24-00865] [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: 08/21/2024] [Revised: 09/19/2024] [Accepted: 12/02/2024] [Indexed: 01/19/2025] Open
Abstract
Amyloid-beta clearance plays a key role in the pathogenesis of Alzheimer's disease. However, the variation in functional proteins involved in amyloid-beta clearance and their correlation with amyloid-beta levels remain unclear. In this study, we conducted meta-analyses and a systematic review using studies from the PubMed, Embase, Web of Science, and Cochrane Library databases, including journal articles published from inception to June 30, 2023. The inclusion criteria included studies comparing the levels of functional proteins associated with amyloid-beta clearance in the blood, cerebrospinal fluid, and brain of healthy controls, patients with mild cognitive impairment, and patients with Alzheimer's disease. Additionally, we analyzed the correlation between these functional proteins and amyloid-beta levels in patients with Alzheimer's disease. The methodological quality of the studies was assessed via the Newcastle‒Ottawa Scale. Owing to heterogeneity, we utilized either a fixed-effect or random-effect model to assess the 95% confidence interval (CI) of the standard mean difference (SMD) among healthy controls, patients with mild cognitive impairment, and patients with Alzheimer's disease. The findings revealed significant alterations in the levels of insulin-degrading enzymes, neprilysin, matrix metalloproteinase-9, cathepsin D, receptor for advanced glycation end products, and P-glycoprotein in the brains of patients with Alzheimer's disease, patients with mild cognitive impairment, and healthy controls. In cerebrospinal fluid, the levels of triggering receptor expressed on myeloid cells 2 and ubiquitin C-terminal hydrolase L1 are altered, whereas the levels of TREM2, CD40, CD40L, CD14, CD22, cathepsin D, cystatin C, and α2 M in peripheral blood differ. Notably, TREM2 and cathepsin D showed changes in both brain (SMD = 0.31, 95% CI: 0.16-0.47, P < 0.001, I2 = 78.4%; SMD = 1.24, 95% CI: 0.01-2.48, P = 0.048, I2 = 90.1%) and peripheral blood (SMD = 1.01, 95% CI: 0.35-1.66, P = 0.003, I2 = 96.5%; SMD = 7.55, 95% CI: 3.92-11.18, P < 0.001, I2 = 98.2%) samples. Furthermore, correlations were observed between amyloid-beta levels and the levels of TREM2 ( r = 0.16, 95% CI: 0.04-0.28, P = 0.009, I2 = 74.7%), neprilysin ( r = -0.47, 95% CI: -0.80-0.14, P = 0.005, I2 = 76.1%), and P-glycoprotein ( r = -0.31, 95% CI: -0.51-0.11, P = 0.002, I2 = 0.0%) in patients with Alzheimer's disease. These findings suggest that triggering receptor expressed on myeloid cells 2 and cathepsin D could serve as potential diagnostic biomarkers for Alzheimer's disease, whereas triggering receptor expressed on myeloid cells 2, neprilysin, and P-glycoprotein may represent potential therapeutic targets.
Collapse
Affiliation(s)
- Letian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Mingyue Liu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Ze Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Bing Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Jiahe Wang
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ke Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
2
|
Wang S, Zhou S, Jiang X, Yang D, He J, Xiu M. Acute hypoxia induces sleep disorders via sima/HIF-1α regulation of circadian rhythms in adult Drosophila. Comp Biochem Physiol C Toxicol Pharmacol 2025; 294:110192. [PMID: 40086680 DOI: 10.1016/j.cbpc.2025.110192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/03/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
The atmospheric oxygen concentration is significantly reduced in highland regions compared to lowland areas. The first entering the plateau can induce sleep disorders in individuals, primarily attributed to insufficient oxygen supply. This study used Drosophila melanogaster as a model organism to better understand the molecular mechanism of acute hypoxia-induced sleep disorders. The Drosophila activity monitoring system (DAMS) was employed to observe the sleep-wake in adult (w1118, simaKG07607, and clockjrk) female flies. Quantifying the relative mRNA expression levels of sima and circadian clock genes in the head of flies was accomplished by utilizing qRT-PCR. Acute hypoxia caused sleep disorders in w1118 flies, such as shortened sleep duration and length, and prolonged sleep latency. PCR results showed that sima and clock genes were up-regulated in ZT6 and ZT12 and down-regulated in ZT0 and ZT18 in acute hypoxic w1118 flies compared to normoxic w1118 flies. Under normoxic conditions, sleep indexes in simaKG07607 flies were not substantially different from w1118 flies. However, clockjrk flies demonstrated a reduced sleep duration, decreased sleep bout length, and increased sleep latency and activities. Sleep and gene expression in simaKG07607 flies under acute hypoxic conditions were not significantly different from those under normoxic conditions. Surprisingly, sleep and gene expression in clockjrk flies showed opposite trends to w1118 flies. The present study indicates that acute hypoxia disrupt circadian rhythms through the activation of sima/HIF-1α, leading to the onset of sleep disorders, with Clock signaling potentially serving as a contributing factor.
Collapse
Affiliation(s)
- Shuwei Wang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou 730000, China; Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou 253000, China
| | - Shihong Zhou
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Xiaolin Jiang
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou 730000, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Dan Yang
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou 730000, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Jianzheng He
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou 730000, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou 730000, China.
| | - Minghui Xiu
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou 730000, China.
| |
Collapse
|
3
|
Gao C, Lim ASP, Haghayegh S, Cai R, Yang J, Yu L, Ibanez A, Buchman AS, Bennett DA, Gao L, Hu K, Li P. Reduced Complexity of Pulse Rate Is Associated With Faster Cognitive Decline in Older Adults. J Am Heart Assoc 2025; 14:e041448. [PMID: 40331928 DOI: 10.1161/jaha.125.041448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Cardiovascular diseases are closely linked to cognitive health. Subclinical cardiovascular functional changes, such as cardiac autonomic dysfunction, precede cardiovascular diseases and improve risk stratification. Continuous monitoring of heart rate or pulse rate is a commonly used approach to evaluate cardiac cycle and autonomic regulation. We investigated whether the complexity of pulse rate is associated with longitudinal cognitive decline in older adults. METHODS Overnight pulse oximetry data were collected from 503 participants (mean age=82±7 [SD] years, 76% female). We used a previously established distribution entropy algorithm to extract the complexity of pulse rate as a proxy for subclinical cardiovascular function. Participants completed a standardized cognitive test battery during the same visit of pulse oximetry and at least 1 follow-up visit. Linear mixed-effects models were conducted to test whether distribution entropy is associated with longitudinal changes in global cognition and separately, in 5 cognitive domains. RESULTS Greater distribution entropy (ie, better complexity) was associated with a slower decline in global cognition; the effect of 1-SD increase in distribution entropy was equivalent to being approximately 3 years younger. No associations were observed between conventional time- or frequency-domain pulse rate variability measures and cognitive changes. CONCLUSIONS Higher complexity of pulse rate is linked with slower cognitive decline in older adults. Future studies should test whether complexity is also associated with future risks of neurodegenerative disorders, such as dementia, and further elucidate the causal directions.
Collapse
Affiliation(s)
- Chenlu Gao
- Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Boston Massachusetts USA
- Division of Sleep and Circadian Disorders Brigham and Women's Hospital Boston Massachusetts USA
- Division of Sleep Medicine Harvard Medical School Boston Massachusetts USA
| | - Andrew S P Lim
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre University of Toronto Ontario Canada
| | - Shahab Haghayegh
- Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Boston Massachusetts USA
- Division of Sleep and Circadian Disorders Brigham and Women's Hospital Boston Massachusetts USA
- Division of Sleep Medicine Harvard Medical School Boston Massachusetts USA
| | - Ruixue Cai
- Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Boston Massachusetts USA
| | - Jingyun Yang
- Rush Alzheimer's Disease Center Rush University Medical Center Chicago Illinois USA
| | - Lei Yu
- Rush Alzheimer's Disease Center Rush University Medical Center Chicago Illinois USA
| | - Agustin Ibanez
- Latin American Brain Health Institute (BrainLat) Universidad Adolfo Ibañez Santiago Chile
- Cognitive Neuroscience Center (CNC) Universidad de San Andres Buenos Aires Argentina
- Global Brain Health Institute (GBHI) Trinity College Dublin Dublin Ireland
| | - Aron S Buchman
- Rush Alzheimer's Disease Center Rush University Medical Center Chicago Illinois USA
| | - David A Bennett
- Rush Alzheimer's Disease Center Rush University Medical Center Chicago Illinois USA
| | - Lei Gao
- Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Boston Massachusetts USA
- Division of Sleep and Circadian Disorders Brigham and Women's Hospital Boston Massachusetts USA
- Division of Sleep Medicine Harvard Medical School Boston Massachusetts USA
| | - Kun Hu
- Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Boston Massachusetts USA
- Division of Sleep and Circadian Disorders Brigham and Women's Hospital Boston Massachusetts USA
- Division of Sleep Medicine Harvard Medical School Boston Massachusetts USA
| | - Peng Li
- Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Boston Massachusetts USA
- Division of Sleep and Circadian Disorders Brigham and Women's Hospital Boston Massachusetts USA
- Division of Sleep Medicine Harvard Medical School Boston Massachusetts USA
| |
Collapse
|
4
|
Khandayataray P, Murthy MK. Exploring the nexus: Sleep disorders, circadian dysregulation, and Alzheimer's disease. Neuroscience 2025; 574:21-41. [PMID: 40189132 DOI: 10.1016/j.neuroscience.2025.03.066] [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: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
We reviewed the connections among Alzheimer's disease (AD), sleep deprivation, and circadian rhythm disorders. Evidence is mounting that disrupted sleep and abnormal circadian rhythms are not merely symptoms of AD, but are also involved in accelerating the disease. Amyloid-beta (Aβ) accumulates, a feature of AD, and worsens with sleep deprivation because glymphatic withdrawal is required to clear toxic proteins from the brain. In addition, disturbances in circadian rhythm can contribute to the induction of neuroinflammation and oxidative stress, thereby accelerating neurodegenerative processes. While these interactions are bidirectional, Alzheimer's pathology further disrupts sleep and circadian function in a vicious cycle that worsens cognitive decline, which is emphasized in the review. The evidence that targeting sleep and circadian mechanisms may serve as therapeutic strategies for AD was strengthened by this study through the analysis of the molecular and physiological pathways. Further work on this nexus could help unravel the neurobiological mechanisms common to the onset of Alzheimer's and disrupted sleep and circadian regulation, which could result in earlier intervention to slow or prevent the onset of the disease.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India.
| |
Collapse
|
5
|
Li P, Hu K. Disturbances in rest-activity rhythms and their neurobiological correlates: implications for Alzheimer's disease and dementia. Sleep 2025; 48:zsaf047. [PMID: 40045738 PMCID: PMC12068051 DOI: 10.1093/sleep/zsaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Affiliation(s)
- Peng Li
- Medical Biodynamics Center, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kun Hu
- Medical Biodynamics Center, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
6
|
Prakash BA, Shah I, Ni G, Vasudevan S, Jagannath A, Foster RG. Dreaming of Better Treatments: Advances in Drug Development for Sleep Medicine and Chronotherapy. J Sleep Res 2025:e70087. [PMID: 40346938 DOI: 10.1111/jsr.70087] [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: 03/18/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/12/2025]
Abstract
Throughout history, the development of new sleep medicines has been driven by progress in our understanding of the mechanisms underlying sleep. Ancient civilisations used their understanding of the sedative nature of natural herbs and compounds to induce sleep. The discovery of barbiturates and bromides heralded a new era of synthetic sleep medicine in the 19th century. This was followed by the development of benzodiazepines that were used to inhibit signalling throughout the brain by promoting gamma-amino butyric acid release and thereby produce loss of consciousness. As our understanding of sleep has deepened, newer therapies have more specifically targeted the wake-inducing neurotransmitter orexin with fewer side effects. Given the newly highlighted role of kinases in sleep/wake regulation, we predict that the next breakthroughs in sleep medicine will likely target these kinases. Given the fundamental role that sleep plays in maintaining brain health through processes such as glymphatic clearance, sleep medicine has therapeutic potential beyond just sleep. Recent evidence suggests that sleep disruptions directly contribute to the build-up of pathological neuronal proteins in neurodegenerative disorders. Therefore, sleep medicine could improve prognosis in disorders such as these. Great attention must be paid to the mechanism of action of each sleep medicine, however, as sleep medicines which do not fully mimic sleep could actually worsen disease progression.
Collapse
Affiliation(s)
| | - Ishani Shah
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Guohao Ni
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Aarti Jagannath
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, Oxford, UK
| | - Russell G Foster
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Tian Y, Kang L, Ha NT, Deng J, Liu D. Hydrogen peroxide in midbrain sleep neurons regulates sleep homeostasis. Cell Metab 2025:S1550-4131(25)00254-2. [PMID: 40378838 DOI: 10.1016/j.cmet.2025.04.016] [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: 04/15/2024] [Revised: 01/13/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
Sleep could protect animals from oxidative damage, yet the dynamic interplay between the redox state and sleep homeostasis remains unclear. Here, we show that acute sleep deprivation (SD) in mice caused a general increase in brain oxidation, particularly in sleep-promoting regions. In vivo imaging of intracellular hydrogen peroxide (H2O2) real-time dynamics revealed that in nigra sleep neurons, the increase in cytosolic but not mitochondrial H2O2 reflects sleep debt and tracks spontaneous wakefulness by positively correlating with wake duration. By controllably manipulating intraneuronal H2O2, we discovered that H2O2 elevation is required for compensatory sleep and causally promotes sleep initiation, at least partly dependent on transient receptor potential melastatin 2 (TRPM2) channel. However, excessive H2O2 induced brain inflammation and sleep fragmentation. Together, our study demonstrates intraneuronal H2O2 as a crucial signaling molecule that translates brain redox imbalance into sleep drive and underscores the significance of oxidative eustress in sleep homeostasis.
Collapse
Affiliation(s)
- Yujing Tian
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Luwei Kang
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ngoc T Ha
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Danqian Liu
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
8
|
Gu J, Yang M, Zhang L, Liu Y, Yan R, Pan D, Qian X, Hu H, Chu D, Hu C, Liu F, Cui H. Rhythmic TDP-43 affects RNA splicing of USP13, resulting in alteration of BMAL1 ubiquitination. J Cell Biol 2025; 224:e202405142. [PMID: 40202498 PMCID: PMC11980682 DOI: 10.1083/jcb.202405142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/20/2024] [Accepted: 02/11/2025] [Indexed: 04/10/2025] Open
Abstract
Circadian rhythm disorders are common characteristics of neurodegenerative diseases. The pathological aggregation of transactive response DNA-binding protein 43 (TDP-43) is associated with multiple neurodegenerative diseases, such as amyotrophic lateral sclerosis. However, the relationship between TDP-43 and circadian rhythm remains unknown. Here, we found that TDP-43 is rhythmically expressed both in vivo and in vitro. TDP-43 knockdown affected the expression of circadian genes, including BMAL1, CLOCK, CRY1, and PER2, and impaired autonomous circadian wheel behavior, cognitive functions, and balance abilities in mice. Furthermore, TDP-43 knockdown induced aberrant splicing of ubiquitin-specific peptidase 13 (USP13) and blocked USP13 rhythmic expression, enhancing the ubiquitination of BMAL1. Meanwhile, TDP-43 knockdown altered the rhythmic expression of phospho-AMPKα (Thr172) and platelet-type phosphofructokinase (PFKP), which may change cellular glucose uptake and ATP production. Our findings further the understanding of the role of TDP-43 dysfunction in circadian rhythm disruption in neurodegenerative diseases and provide new mechanistic evidence supporting the interaction between circadian rhythm disruption and neurodegeneration.
Collapse
Affiliation(s)
- Jianlan Gu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Mingming Yang
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Liti Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yuxiao Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Ruolan Yan
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Danmin Pan
- Department of Cell Biology, School of Life Sciences, Nantong University, Nantong, China
| | - Xiaowei Qian
- Department of Cell Biology, School of Life Sciences, Nantong University, Nantong, China
| | - Hanjing Hu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Dandan Chu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Chen Hu
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Hengxiang Cui
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Galushkin A, Gozes I. Intranasal NAP (Davunetide): Neuroprotection and circadian rhythmicity. Adv Drug Deliv Rev 2025; 220:115573. [PMID: 40185278 DOI: 10.1016/j.addr.2025.115573] [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: 11/18/2024] [Revised: 02/05/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
In this review we examine the neuroprotective potential of NAP (davunetide), a small peptide derived from Activity-Dependent Neuroprotective Protein (ADNP), in the context of neurodevelopmental and neurodegenerative disorders. ADNP, a protein essential for brain development and function, is associated with tauopathy-related diseases, such as Alzheimer's Disease (AD), and circadian rhythm regulation. NAP enhances microtubule stability and prevents tauopathy. In preclinical studies, NAP shows promise in improving cognitive performance and correcting behavioral deficits in different models. Clinical studies on NAP (davunetide) administered via intranasal delivery have demonstrated its safety, favorable bioavailability, and potential efficacy in improving cognitive function, making it a viable therapeutic option. In the pure tauopathy, progressive supranuclear palsy, NAP (davunetide) significantly slowed disease progression in women in a phase II-III clinical trial. Additionally, the complex interactions between ADNP, associated pathways, and circadian regulation and the extensive NAP compensation upon ADNP deficiency attest to further clinical development. Thus, NAP is an example of a reductionist approach in drug delivery, replacing/enhancing the critical large ADNP-related pathways including dysregulated microtubules and tauopathy with a small brain bioavailable investigational drug, davunetide.
Collapse
Affiliation(s)
- Artur Galushkin
- Dr. Diana and Zelman Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical & Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Illana Gozes
- Dr. Diana and Zelman Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical & Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
10
|
Seke M, Stankovic A, Zivkovic M. Capacity of fullerenols to modulate neurodegeneration induced by ferroptosis: Focus on multiple sclerosis. Mult Scler Relat Disord 2025; 97:106378. [PMID: 40088719 DOI: 10.1016/j.msard.2025.106378] [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: 12/27/2023] [Revised: 01/10/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Multiple sclerosis is an inflammatory disease of the central nervous system (CNS), characterized by oligodendrocyte loss and demyelination of axons leading to neurodegeneration and severe neurological disability. Despite the existing drugs that have immunomodulatory effects an adequate therapy that slow down or stop neuronal death has not yet been found. Oxidative stress accompanied by excessive release of iron into the extracellular space, mitochondrial damage and lipid peroxidation are important factors in the controlled cell death named ferroptosis, latterly recognized in MS. As the fullerenols exhibit potent antioxidant activity, recent results imply that they could have protective effects by suppressing ferroptosis. Based on the current knowledge we addressed the main mechanisms of the protective effects of fullerenols in the CNS in relation to ferroptosis. Inhibition of inflammation, iron overload and lipid peroxidation through the signal transduction mechanism of Nuclear Factor Erythroid 2-Related Factor 2 (NRF2), chelation of heavy metals and free radical scavenging using fullerenols are proposed as benefitial strategy preventing MS progression. Current review connects ferroptosis molecular targets and important factors of MS progression, with biomedical properties and mechanisms of fullerenols' actions, to propose new treatment strategies that could be addaptobale in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Mariana Seke
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia.
| |
Collapse
|
11
|
Tafuri B, Giugno A, Nigro S, Zoccolella S, Barone R, Tamburrino L, Gnoni V, Urso D, Rollo E, De Blasi R, Logroscino G. Radiomic alterations and clinical correlates of hypothalamic nuclei in ALS. Comput Biol Med 2025; 189:109906. [PMID: 40020551 DOI: 10.1016/j.compbiomed.2025.109906] [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: 10/28/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
OBJECTIVE The aim of this study is to analyze hypothalamic changes and clinical/metabolic correlates with a radiomic approach in Amyotrophic Lateral Sclerosis (ALS). METHODS We retrospectively identified 54 sporadic ALS patients and 53 matched controls. We compared radiomics features over hypothalamic subunits in T1-weighted. Semi-partial correlation (Spearman's correlation) assessed the relationship between Body Mass Index (BMI) and clinical scores with radiomics features. We considered only moderate correlations (rho>|0.4|). RESULTS Compared to HC, individuals with ALS showed significantly higher values of radiomic measures in the left Anterior-Inferior, Posterior and Inferior-Tubular hypothalamic subunits. Similarly, right hypothalamic nuclei reported significant differences in Anterior-Superior, Posterior and Inferior-Tubular nuclei. Two radiomics measures of randomness of the intensities in left Anterior-Inferior subunit showed highly significant correlation with greater BMI values. Higher local homogeneity of the right Inferior-Tubular subunit corresponded to higher ALS Functional Rating Scale-Revised (ALSFRS-r), while finer textures of the left Anterior-Superior subunit were negatively related with disease progression rate. CONCLUSIONS These results support the hypothesis that a degenerative process affecting hypothalamus in ALS extends beyond the atrophy process. Intriguingly, the close relationship between the entropy of left Anterior-Inferior nucleus and the higher BMI may further demonstrate the critical role of hypothalamus in eating abnormalities. Furthermore, the inhomogeneity of the right Inferior-Tubular subunit reflects a more severe clinical condition by ALSFRS-R. This work represents a significant advancement in the study of ALS and its association with hypothalamic changes through a novel radiological approach, uncovering new associations between sub-hypothalamic radiomic changes, anthropometric measures, and disease outcomes.
Collapse
Affiliation(s)
- Benedetta Tafuri
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Bari, Italy; Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy; Department of Engineering of Innovation, University of Salento, Lecce, Italy.
| | - Alessia Giugno
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy; Department of Medical and Surgical Sciences, Institute of Neurology, Magna Graecia University, Catanzaro, Italy.
| | - Salvatore Nigro
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy; Institute of Nanotechnology, National Research Council (CNR-NANOTEC), Lecce, Italy.
| | - Stefano Zoccolella
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy; Neurology Unit, San Paolo Hospital, Azienda Sanitaria Locale (ASL) Bari, Bari, Italy.
| | - Roberta Barone
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy.
| | - Ludovica Tamburrino
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Bari, Italy; Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy.
| | - Valentina Gnoni
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy.
| | - Daniele Urso
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy.
| | - Eleonora Rollo
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy; Department of Neurosciences, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Roberto De Blasi
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy.
| | - Giancarlo Logroscino
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Bari, Italy; Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione "Card. G. Panico", Tricase, Italy.
| |
Collapse
|
12
|
Song QX, Suadicani SO, Negoro H, Jiang HH, Jabr R, Fry C, Xue W, Damaser MS. Disruption of circadian rhythm as a potential pathogenesis of nocturia. Nat Rev Urol 2025; 22:276-293. [PMID: 39543359 DOI: 10.1038/s41585-024-00961-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
Increasing evidence suggested the multifactorial nature of nocturia, but the true pathogenesis of this condition still remains to be elucidated. Contemporary clinical medications are mostly symptom based, aimed at either reducing nocturnal urine volume or targeting autonomic receptors within the bladder to facilitate urine storage. The day-night switch of the micturition pattern is controlled by circadian clocks located both in the central nervous system and in the peripheral organs. Arousal threshold and secretion of melatonin and vasopressin increase at night-time to achieve high-quality sleep and minimize nocturnal urine production. In response to the increased vasopressin, the kidney reduces the glomerular filtration rate and facilitates the reabsorption of water. Synchronously, in the bladder, circadian oscillation of crucial molecules occurs to reduce afferent sensory input and maintain sufficient bladder capacity during the night sleep period. Thus, nocturia might occur as a result of desynchronization in one or more of these circadian regulatory mechanisms. Disrupted rhythmicity of the central nervous system, kidney and bladder (known as the brain-kidney-bladder circadian axis) contributes to the pathogenesis of nocturia. Novel insights into the chronobiological nature of nocturia will be crucial to promote a revolutionary shift towards effective therapeutics targeting the realignment of the circadian rhythm.
Collapse
Affiliation(s)
- Qi-Xiang Song
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sylvia O Suadicani
- Department of Urology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hiromitsu Negoro
- Department of Urology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hai-Hong Jiang
- Department of Urology and Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rita Jabr
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Christopher Fry
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Margot S Damaser
- Department of Biomedical Engineering, Lerner Research Institute and Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
13
|
Bianchin MM, Zimmer ER. Wake-Up Call: The Association Between Sleep Disturbances and Dementia. Neurology 2025; 104:e213516. [PMID: 40106754 DOI: 10.1212/wnl.0000000000213516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/28/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
- Marino Muxfeldt Bianchin
- Division of Neurology, Hospital de Clínicas de Porto Alegre, Brazil
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Brazil
- BRAIN - Basic Research and Advanced Investigations in Neurosciences, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Brazil
| | - Eduardo Rigon Zimmer
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica (PPGbioq) e Farmacologia e Terapêutica (PPGFT), Universidade Federal do Rio Grande do Sul, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Brazil; and
- McGill University, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Sarsembayeva D, Schreuder MJ, Huisman M, Kok A, Wagner M, Capuano AW, Hartman CA. Individual Sleep Problems Are Associated With an Accelerated Decline in Multiple Cognitive Functions in Older Adults. J Sleep Res 2025:e70067. [PMID: 40262553 DOI: 10.1111/jsr.70067] [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: 07/11/2024] [Revised: 03/06/2025] [Accepted: 04/06/2025] [Indexed: 04/24/2025]
Abstract
Poor sleep is a known risk factor of cognitive disorders, but the role of individual sleep problems in age-related cognitive changes remains unclear. This study used two complementary statistical models to estimate nonlinear trajectories of decline in four domains of cognitive functioning in the age period between 55 and 100 years depending on the severity of problems with falling asleep, night awakenings, and early morning awakenings, and short/long sleep duration. The sample included 5132 older adults (M = 67 years, 48% male) from the Longitudinal Aging Study Amsterdam (LASA), assessed 4-10 times every 2-3 years. Sleep problems were self-reported, and cognitive functioning was measured with the 15-Word test (reflecting episodic memory as immediate and delayed recall), Coding task (information processing speed) and Mini-mental State Examination/MMSE (global cognition). Data were analysed using quadratic and piecewise changepoint mixed models. The piecewise models provided more precise and interpretable findings. Decline in information processing speed accelerated significantly earlier in participants with short sleep duration (regression coefficient (B) = -2.3[95% confidence interval (CI): -3.86; -0.81]; p < 0.01) and faster with more severe early morning awakenings (B = -0.07 [-0.1; -0.03]; p < 0.01). Decline in immediate recall accelerated earlier in those with short sleep (B = -2.8 [-4.44; -1.14]; p < 0.01) and severe problems with falling asleep (B = -1.22 [-2.06; -0.39]; p = 0.01). Decline in delayed recall was faster with long sleep (B = -0.06 [-0.08;-0.03]; p < 0.01). Decline in global cognition accelerated faster in those with short/long sleep duration (B = -0.07 [-0.13; -0.01]/-0.10 [-0.18; -0.03]; p < 0.01) and severe night awakenings (B = -0.04 [-0.07;-0.02]; p < 0.01). To conclude, this study showed that some sleep problems can differentially predict earlier acceleration of deterioration in specific cognitive functions in older adults.
Collapse
Affiliation(s)
- Dina Sarsembayeva
- Interdisciplinary Centre Psychopathology and Emotion Regulation, University Center Psychiatry, University Medical Center Groningen, Groningen, the Netherlands
| | - Marieke J Schreuder
- Department of Psychology and Education Sciences, Quantitative Psychology and Individual Differences, KU Leuven, Leuven, Belgium
- Department of Developmental Psychology, Tilburg University, Tilburg, the Netherlands
| | - Martijn Huisman
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, the Netherlands
| | - Almar Kok
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, the Netherlands
| | - Maude Wagner
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Ana W Capuano
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Catharina A Hartman
- Interdisciplinary Centre Psychopathology and Emotion Regulation, University Center Psychiatry, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
15
|
Milton S, Cavaillès C, Ancoli-Israel S, Stone KL, Yaffe K, Leng Y. Five-Year Changes in 24-Hour Sleep-Wake Activity and Dementia Risk in Oldest Old Women. Neurology 2025; 104:e213403. [PMID: 40106755 PMCID: PMC11919274 DOI: 10.1212/wnl.0000000000213403] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Sleep disruptions are associated with cognitive aging in older adults. However, little is known about longitudinal sleep changes in the oldest old and whether these changes are linked to cognitive impairment. We aimed to determine whether changes in 24-hour multidimensional sleep-wake activity are associated with mild cognitive impairment (MCI) and dementia in oldest old women. METHODS We studied cognitively unimpaired women enrolled in the Study of Osteoporotic Fractures who completed wrist actigraphy twice (baseline and follow-up) and had cognitive status evaluated at follow-up using a neuropsychological battery and adjudication. To identify multidimensional sleep-wake change profiles, we performed hierarchical clustering on principal components on the 5-year changes (median 5.0 [range 3.5-6.3] years) in nighttime sleep (sleep duration, sleep efficiency [SE], and wake after sleep onset [WASO]), napping (duration and frequency), and circadian rest-activity rhythms (RARs; acrophase, amplitude, mesor, and robustness). Using multinomial logistic regression, we evaluated the associations between these profiles-and individual parameter changes-and MCI and dementia risk at follow-up. RESULTS Of 733 participants (mean age 82.5 ± 2.9 years), 164 (22.4%) developed MCI and 93 (12.7%) developed dementia by the follow-up visit. We identified 3 sleep-wake change profiles: stable sleep (SS; n = 321 [43.8%]) was characterized by stability or small improvements; declining nighttime sleep (n = 256 [34.9%]) showed decreases in nighttime sleep quality and duration, moderate napping increases, and worsening circadian RARs; and increasing sleepiness (IS; n = 156 [21.3%]) exhibited large increases in daytime and nighttime sleep duration and quality, and worsening circadian RARs. After adjustment for age, education, race, body mass index, diabetes, hypertension, myocardial infarction, antidepressant use, and baseline cognition, women with IS had approximately double the risk of dementia (odds ratio 2.21, 95% CI 1.14-4.26) compared with those with SS. SE, WASO, nap duration, and nap frequency were individually associated with dementia. Neither sleep-wake change profiles nor individual parameters were associated with MCI. DISCUSSION Among community-dwelling women in their 80s, those with increasing 24-hour sleepiness over 5 years had doubled dementia risk during that time. Change in multidimensional 24-hour sleep-wake activity may serve as an early marker or risk factor for dementia in oldest old women.
Collapse
Affiliation(s)
- Sasha Milton
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| | - Clémence Cavaillès
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| | | | - Katie L Stone
- Research Institute, California Pacific Medical Center, San Francisco
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Department of Neurology, University of California, San Francisco; and
- San Francisco Veterans Affairs Health Care System, CA
| | - Yue Leng
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| |
Collapse
|
16
|
Maybrier HR, Jackson JJ, Toedebusch CD, Lucey BP, Head D. Influence of sleep and cardiovascular health on cognitive trajectories in older adults. Neurobiol Aging 2025; 152:34-42. [PMID: 40318496 DOI: 10.1016/j.neurobiolaging.2025.04.008] [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: 10/18/2024] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025]
Abstract
Age-related changes in sleep have been associated with cognitive decline, yet causal pathways have not been identified. Evidence suggests reduced cardiovascular health may be a consequence of poor sleep and a precursor to cognitive decline. This observational cohort study used path analyses to determine whether cardiovascular disease risk mediated or moderated effects of sleep on yearly longitudinal change in cognition, estimated with linear growth models. Total sleep time (TST), sleep efficiency (SE), and relative spectral power of slow wave activity (SWA; 1-4 Hz) and slow oscillations (SO; 0.5-1 Hz), were measured with single-channel home EEG. Cardiovascular disease risk (CVR) was estimated as 10-year Framingham Risk Score 1-year post-sleep. Outcomes were yearly change in executive function (EF), episodic memory (EM), and processing speed (PS) over 2-5 years post-sleep. 342 participants (mean age 73.5 +/- 5.6 years, 51 % female) were included. Shorter TST was linearly associated with increased CVR across all models (βs = -0.18(0.058) - -0.19(0.059), ps< 0.002). TST was indirectly associated with EF and PS decline through CVR, such that associations between short TST and cognitive decline were partially due to higher CVR. All other mediating and moderating effects were nonsignificant after multiple comparisons. Indirect associations between short sleep duration and greater decline in executive function and processing speed were found through higher CVR, suggesting a potential mechanism by which sleep leads to cognitive decline. Findings support the prioritization of adequate sleep duration to preserve both cardiovascular and cognitive health in later life.
Collapse
Affiliation(s)
- Hannah R Maybrier
- Department of Psychological and Brain Sciences, Washington University, St. Louis, MO, United States
| | - Joshua J Jackson
- Department of Psychological and Brain Sciences, Washington University, St. Louis, MO, United States
| | - Cristina D Toedebusch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Denise Head
- Department of Psychological and Brain Sciences, Washington University, St. Louis, MO, United States; Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, United States; Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
17
|
Sirimaharaj N, Thiankhaw K, Chattipakorn N, Chattipakorn SC. Unveiling the Protective Roles of Melatonin on Glial Cells in the Battle Against Alzheimer's Disease-Insights from In Vivo and In Vitro Studies. Mol Neurobiol 2025:10.1007/s12035-025-04904-7. [PMID: 40208552 DOI: 10.1007/s12035-025-04904-7] [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: 11/09/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder that predominantly affects the elderly. Characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles, AD leads to memory loss, cognitive decline, and severe behavioral changes. As the most common form of dementia, AD imposes a significant global health burden, highlighting the need for interventions that address underlying disease mechanisms rather than only symptomatic treatment. Glial cells, including microglia and astrocytes, play a crucial role in AD progression by mediating neuroinflammatory responses and modulating Aβ clearance and neuronal health. Dysfunction in these cells can exacerbate neuroinflammation and neuronal damage, making glial cells an important target for therapeutic intervention. This review synthesizes findings from in vivo and in vitro studies on melatonin's effects on glial cell dysfunction in AD, emphasizing the multi-mechanistic nature of its neuroprotective properties. Recent studies highlight melatonin's potential as a therapeutic agent that addresses AD-related mechanisms through its interactions with glial cells. Melatonin has demonstrated protective effects, including reducing oxidative stress, apoptosis, and inflammation, inhibiting Aβ fibrillogenesis, and modulating amyloid precursor proteins. Additionally, its influence on glial cell activity, through melatonin receptor pathways, suggests it can alleviate neuroinflammation, a key component of AD progression. The collective evidence points to melatonin's promise as a therapeutic tool with potential roles in both preventive and adjunctive treatments for AD. However, further research is necessary to establish its efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Nopdanai Sirimaharaj
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
18
|
Wang J, Pan H, Tang H, Zhang J, Li T, Liu Y, Huang Y, Fei Z, Wang Y. Shuangxia Decoction attenuates sleep disruption in 5×FAD mice through neuroinflammation inhibition: An integrative analysis of transcriptomic and molecular biology investigations. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119642. [PMID: 40101857 DOI: 10.1016/j.jep.2025.119642] [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: 12/13/2024] [Revised: 02/23/2025] [Accepted: 03/15/2025] [Indexed: 03/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory and learning deficits. Circadian rhythm disruption-induced sleep disruption is frequently observed in AD patients. The Shuangxia Decoction (SXD) comprising Pinellia ternata (Thunb.) Breit. (Banxia) and Prunella vulgaris L. (Xiakucao), has been effectively used to treate sleep disruption for thousands of years. However, the mechanisms by which SXD treated AD through circadian rhythm-related pathways remain unexplored. AIMS OF THE STUDY This research sought to determine the efficacy, mechanisms, and active compounds of SXD in AD treatment via an integrative approach. MATERIALS AND METHODS We conducted a chronic jet lag (CJL) protocol in wild-type (WT) mice and monitored their rest/activity to compare their rest/activity period among WT, CJL, and CJD + SXD groups. In addition, we evaluated the impact of SXD on the cognitive and Aβ burden of 5 × FAD mice by behavioral tests and Thioflavin staining. The underlying pathway analysis of SXD was revealed through transcriptomic and biology experimental validation. The active compounds of SXD were further analyzed using the UPLC-MS, molecular docking, and cellular thermal shift assay (CESTA). RESULTS Our study demonstrated a rapid recovery of rest/activity period in CJL mice following SXD treatment. Additionally, SXD treatment alleviated Aβ plaque accumulation, subsequently preserving cognitive behavior and motor ability in 5 × FAD mice. Moreover, SXD significantly enhanced neuronal synaptic plasticity dendritic plasticity in CA1 neurons of 5 × FAD mice. Transcriptomic analysis showed upregulation of the neuroinflammation-related pathway in 5 × FAD mice. Subsequent heatmap analysis indicated a suppression of inflammatory factor secretion (Cd68, Trem2, IL-6, IL-1β, Cxc3r1, Tnf et al.) and an increase of anti-inflammatory factor secretion (IL4, Ccl19, Ccl21a et al.) following SXD treatment in the 5 × FAD mice. Meanwhile, SXD upregulated positive regulators involved in the circadian rhythm like Bmal1 and Clock, and downregulated negative regulators like Nr1d1. Moreover, microglia exhibited an amoeboid morphology characterized by few processes and rounded cell bodies in 5 × FAD mice, whereas the age-matched SXD group maintained microglia with a ramified appearance. Additionally, our study identified 20 major components of SXD and identified 3-(3,4-Dihydroxyphenyl) lactic acid, Salviaflaside, and Ilexhainanoside D for further molecular docking with REV-ERBα (NR1D1), a commonly used circadian target. Salviaflaside further showed a strong bind with REV-ERBα via CESTA. CONCLUSIONS Our findings indicate that SXD may rescue circadian rhythm in 5 × FAD mice through specifically binding to REV-ERBα in microglia to activate the BMAL1/CLOCK pathway, thus inhibiting transcription of inflammatory factors, contributing to alleviating neuroinflammation and impeding AD progression. Our results offer a scientific foundation for developing SXD-based therapies in the early stage of AD, where sleep disruption precedes cognitive decline, offering potential leads for clinical trials to improve sleep quality thus delaying neurodegeneration in AD patients.
Collapse
Affiliation(s)
- Jie Wang
- Department of Chinese Medicine & Integrative Medicine, Shanghai Geriatric Medical Center, Zhongshan Hospital, Fudan University, 2560 Chunshen Road, Shanghai, 201104, China; Department of Chinese Medicine & Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 201104, China
| | - Hao Pan
- Department of Neurosurgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haiyan Tang
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jingsi Zhang
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tingting Li
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yizhou Liu
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiwen Huang
- Endocrinology Department of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Zhimin Fei
- Department of Neurosurgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yu Wang
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
19
|
Tian ZY, Jiang B, Jin M, Yu XK, Chen QL, Wang JH. Alzheimer's disease and insomnia: a bibliometric study and visualization analysis. Front Aging Neurosci 2025; 17:1542607. [PMID: 40264463 PMCID: PMC12011777 DOI: 10.3389/fnagi.2025.1542607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Background Alzheimer's disease (AD) is the fastest-growing neurodegenerative disorder globally, with patient numbers expected to rise to 130 million by 2050. Insomnia, a prevalent comorbidity, exhibits a bidirectional relationship with AD: insomnia accelerates AD pathology, while AD worsens sleep disorders. This relationship has emerged as a key area of research. Current mechanisms involve oxidative stress, inflammatory responses, and glymphatic system dysfunction, yet a comprehensive review of these processes remains absent. Objective To conduct a visual analysis of the relationship between Alzheimer's disease and insomnia using CiteSpace. Methods Literature on "insomnia" and "Alzheimer's disease" published between January 1, 2000, and October 31, 2024, was retrieved from the Web of Science Core Collection. CiteSpace and VOSviewer software were used to analyze institutions, authors, and keywords. Results A total of 1,907 articles were analyzed, revealing a consistent upward trend in publication volume. The United States and the Mayo Clinic were identified as leading contributors, producing 704 and 57 publications, respectively. Boeve Bradley F the most prolific author contributed 30 publications. Collaboration was actively observed among countries, institutions, and authors. High-frequency keywords identified were "Parkinson's disease," "cognitive impairment," and "sleep behavior disorder." Emerging research areas are likely to focus on "sleep quality" and the "glymphatic system." Conclusion This study is the first to apply bibliometric analysis to identify three key trends in AD and insomnia research: the dominance of the United States and Mayo Clinic, strong international collaboration, and a focus on critical areas such as cognitive impairment, the glymphatic system, and sleep interventions. Insomnia may accelerate AD progression via multiple pathways, indicating that enhancing sleep quality could provide new strategies for early intervention. Future research should prioritize advancing the clinical translation of sleep interventions and investigating the mechanisms of the glymphatic system.
Collapse
Affiliation(s)
- Zi-Yue Tian
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Meng Jin
- The Third Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Xiao-Kun Yu
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Qi-Lin Chen
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- KweiChow Moutai Hospital, Zunyi, Guizhou, China
| | - Jia-Hui Wang
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
20
|
Yang H, Niu L, Tian L, Hu Y, Cheng C, Li S, Le W. Circadian rhythm disturbances in Alzheimer's disease: insights from plaque-free and plaque-burdened stages in APP SWE/PS1 dE9 mice. Alzheimers Res Ther 2025; 17:76. [PMID: 40188157 PMCID: PMC11971749 DOI: 10.1186/s13195-025-01724-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Disruptions in circadian rhythms are commonly observed in patients with Alzheimer's disease (AD) and could potentially accelerate the progression of the condition. However, the relationship between circadian rhythm disruptions and AD development, as well as the mechanisms involved, remain poorly understood. METHODS This study investigated the circadian behavior, rhythmic gene expression in multiple brain regions, and its correlation with sleep architecture of AD mice at two disease stages: plaque-free stage (2-month-old) and plaque-burdened stage (10-month-old) as compared to age-matched wild-type (WT) mice. RESULTS Two-month-old AD mice already displayed alteration in the activity patterns compared to WT mice, showing increased activity during the light phase and decreased activity during the dark phase, and the change in the activity pattern of 10-month-old AD mice was more significant. Further, electroencephalogram (EEG) examination showed increased wakefulness and reduced non-rapid eye movement (NREM) sleep in 2- and 10-month-old AD mice. In addition, we documented a significant change in circadian core clock genes in the suprachiasmatic nucleus (SCN), hippocampus, and cortex of 2- and 10-month-old AD mice. Correlation analyses demonstrated the close relationship between circadian clock gene expression level and specific sleep-wake parameters, especially within the SCN and hippocampus. CONCLUSIONS These findings revealed that circadian rhythm disturbances in AD mice preceded Aβ deposition. The circadian rhythm disturbances observed in the early AD might be attributed to the abnormal expression of core clock genes in the brain regions involved in circadian rhythm regulation.
Collapse
Affiliation(s)
- Huijia Yang
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Long Niu
- Department of Neurology, Heping Hospital affiliated to Changzhi Medical College, Changzhi, China
| | - Lulu Tian
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Yiying Hu
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Cheng Cheng
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Weidong Le
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China.
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
21
|
King MW, Jacob S, Sharma A, Lawrence JH, Weaver DR, Musiek ES. Circadian rhythms and the light-dark cycle interact to regulate amyloid plaque accumulation and tau phosphorylation in 5xFAD mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.645805. [PMID: 40236233 PMCID: PMC11996435 DOI: 10.1101/2025.03.31.645805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background Circadian disruption has long been appreciated as a downstream consequence of Alzheimer's Disease in humans. However, an upstream role for behavioral circadian disruption in regulating AD pathology remains an open question. Methods To determine the role of the central circadian clock in the suprachiasmatic nucleus (SCN) in regulating amyloid pathology, we crossed the 5xFAD amyloid mouse model with mice harboring deletion of the critical clock gene Bmal1 in GABAergic neurons using VGAT-iCre, which is expressed in >95% of SCN cells. To examine the role the light-dark cycle in this process, we aged these mice in either regular 12:12 light-dark (LD) or constant darkness (DD) conditions. Transcriptional, behavioral, and physiological rhythms were examined in VGAT-iCre; 5xFAD; Bmal1 fl/fl (VGAT-BMAL1KO;5xFAD) mice under varying light conditions. Amyloid plaque deposition, peri-plaque tau phosphorylation, and other pathology was examined by immunohistochemistry, and transcriptomic changes were examined by high-throughput qPCR. Results VGAT-BMAL1KO;5xFAD mice showed loss of SCN BMAL1 expression and severe disruption of behavioral rhythms in both LD and DD, with loss of day-night rhythms in consolidated sleep and blunting of rhythmic clock gene expression in the brain. Surprisingly, VGAT-BMAL1KO;5xFAD mice kept under LD showed reduced total plaque accumulation and peri-plaque tau phosphorylation, compared to Cre-negative controls. These changes were gated by the light-dark cycle, as they were absent in VGAT-BMAL1KO;5xFAD mice kept in DD conditions. Total plaque accumulation was also reduced in control 5xFAD mice kept in DD as compared to LD, suggesting a general effect of light-dark cycle on amyloid aggregation. Expression of murine presenilin 1 (Psen1) -- which catalyzes the processing of sAPPβ into Aβ -- as well as APP cleavage to C-terminal fragments, were suppressed in VGAT-BMAL1KO;5xFAD under LD conditions. Conclusions These studies elucidated an interaction between the circadian clock in GABAergic neurons and the light-dark cycle in regulating amyloid pathology and suggest that decoupling the central clock form the light-dark cycle may reduce APP cleavage and plaque formation. These results call into question the proposed simple positive feedback loop between circadian rhythm disruption and Alzheimer's Disease pathology.
Collapse
|
22
|
Xu X, Xu L, Lang Z, Sun G, Pan J, Li X, Bian Z, Wu X. Identification of potential susceptibility loci for non-small cell lung cancer through whole genome sequencing in circadian rhythm genes. Sci Rep 2025; 15:7825. [PMID: 40050692 PMCID: PMC11885630 DOI: 10.1038/s41598-025-92083-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
Lung cancer is a malignant tumor with a high morbidity and mortality rate worldwide, causing an increasing disease burden. Of these, the most common type is non-small cell lung cancer (NSCLC), which accounts for 80-85% of all lung cancer cases. Genetic research is crucial for continuously discovering susceptibility genes related to lung cancer for in-depth study. The role of genetic predisposition in the development of NSCLC, particularly within circadian rhythm pathways known to govern various physiological processes, is increasingly acknowledged. Yet, the association between genetic variants of circadian rhythm-related genes and NSCLC susceptibility among Chinese populations is not fully understood. This study carried out a two-phase (discovery and validation stages) research design to identify genetic variants associated with NSCLC risk within the circadian rhythm pathway. We employed extensive whole-genome sequencing (WGS) for 1,104 NSCLC cases and 9,635 controls. FastGWA-GLMM was used for single-locus risk association analysis of NSCLC, and we screened candidate SNPs in the validation set that comprised 4,444 cases and 174,282 controls from the Biobank Japan Project (BBJ). Furthermore, GCTA-COJO conditional analysis was utilized to confirm SNPs related to NSCLC risk. Finally, potential genetic variations that may regulate gene expression were explored in GTEx and QTLbase. RNA sequencing data were utilized for transcriptomic verification. Our study identified eight candidate SNPs associated with NSCLC susceptibility within the circadian rhythm pathway that met the requirement with P < 0.05 in both the discovery and validation populations. After conditional analysis, five of these SNPs remained. The A allele of CUL1 rs78524436 (ORmeta = 1.18, 95%CI: 1.09-1.29, Pmeta = 7.99e-5) and the A allele of TEF rs9611588 (ORmeta = 1.06, 95%CI: 1.02-1.10, Pmeta = 1.28e-3) were associated with an increased risk of NSCLC. The A allele of FBXL21 rs2069868 (ORmeta = 0.86, 95%CI: 0.80-0.96, Pmeta = 4.78e-4), the T allele of CSNK1D rs147316973 (ORmeta = 0.76, 95%CI: 0.65-0.88, Pmeta = 5.93e-4), and the A allele of RORA rs1589701 (ORmeta = 0.94, 95%CI: 0.91-0.98, Pmeta = 3.40e-3) were associated with a lower risk of NSCLC, separately. The eQTL results revealed an association between RORA rs1589701 and TEF rs9611588 with the expression levels of RORA and TEF, respectively. Transcriptome data indicated that RORA and TEF showed lower expression levels in tumor tissues compared to normal tissues (P < 0.001). Moreover, poorer survival was observed in patients with lower RORA and TEF expressions (log-rank P < 0.05). Our findings spotlight potential susceptibility loci within circadian rhythm pathway genes that modulate NSCLC carcinogenesis, which enriches the understanding of the genetic susceptibility of NSCLC in the Chinese population and provides a more solid basis for exploring the biological mechanism of circadian rhythm genes in NSCLC.
Collapse
Affiliation(s)
- Xiaohang Xu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, 310058, China
| | - Luopiao Xu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, 310058, China
| | - Zeyong Lang
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Gege Sun
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Junlong Pan
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xue Li
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, 310058, China
| | - Zilong Bian
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xifeng Wu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, 310058, China.
- National Institute for Data Science in Health and Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
- School of Medicine and Health Science, George Washington University, Washington, DC, USA.
| |
Collapse
|
23
|
Yang MY, Wu CN, Lin YT, Tsai MH, Hwang CF, Yang CH. Dissecting the Circadian Clock and Toll-like Receptor Gene Alterations in Meniere's Disease and Vestibular Migraine. Otolaryngol Head Neck Surg 2025; 172:999-1005. [PMID: 39675041 DOI: 10.1002/ohn.1085] [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: 08/27/2024] [Revised: 11/03/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE To investigate alterations in the expression of circadian clock and Toll-like receptor (TLR) genes in peripheral blood (PB) leukocytes of patients with Meniere's disease (MD) and vestibular migraine (VM), and determine whether these gene expressions can differentiate MD from VM. STUDY DESIGN Observational prospective study. SETTING Tertiary academic medical center. METHODS PB leukocytes were collected from patients diagnosed with MD and VM during recent vertigo attacks, as well as from healthy controls. The expression levels of 9 circadian clock genes and 6 TLR genes were analyzed using real-time quantitative reverse transcriptase-polymerase chain reaction. RESULTS Sixty-nine participants were enrolled, including 28 patients with MD, 14 patients with VM, and 27 healthy controls. Both MD and VM groups showed lower expression of PER1 compared to the control group (P < .01). The VM group exhibited significantly lower expression of PER1, PER2, CRY1, BMAL1, CLOCK, and TIM compared to the MD group (all P < .001). The MD group had higher TLR9 expression than the control group, and elevated TLR4, TLR8, and TLR9 expression compared to the VM group (P < .05). In the VM group, patients with severe dizziness handicaps had significantly lower expression of PER2, CRY1, CRY2, and CK1ε compared to those with mild to moderate handicaps (P < .05). CONCLUSION This study identifies distinct alterations in the circadian clock and TLR gene expression in MD and VM, suggesting potential differences in the pathogenesis of these 2 vertiginous disorders and highlighting the possibility of these gene expressions as biomarkers for differentiation.
Collapse
Affiliation(s)
- Ming-Yu Yang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Ching-Nung Wu
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Tsai Lin
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ming-Hsien Tsai
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chung-Feng Hwang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chao-Hui Yang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- School of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
24
|
Tofani GSS, Clarke G, Cryan JF. I "Gut" Rhythm: the microbiota as a modulator of the stress response and circadian rhythms. FEBS J 2025; 292:1454-1479. [PMID: 39841560 PMCID: PMC11927059 DOI: 10.1111/febs.17400] [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: 08/02/2024] [Revised: 11/20/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
Modern habits are becoming more and more disruptive to health. As our days are often filled with circadian disruption and stress exposures, we need to understand how our responses to these external stimuli are shaped and how their mediators can be targeted to promote health. A growing body of research demonstrates the role of the gut microbiota in influencing brain function and behavior. The stress response and circadian rhythms, which are essential to maintaining appropriate responses to the environment, are known to be impacted by the gut microbiota. Gut microbes have been shown to alter the host's response to stress and modulate circadian rhythmicity. Although studies demonstrated strong links between the gut microbiota, circadian rhythms and the stress response, such studies were conducted in an independent manner not conducive to understanding the interface between these factors. Due to the interconnected nature of the stress response and circadian rhythms, in this review we explore how the gut microbiota may play a role in regulating the integration of stress and circadian signals in mammals and the consequences for brain health and disease.
Collapse
Affiliation(s)
- Gabriel S. S. Tofani
- APC MicrobiomeUniversity College CorkIreland
- Department of Anatomy & NeuroscienceUniversity College CorkIreland
| | - Gerard Clarke
- APC MicrobiomeUniversity College CorkIreland
- Department of Psychiatry & Neurobehavioural ScienceUniversity College CorkIreland
| | - John F. Cryan
- APC MicrobiomeUniversity College CorkIreland
- Department of Anatomy & NeuroscienceUniversity College CorkIreland
| |
Collapse
|
25
|
Korkmaz H, Anstötz M, Wellinghof T, Fazari B, Hallenberger A, Bergmann AK, Niggetiedt E, Güven FD, Tundo-Lavalle F, Purath FFA, Bochinsky K, Gremer L, Willbold D, von Gall C, Ali AAH. Loss of Bmal1 impairs the glutamatergic light input to the SCN in mice. Front Cell Neurosci 2025; 19:1538985. [PMID: 40083633 PMCID: PMC11903712 DOI: 10.3389/fncel.2025.1538985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction Glutamate represents the dominant neurotransmitter that conveys the light information to the brain, including the suprachiasmatic nucleus (SCN), the central pacemaker for the circadian system. The neuronal and astrocytic glutamate transporters are crucial for maintaining efficient glutamatergic signaling. In the SCN, glutamatergic nerve terminals from the retina terminate on vasoactive intestinal polypeptide (VIP) neurons, which are essential for circadian functions. To date, little is known about the role of the core circadian clock gene, Bmal1, in glutamatergic neurotransmission of light signal to various brain regions. Methods The aim of this study was to further elucidate the role of Bmal1 in glutamatergic neurotransmission from the retina to the SCN. We therefore examined the spontaneous rhythmic locomotor activity, neuronal and glial glutamate transporters, as well as the ultrastructure of the synapse between the retinal ganglion cells (RGCs) and the SCN in adult male Bmal1-/- mice. Results We found that the deletion of Bmal1 affects the light-mediated behavior in mice, decreases the retinal thickness and affects the vesicular glutamate transporters (vGLUT1, 2) in the retina. Within the SCN, the immunoreaction of vGLUT1, 2, glial glutamate transporters (GLAST) and VIP was decreased while the glutamate concentration was elevated. At the ultrastructure level, the presynaptic terminals were enlarged and the distance between the synaptic vesicles and the synaptic cleft was increased, indicative of a decrease in the readily releasable pool at the excitatory synapses in Bmal1-/-. Conclusion Our data suggests that Bmal1 deletion affects the glutamate transmission in the retina and the SCN and affects the behavioral responses to light.
Collapse
Affiliation(s)
- Hüseyin Korkmaz
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Max Anstötz
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Tim Wellinghof
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Benedetta Fazari
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Angelika Hallenberger
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Ann Kathrin Bergmann
- Core Facility for Electron Microscopy, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Elena Niggetiedt
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Fatma Delâl Güven
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Federica Tundo-Lavalle
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Fathima Faiba A. Purath
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Kevin Bochinsky
- Jülich Research Center, Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Jülich, Germany
| | - Lothar Gremer
- Jülich Research Center, Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Jülich, Germany
- Institute of Physical Biology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Dieter Willbold
- Jülich Research Center, Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Jülich, Germany
- Institute of Physical Biology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Charlotte von Gall
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
| | - Amira A. H. Ali
- Faculty of Medicine, Institute of Anatomy II, Heinrich Heine University, Düsseldorf, Germany
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
26
|
Peng J, Huang X, Liu P, Hu Y, Kang L. SCH58261 effectively prevents the reduction in excitability of striatal MSNs in mice following 20 h of sleep deprivation. Purinergic Signal 2025:10.1007/s11302-025-10072-z. [PMID: 39969768 DOI: 10.1007/s11302-025-10072-z] [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: 06/30/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
Adenosine, a sleep-associated neuromodulator, is crucial in various physiological and pathological processes. Previous studies have demonstrated that sleep deprivation (SD) alters striatal neuronal activity. In this study, we used in vitro electrophysiological recordings to investigate the effects of 20 h of SD on the neuronal excitability of mouse dorsal striatal medium spiny neurons (MSNs). Our findings revealed that SD resulted in altered action potential (AP) discharge properties and reduced neuronal excitability compared to the control group. Importantly, these changes were partially offset by the prophylactic injection of the A2A receptor (A2AR) antagonist SCH58261. Additionally, 20 h of SD caused a decrease in the amplitude and an increase in the interval of spontaneous excitatory postsynaptic currents (sEPSCs) compared to control. However, the prophylactic injection of the A2AR antagonism shortened the sEPSC interval, while the A1 receptor (A1R) antagonist DPCPX not only shortened the interval but also further reduced the amplitude of sEPSCs. Thus, it can be concluded that SCH58261 effectively prevents the reduction in excitability of striatal MSNs in mice following 20 h of sleep deprivation, whereas DPCPX does not.
Collapse
Affiliation(s)
- Jin Peng
- College of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China
| | - Xinyu Huang
- College of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China
| | - Peijie Liu
- College of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China
| | - Yushi Hu
- College of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China
| | - Liang Kang
- Institute of Sport Medicine and Health, Chengdu Sport University, Chengdu, 610041, China.
| |
Collapse
|
27
|
Antonioni A, Di Lorenzo F. Topic Issue: "Translational Advances in Neurodegenerative Dementias". Neurol Int 2025; 17:31. [PMID: 39997662 PMCID: PMC11858273 DOI: 10.3390/neurolint17020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Neurodegenerative dementia, a collective term for a range of disorders characterized by progressive cognitive and functional decline, is an urgent challenge in global healthcare [...].
Collapse
Affiliation(s)
- Annibale Antonioni
- Doctoral Program in Translational Neurosciences and Neurotechnologies, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Francesco Di Lorenzo
- Experimental Neuropsychophysiology Lab, Department of Clinical and Behavioural Neurology, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
| |
Collapse
|
28
|
Fu C, Sun L, Feng C, Zhou T, Bi Y. A prognostic model of lung adenocarcinoma constructed based on circadian rhythm genes and its potential clinical significance. Front Oncol 2025; 15:1464578. [PMID: 40040723 PMCID: PMC11876053 DOI: 10.3389/fonc.2025.1464578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a common pathological category of lung cancer. Circadian rhythm (CR) disruption has been demonstrated to impact on lung tumorigenesis in mouse models. The aim of this study was to mine genes relevant to CR in LUAD and construct a corresponding risk model. Methods CRRGs from GSEA-MsigDB were filtered by overlapping DEGs in LUAD and NC specimens, two clusters with survival and clinical discrepancies, and CRRGs. Cox regression analysis (univariate and multivariate) was used to establish a CR-relevant risk model, which was validated in both the training and validation sets. Differences in immune infiltration, immunotherapy, and drug sensitivity between subgroups were explored. Prognostic gene expression was tested in clinical cancer and paracancer tissue samples using RT-qPCR. Results A grand total of two prognostic genes (CDK1 and HLA-DMA) related to CR were screened. The AUC values of a CR-relevant risk model in predicting 1/3/5-years survival in LUAD patients were greater than 0.6, indicating that the efficiency of the model was decent. Then, the results of CIBERSORT demonstrated noticeable differences in the tumor microenvironment between CR-relevant high- and low-risk subgroups. In addition, the CR-relevant risk score could be performed to estimate the effectiveness of immunotherapy in LUAD patients. The sensitivity of three common drugs (homoharringtonine, lapatinib, and palbociclib) in LUAD could be evaluated by the CR-relevant risk model. Ultimately, the experimental results confirmed that the expression trends of CDK1 and HLA-DMA in our collected clinical samples were in line with the expression trends in the TCGA-LUAD dataset. Conclusion In conclusion, a CR-relevant risk model based on CDK1 and HLA-DMA was constructed by using bioinformatics analysis, which might supply a new insight into the improved prognosis of LUAD.
Collapse
Affiliation(s)
- Cong Fu
- Department of Oncology, Changzhou Cancer (Fourth People’s) Hospital, Changzhou, China
| | - Lin Sun
- Department of Oncology, Affiliated Hospital of Soochow University, Changzhou, China
| | - Cuncheng Feng
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People’s Hospital, Changzhou, China
| | - Tong Zhou
- Department of Oncology, Changzhou Cancer (Fourth People’s) Hospital, Changzhou, China
| | - Yanzhi Bi
- Department of Oncology, Changzhou Cancer (Fourth People’s) Hospital, Changzhou, China
| |
Collapse
|
29
|
Kim S, Kang SW, Kim SE, Kim HJ, Kim SA, Lee YW, Kim EY, Shin C, Lee HW. Genome-wide identification and functional validation of the WW domain containing oxidoreductase gene associated with sleep duration. Sci Rep 2025; 15:5552. [PMID: 39952983 PMCID: PMC11828923 DOI: 10.1038/s41598-024-81158-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 11/25/2024] [Indexed: 02/17/2025] Open
Abstract
Individual differences in sleep duration have been reported, and genetic components of sleep duration have been identified showing various heritability. To identify genetic variants that contribute to sleep duration, we conducted a human genome-wide identification on sleep duration and performed confirmatory experiments using a Drosophila model. Genome-wide association study in human was analyzed to determine the association of the genetic variants with self-aware sleep duration from two community-based cohort, Ansan (cohort 1, n = 4635) and Ansung (cohort 2, n = 4205), recruited from the Korean Genome and Epidemiology Study. Individual single nucleotide variants (rs16948804 and rs4887991) in the WW domain containing oxidoreductase (WWOX) gene were associated with self-aware sleep duration in human (p-values, 1.11 × 10- 7 and 2.05 × 10- 7, retrospectively). To examine the functional relevance of the WWOX gene identified in the genome-wide association study, we analyzed the sleep duration of Drosophila loss-of-function mutants. The deletion of Wwox in flies reduced sleep duration and quality with average bout length during daytime and increased night-time sleep duration (all of p-values < 0.01). Our findings suggested that WWOX expression is associated with sleep duration in both humans and Drosophila and genetic factors play a role in inter-individual variability in sleep characteristics.
Collapse
Affiliation(s)
- Soriul Kim
- Department of Paramedicine, Seowon University, Cheongju, South Korea
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul, South Korea
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - So Who Kang
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Song E Kim
- Departments of Neurology and Medical Science, Computational Medicine, System Health Science and Engineering and Artificial Intelligence Graduate Programs, Ewha Womans University School of Medicine and Ewha Medical Research Institute, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, South Korea
| | - Hyeon Jin Kim
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul, South Korea
- Department of Neurology, Asan Medical Center, Seoul, South Korea
| | - Sol Ah Kim
- Departments of Neurology and Medical Science, Computational Medicine, System Health Science and Engineering and Artificial Intelligence Graduate Programs, Ewha Womans University School of Medicine and Ewha Medical Research Institute, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, South Korea
| | - Yae Won Lee
- Departments of Neurology and Medical Science, Computational Medicine, System Health Science and Engineering and Artificial Intelligence Graduate Programs, Ewha Womans University School of Medicine and Ewha Medical Research Institute, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, South Korea
| | - Eun Young Kim
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Chol Shin
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul, South Korea.
- Institute of Human Genomic Study, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Hyang Woon Lee
- Departments of Neurology and Medical Science, Computational Medicine, System Health Science and Engineering and Artificial Intelligence Graduate Programs, Ewha Womans University School of Medicine and Ewha Medical Research Institute, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, South Korea.
- Computational Medicine, System Health Science and Engineering, Ewha Womans University, Seoul, South Korea.
- Artificial Intelligence Convergence Graduate Programs, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
30
|
Arora S, Houdek P, Čajka T, Dočkal T, Sládek M, Sumová A. Chronodisruption that dampens output of the central clock abolishes rhythms in metabolome profiles and elevates acylcarnitine levels in the liver of female rats. Acta Physiol (Oxf) 2025; 241:e14278. [PMID: 39801395 PMCID: PMC11726269 DOI: 10.1111/apha.14278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/02/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
AIM Exposure to light at night and meal time misaligned with the light/dark (LD) cycle-typical features of daily life in modern 24/7 society-are associated with negative effects on health. To understand the mechanism, we developed a novel protocol of complex chronodisruption (CD) in which we exposed female rats to four weekly cycles consisting of 5-day intervals of constant light and 2-day intervals of food access restricted to the light phase of the 12:12 LD cycle. METHODS We examined the effects of CD on behavior, estrous cycle, sleep patterns, glucose homeostasis and profiles of clock- and metabolism-related gene expression (using RT qPCR) and liver metabolome and lipidome (using untargeted metabolomic and lipidomic profiling). RESULTS CD attenuated the rhythmic output of the central clock in the suprachiasmatic nucleus via Prok2 signaling, thereby disrupting locomotor activity, the estrous cycle, sleep patterns, and mutual phase relationship between the central and peripheral clocks. In the periphery, CD abolished Per1,2 expression rhythms in peripheral tissues (liver, pancreas, colon) and worsened glucose homeostasis. In the liver, it impaired the expression of NAD+, lipid, and cholesterol metabolism genes and abolished most of the high-amplitude rhythms of lipids and polar metabolites. Interestingly, CD abolished the circadian rhythm of Cpt1a expression and increased the levels of long-chain acylcarnitines (ACar 18:2, ACar 16:0), indicating enhanced fatty acid oxidation in mitochondria. CONCLUSION Our data show the widespread effects of CD on metabolism and point to ACars as biomarkers for CD due to misaligned sleep and feeding patterns.
Collapse
Affiliation(s)
- Shiyana Arora
- Laboratory of Biological RhythmsInstitute of Physiology of the Czech Academy of SciencesPragueCzech Republic
| | - Pavel Houdek
- Laboratory of Biological RhythmsInstitute of Physiology of the Czech Academy of SciencesPragueCzech Republic
| | - Tomáš Čajka
- Laboratory of Translational MetabolismInstitute of Physiology of the Czech Academy of SciencesPragueCzech Republic
| | - Tereza Dočkal
- Laboratory of Biological RhythmsInstitute of Physiology of the Czech Academy of SciencesPragueCzech Republic
| | - Martin Sládek
- Laboratory of Biological RhythmsInstitute of Physiology of the Czech Academy of SciencesPragueCzech Republic
| | - Alena Sumová
- Laboratory of Biological RhythmsInstitute of Physiology of the Czech Academy of SciencesPragueCzech Republic
| |
Collapse
|
31
|
Lin S, Gao M, Zhang J, Wu Y, Yu T, Peng Y, Jia Y, Zou H, Lu L, Li D, Ma Y. Sleep onset time as a mediator in the association between screen exposure and aging: a cross-sectional study. GeroScience 2025; 47:1239-1249. [PMID: 39190220 PMCID: PMC11872958 DOI: 10.1007/s11357-024-01321-x] [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: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024] Open
Abstract
Excessive screen exposure has become a significant health concern. This study investigates the impact of screen time on aging in middle-aged and elderly populations. Healthy working adults over 45 years old in Shanghai, China, underwent general and ocular examinations. Questionnaires collected demographics, medical history, and screen exposure details. Aging was assessed using the retinal age gap, defined as the difference between the retinal age predicted by deep learning algorithms based on fundus images and chronological age. Pathway analysis tested the mediation effect of sleep duration and onset time on the relationship between screen usage and retinal age gap. The retinal age gap increased with longer screen exposure, from 0.49 ± 3.51 years in the lowest tertile to 5.13 ± 4.96 years in the highest tertile (Jonckheere-Terpstra test, p < 0.001). Each additional hour of screen exposure accelerated the retinal age gap by 0.087 years (95% CI, 0.027, 0.148, p = 0.005) in the fully adjusted linear model. Sleep onset time mediated the impact of screen usage on the retinal age gap (indirect effect, β = 0.11; 95% CI 0.04-0.24). The impact of screen usage in a light-off environment on the retinal age gap was fully mediated by sleep onset time (indirect effect, β = 0.22; 95% CI 0.07-0.38), with the proportion being 100%. Our study identified a correlation between excessive screen time and a wider retinal age gap in middle-aged and elderly individuals, likely due to delayed sleep onset. To mitigate the adverse effects on the retina and aging, it is important to limit screen usage and avoid screens before bedtime.
Collapse
Affiliation(s)
- Senlin Lin
- Shanghai Eye Diseases Prevention &Treatment Center/ Shanghai Eye Hospital, School of Medicine, Tongji University, No. 1440, Hongqiao Road, Shanghai, 200336, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Engineering Research Center of Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Meng Gao
- Sijing Community Health Service Center, Shanghai, China
| | - Juzhao Zhang
- Shanghai Eye Diseases Prevention &Treatment Center/ Shanghai Eye Hospital, School of Medicine, Tongji University, No. 1440, Hongqiao Road, Shanghai, 200336, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Engineering Research Center of Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yuting Wu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tao Yu
- Shanghai Eye Diseases Prevention &Treatment Center/ Shanghai Eye Hospital, School of Medicine, Tongji University, No. 1440, Hongqiao Road, Shanghai, 200336, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Engineering Research Center of Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yajun Peng
- Shanghai Eye Diseases Prevention &Treatment Center/ Shanghai Eye Hospital, School of Medicine, Tongji University, No. 1440, Hongqiao Road, Shanghai, 200336, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Engineering Research Center of Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yingnan Jia
- Key Lab of Public Health Safety of the Ministry of Education, School of Public Health, Fudan University, 130 Dongan Road, Shanghai, 200032, China
- Health Communication Institute, Fudan University, Shanghai, 200032, China
| | - Haidong Zou
- Shanghai Eye Diseases Prevention &Treatment Center/ Shanghai Eye Hospital, School of Medicine, Tongji University, No. 1440, Hongqiao Road, Shanghai, 200336, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Engineering Research Center of Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, China
| | - Lina Lu
- Shanghai Eye Diseases Prevention &Treatment Center/ Shanghai Eye Hospital, School of Medicine, Tongji University, No. 1440, Hongqiao Road, Shanghai, 200336, China.
- National Clinical Research Center for Eye Diseases, Shanghai, China.
- Shanghai Engineering Research Center of Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.
| | - Deshang Li
- Shihudang Community Health Service Center, No. 1 to 5, Lane 50, Yanshou Road, Shanghai, China.
| | - Yingyan Ma
- Shanghai Eye Diseases Prevention &Treatment Center/ Shanghai Eye Hospital, School of Medicine, Tongji University, No. 1440, Hongqiao Road, Shanghai, 200336, China.
- National Clinical Research Center for Eye Diseases, Shanghai, China.
- Shanghai Engineering Research Center of Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, China.
| |
Collapse
|
32
|
Canonichesi J, Bellingacci L, Rivelli F, Tozzi A. Enhancing sleep quality in synucleinopathies through physical exercise. Front Cell Neurosci 2025; 19:1515922. [PMID: 39959465 PMCID: PMC11825755 DOI: 10.3389/fncel.2025.1515922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/06/2025] [Indexed: 02/18/2025] Open
Abstract
During sleep, several crucial processes for brain homeostasis occur, including the rearrangement of synaptic connections, which is essential for memory formation and updating. Sleep also facilitates the removal of neurotoxic waste products, the accumulation of which plays a key role in neurodegeneration. Various neural components and environmental factors regulate and influence the physiological transition between wakefulness and sleep. Disruptions in this complex system form the basis of sleep disorders, as commonly observed in synucleinopathies. Synucleinopathies are neurodegenerative disorders characterized by abnormal build-up of α-synuclein protein aggregates in the brain. This accumulation in different brain regions leads to a spectrum of clinical manifestations, including hypokinesia, cognitive impairment, psychiatric symptoms, and neurovegetative disturbances. Sleep disorders are highly prevalent in individuals with synucleinopathies, and they not only affect the overall well-being of patients but also directly contribute to disease severity and progression. Therefore, it is crucial to develop effective therapeutic strategies to improve sleep quality in these patients. Adequate sleep is vital for brain health, and the role of synucleinopathies in disrupting sleep patterns must be taken into account. In this context, it is essential to explore the role of physical exercise as a potential non-pharmacological intervention to manage sleep disorders in individuals with synucleinopathies. The current evidence on the efficacy of exercise programs to enhance sleep quality in this patient population is discussed.
Collapse
Affiliation(s)
| | | | | | - Alessandro Tozzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
33
|
Ishikawa H, Hoshino T, Hamanaka G, Mandeville ET, Guo S, Kimura S, Fukuda N, Li W, Shindo A, Sakadzic S, Harrington ME, Lo EH, Arai K. Effects of aging on diurnal transcriptome change in the mouse corpus callosum. iScience 2025; 28:111556. [PMID: 39845418 PMCID: PMC11750567 DOI: 10.1016/j.isci.2024.111556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/06/2024] [Accepted: 12/05/2024] [Indexed: 01/24/2025] Open
Abstract
The corpus callosum, a major white matter region central to cognitive function, is vulnerable to aging. Using zeitgeber time (ZT) aligned with environmental light/dark cycles, we investigated temporal gene expression patterns in the corpus callosum of young (5-month-old) and aged (24-month-old) mice using RNA-seq. Comparative analysis revealed more differentially expressed genes across ZT pairs in young mice than aged mice. In addition, complement pathway genes, including C4b, C3, C1qa, C1qb, and C1qc, were consistently upregulated in aged mice regardless of ZT. Furthermore, genes such as Etnppl, Tinagl1, Hspa12b, Ppp1r3c, Thbd, Pla2g3, and Tsc22d3 exhibited ZT-dependent rhythmicity in young mice, but their rhythmic patterns were altered with age. This study provides an important dataset of the interplay between aging, diurnal rhythms, and gene expression in the corpus callosum, highlighting potential molecular mechanisms mediating white matter aging. Further investigation is warranted to dissect these gene's specific roles in neurological health during aging.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Tomonori Hoshino
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Emiri T. Mandeville
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Shuzhen Guo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Shintaro Kimura
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Norito Fukuda
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Wenlu Li
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Sava Sakadzic
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | - Eng H. Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
34
|
Oladapo A, Deshetty UM, Callen S, Buch S, Periyasamy P. Single-Cell RNA-Seq Uncovers Robust Glial Cell Transcriptional Changes in Methamphetamine-Administered Mice. Int J Mol Sci 2025; 26:649. [PMID: 39859365 PMCID: PMC11766323 DOI: 10.3390/ijms26020649] [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: 12/15/2024] [Revised: 01/07/2025] [Accepted: 01/12/2025] [Indexed: 01/30/2025] Open
Abstract
Methamphetamine is a highly addictive stimulant known to cause neurotoxicity, cognitive deficits, and immune dysregulation in the brain. Despite significant research, the molecular mechanisms driving methamphetamine-induced neurotoxicity and glial cell dysfunction remain poorly understood. This study investigates how methamphetamine disrupts glial cell function and contributes to neurodevelopmental and neurodegenerative processes. Using single-cell RNA sequencing (scRNA-seq), we analyzed the transcriptomes of 4000 glial cell-associated genes from the cortical regions of mice chronically administered methamphetamine. Methamphetamine exposure altered the key pathways in astrocytes, including the circadian rhythm and cAMP signaling; in microglia, affecting autophagy, ubiquitin-mediated proteolysis, and mitophagy; and in oligodendrocytes, disrupting lysosomal function, cytoskeletal regulation, and protein processing. Notably, several transcription factors, such as Zbtb16, Hif3a, Foxo1, and Klf9, were significantly dysregulated in the glial cells. These findings reveal profound methamphetamine-induced changes in the glial transcriptomes, particularly in the cortical regions, highlighting potential molecular pathways and transcription factors as targets for therapeutic intervention. This study provides novel insights into the glial-mediated mechanisms of methamphetamine toxicity, contributing to our understanding of its effects on the central nervous system and laying the groundwork for future strategies to mitigate its neurotoxic consequences.
Collapse
Affiliation(s)
| | | | | | | | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.O.); (U.M.D.); (S.C.); (S.B.)
| |
Collapse
|
35
|
Crespo MT, Trebucq LL, Senna CA, Hokama G, Paladino N, Agostino PV, Chiesa JJ. Circadian disruption of feeding-fasting rhythm and its consequences for metabolic, immune, cancer, and cognitive processes. Biomed J 2025:100827. [PMID: 39756653 DOI: 10.1016/j.bj.2025.100827] [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: 06/28/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025] Open
Abstract
The circadian system is composed by a central hypothalamic clock at the suprachiasmatic nuclei (SCN) that communicates with peripheral circadian oscillators for daily coordination of behavior and physiology. The SCN entrain to the environmental 24-h light-dark (LD) cycle and drive daily rhythms of internal synchronizers such as core body temperature, hypothalamic-hypophysary hormones, sympathetic/parasympathetic activity, as well as behavioral and feeding-fasting rhythms, which supply signals setting core molecular clocks at central and peripheral tissues. Steady phase relationships between the SCN and peripheral oscillators keep homeostatic processes such as microbiota/microbiome composition/activity, metabolic supply/demand, energy balance, immunoinflammatory process, sleep amount and quality, psychophysiological stress, etc. Indeed, the risk of health alterations increase when these phase relationships are chronically changed prompting circadian disruption (CD), as occurring after sudden LD cycle changes (so-called jet-lag), or due to changes of activity/feeding-rest/fasting rhythm with respect to LD cycles (as humans subjected to nightwork, or restricting food access at rest in mice). Typical pathologies observed in animal models of CD and epidemiological studies include metabolic syndrome, type-2 diabetes, obesity, chronic inflammation, cancer, sleep disruption, decrease in physical and cognitive performance, and mood, among others. The present review discusses different aspects of such physiological dysregulations observed in animal models of CD having altered feeding-fasting rhythms, with potential translation to human health.
Collapse
Affiliation(s)
- Manuel Tomás Crespo
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Laura Lucía Trebucq
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Camila Agustina Senna
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Guido Hokama
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Natalia Paladino
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Patricia Verónica Agostino
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Juan José Chiesa
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina.
| |
Collapse
|
36
|
Kollarik S, Bimbiryte D, Sethi A, Dias I, Moreira CG, Noain D. Pharmacological enhancement of slow-wave activity at an early disease stage improves cognition and reduces amyloid pathology in a mouse model of Alzheimer's disease. Front Aging Neurosci 2025; 16:1519225. [PMID: 39831085 PMCID: PMC11739298 DOI: 10.3389/fnagi.2024.1519225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Improving sleep in murine Alzheimer's disease (AD) is associated with reduced brain amyloidosis. However, the window of opportunity for successful sleep-targeted interventions, regarding the reduction in pathological hallmarks and related cognitive performance, remains poorly characterized. Methods Here, we enhanced slow-wave activity (SWA) during sleep via sodium oxybate (SO) oral administration for 2 weeks at early (6 months old) or moderately late (11 months old) disease stages in Tg2576 mice and evaluated resulting neuropathology and behavioral performance. Results We observed that the cognitive performance of 6-month-old Tg2576 mice significantly improved upon SO treatment, whereas no change was observed in 11-month-old mice. Histochemical assessment of amyloid plaques demonstrated that SO-treated 11-month-old Tg2576 mice had significantly less plaque burden than placebo-treated ones, whereas ELISA of insoluble protein fractions from brains of 6-month-old Tg2576 mice indicated lower Aβ-42/Aβ-40 ratio in SO-treated group vs. placebo-treated controls. Discussion Altogether, our results suggest that SWA-dependent reduction in brain amyloidosis leads to alleviated behavioral impairment in Tg2576 mice only if administered early in the disease course, potentially highlighting the key importance of early sleep-based interventions in clinical cohorts.
Collapse
Affiliation(s)
- Sedef Kollarik
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich (ZNZ), Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Dorita Bimbiryte
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Aakriti Sethi
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Inês Dias
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich (ZNZ), Zurich, Switzerland
- D-HEST, ETHZurich, Zurich, Switzerland
| | - Carlos G. Moreira
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich (ZNZ), Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
- University Center of Competence Sleep and Health Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Rothwell ES, Viechweg SS, Prokai L, Mong JA, Lacreuse A. Oral administration of ethinyl estradiol and the brain-selective estrogen prodrug DHED in a female common marmoset model of menopause: Effects on cognition, thermoregulation, and sleep. Horm Behav 2025; 167:105670. [PMID: 39721460 DOI: 10.1016/j.yhbeh.2024.105670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/15/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024]
Abstract
Menopausal symptoms of sleep disturbances, cognitive deficits, and hot flashes are understudied, in part due to the lack of animal models in which they co-occur. Common marmosets (Callithrix jacchus) are valuable nonhuman primates for studying these symptoms, and we examined changes in cognition (reversal learning), sleep (48 h/wk of sleep recorded by telemetry), and thermoregulation (nose temperature in response to mild external warming) in middle-aged, surgically-induced menopausal marmosets studied at baseline, during 3-week phases of ethinyl estradiol (EE2, 4 μg/kg/day, p.o.) treatment and after EE2 withdrawal. We also assessed a brain-selective hormonal therapy devoid of estrogenic effects in peripheral tissues on the same measures (cognition, sleep, thermoregulation) after treatment with the estrogen prodrug 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED, 100 μg/kg/day, p.o) and DHED withdrawal. Reversal learning performance was improved with EE2 or DHED treatment relative to phases without hormone administration, as indicated by a faster reversal of the stimulus/reward contingencies. Both EE2 and DHED increased non-REM sleep and reduced nighttime awakenings relative to baseline, but to the detriment of REM sleep which was highest at baseline. Nasal temperature in response to mild external warming was highest, and overnight core body temperature lowest, in the DHED treatment phase compared to both the EE2 and baseline phases. These results suggest that low dose estradiol, delivered either peripherally or centrally via DHED, benefits selective aspects of cognition and sleep in a marmoset menopause model. DHED appears a promising therapeutic candidate for alleviating the cognitive and sleep disruptions associated with estrogen deficiency in primates.
Collapse
Affiliation(s)
- Emily S Rothwell
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States of America.
| | - Shaun S Viechweg
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, United States of America
| | - Laszlo Prokai
- Department of Pharmacology and Neuroscience, The University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Jessica A Mong
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, United States of America
| | - Agnès Lacreuse
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States of America
| |
Collapse
|
38
|
Liu YJ, Swaab DF, Zhou JN. Sleep-wake modulation and pathogenesis of Alzheimer disease: Suggestions for postponement and treatment. HANDBOOK OF CLINICAL NEUROLOGY 2025; 206:211-229. [PMID: 39864928 DOI: 10.1016/b978-0-323-90918-1.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Sleep-wake disorders are recognized as one of the earliest symptoms of Alzheimer disease (AD). Accumulating evidence has highlighted a significant association between sleep-wake disorders and AD pathogenesis, suggesting that sleep-wake modulation could be a promising approach for postponing AD onset. The suprachiasmatic nucleus (SCN) and the pineal hormone melatonin are major central modulating components of the circadian rhythm system. Cerebrospinal fluid (CSF) melatonin levels are dramatically decreased in AD. Interestingly, the number of neurofibrillary tangles in the hippocampus, which is one of the two major neuropathologic AD biomarkers, increases in parallel with the decrease in CSF melatonin levels. Furthermore, a decrease in salivary melatonin levels in middle-aged persons is a significant risk factor for the onset of the early stages of AD. Moreover, the disappearance of rhythmic fluctuations in melatonin may be one of the best biomarkers for AD diagnosis. Light therapy combined with melatonin supplementation is the recommended first-line treatment for sleep-wake disorders in AD patients and may be beneficial for ameliorating cognitive impairment. Sleep-wake cycle modulation based on AD risk gene presence is a promising early intervention for AD onset postponement.
Collapse
Affiliation(s)
- Ya-Jing Liu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dick F Swaab
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Jiang-Ning Zhou
- Institute of Brain Science, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
39
|
Lee Y, English EL, Schwartzmann CM, Liu Y, Krueger JM. Sleep loss-induced oncogenic pathways are mediated via the neuron-specific interleukin-1 receptor accessory protein (AcPb). Brain Behav Immun 2025; 123:411-421. [PMID: 39343106 PMCID: PMC11624092 DOI: 10.1016/j.bbi.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/04/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024] Open
Abstract
Interleukin-1β (IL1), a pleiotropic cytokine, is involved in sleep regulation, tumor ontogeny, and immune responses. IL1 receptor adaptor proteins, including the IL1 receptor accessory protein (AcP), and its neuron-specific isoform, AcPb, are required for IL1 signaling. The AcPb isoform is resultant from alternate splicing of the AcP transcript. Our previous studies using AcPb null (AcPb-/-) mice characterized its participation in sleep regulation and emergent neuronal/glial network properties. Here, we investigated the impact of acute sleep disruption (SD) on brain cancer-related pathways in wild-type (WT) and AcPb-/- mice, employing RNA sequencing methods. In WT mice, SD increased AcPb mRNA levels, but not AcP mRNA, confirming prior similar work in rats. Transcriptome and pathway enrichment analyses demonstrated significant alterations in cancer, immune, and viral disease-related pathways in WT mice after SD, which were attenuated in AcPb-/- mice including multiple upregulated Src phosphorylation-signaling-dependent genes associated with cancer progression and metastasis. Our RNAseq findings, were analyzed within the context of The Cancer Genome Atlas Program (TCGA) data base; revealing an upregulation of sleep- and cancer-linked genes (e.g., IL-17B, IL-17RA, LCN2) across various tumors, including brain tumors, compared to normal tissues. Sleep-linked factors, identified through TCGA analyses, significantly impact patient prognosis and survival, particularly in low-grade glioma (LGG) and glioblastoma multiforme (GBM) patients. Overall, our findings suggest that SD promotes a pro-tumor environment through AcPb-modulated pathways.
Collapse
Affiliation(s)
- Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA 99202, USA; Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA.
| | - Erika L English
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA
| | - Catherine M Schwartzmann
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Yiyong Liu
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; Genomics Core, Washington State University, Spokane, WA, USA
| | - James M Krueger
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA; Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
40
|
Silvestri R, Guarnieri B. Advanced sleep phase syndrome: Role of genetics and aging. HANDBOOK OF CLINICAL NEUROLOGY 2025; 206:61-70. [PMID: 39864932 DOI: 10.1016/b978-0-323-90918-1.00005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Advanced sleep phase (ASP) is seldom brought to medical attention because many individuals easily adapt to their early chronotype, especially if it emerges before the age of 30 and is present in a first-degree relative. In this case, the disorder is considered familial (FASP) and is mostly discovered coincidentally in the presence of other sleep disorders, mainly obstructive sleep apnea syndrome (OSAS). The prevalence of FASP is currently estimated to be between 0.21% and 0.5%. Autosomal dominant mutations in circadian clock genes like PER2, CK1, PER3, CRY2, TIMELESS, and DEC2 have been linked to FASP, some with pleiotropic effects influencing other health aspects like migraine and depression. Early morning awakening is, instead, more common among older individuals, occurring in almost 4% of cases, without considering associated comorbidities. Advanced sleep-wake phase disorder (ASWPD) is characterized by a consistent and distressing anticipation of sleep-wake timing, affecting almost 1% of middle-aged individuals. On average, women have a shorter circadian period than men, making them more susceptible to ASWPD, albeit no significant gender discrepancies have been observed. Age-related alterations in circadian rhythms are exacerbated and compounded by neurodegenerative disorders, impacting the suprachiasmatic nucleus (SCN), sensitivity to light, and light responsiveness in those affected. Conflicting data has surfaced regarding the protective or detrimental effects of ASWPD in studies on aging, mild cognitive impairment (MCI), and diverse dementia conditions.
Collapse
Affiliation(s)
- Rosalia Silvestri
- Sleep Medicine Center, Department of Clinical and Experimental Medicine, AOU G. Martino, Messina, Italy
| | - Biancamaria Guarnieri
- Sleep Medicine Center, Department of Neurology, Villa Serena Hospital, Città S. Angelo, Pescara, Italy; Villaserena Research Foundation, Città S. Angelo, Pescara, Italy
| |
Collapse
|
41
|
Fu M, Lu S, Gong L, Zhou Y, Wei F, Duan Z, Xiang R, Gonzalez FJ, Li G. Intermittent fasting shifts the diurnal transcriptome atlas of transcription factors. Mol Cell Biochem 2025; 480:491-504. [PMID: 38528297 DOI: 10.1007/s11010-024-04928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/05/2024] [Indexed: 03/27/2024]
Abstract
Intermittent fasting remains a safe and effective strategy to ameliorate various age-related diseases, but its specific mechanisms are not fully understood. Considering that transcription factors (TFs) determine the response to environmental signals, here, we profiled the diurnal expression of 600 samples across four metabolic tissues sampled every 4 over 24 h from mice placed on five different feeding regimens to provide an atlas of TFs in biological space, time, and feeding regimen. Results showed that 1218 TFs exhibited tissue-specific and temporal expression profiles in ad libitum mice, of which 974 displayed significant oscillations at least in one tissue. Intermittent fasting triggered more than 90% (1161 in 1234) of TFs to oscillate somewhere in the body and repartitioned their tissue-specific expression. A single round of fasting generally promoted TF expression, especially in skeletal muscle and adipose tissues, while intermittent fasting mainly suppressed TF expression. Intermittent fasting down-regulated aging pathway and upregulated the pathway responsible for the inhibition of mammalian target of rapamycin (mTOR). Intermittent fasting shifts the diurnal transcriptome atlas of TFs, and mTOR inhibition may orchestrate intermittent fasting-induced health improvements. This atlas offers a reference and resource to understand how TFs and intermittent fasting may contribute to diurnal rhythm oscillation and bring about specific health benefits.
Collapse
Affiliation(s)
- Min Fu
- Department of Neurology, The Fourth Hospital of Changsha, Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China
| | - Siyu Lu
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Lijun Gong
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yiming Zhou
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Fang Wei
- Department of Neurology, The Fourth Hospital of Changsha, Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China.
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Zhigui Duan
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Rong Xiang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 41001, Hunan, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guolin Li
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China.
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
42
|
Hussain Y, Dar MI, Pan X. Circadian Influences on Brain Lipid Metabolism and Neurodegenerative Diseases. Metabolites 2024; 14:723. [PMID: 39728504 DOI: 10.3390/metabo14120723] [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: 11/18/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Circadian rhythms are intrinsic, 24 h cycles that regulate key physiological, mental, and behavioral processes, including sleep-wake cycles, hormone secretion, and metabolism. These rhythms are controlled by the brain's suprachiasmatic nucleus, which synchronizes with environmental signals, such as light and temperature, and consequently maintains alignment with the day-night cycle. Molecular feedback loops, driven by core circadian "clock genes", such as Clock, Bmal1, Per, and Cry, are essential for rhythmic gene expression; disruptions in these feedback loops are associated with various health issues. Dysregulated lipid metabolism in the brain has been implicated in the pathogenesis of neurological disorders by contributing to oxidative stress, neuroinflammation, and synaptic dysfunction, as observed in conditions such as Alzheimer's and Parkinson's diseases. Disruptions in circadian gene expression have been shown to perturb lipid regulatory mechanisms in the brain, thereby triggering neuroinflammatory responses and oxidative damage. This review synthesizes current insights into the interconnections between circadian rhythms and lipid metabolism, with a focus on their roles in neurological health and disease. It further examines how the desynchronization of circadian genes affects lipid metabolism and explores the potential mechanisms through which disrupted circadian signaling might contribute to the pathophysiology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yusuf Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| | - Mohammad Irfan Dar
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| | - Xiaoyue Pan
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| |
Collapse
|
43
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [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: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
44
|
Das S, Khan R, Banerjee S, Ray S, Ray S. Alterations in Circadian Rhythms, Sleep, and Physical Activity in COVID-19: Mechanisms, Interventions, and Lessons for the Future. Mol Neurobiol 2024; 61:10115-10137. [PMID: 38702566 DOI: 10.1007/s12035-024-04178-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/04/2024] [Indexed: 05/06/2024]
Abstract
Although the world is acquitting from the throes of COVID-19 and returning to the regularity of life, its effects on physical and mental health are prominently evident in the post-pandemic era. The pandemic subjected us to inadequate sleep and physical activities, stress, irregular eating patterns, and work hours beyond the regular rest-activity cycle. Thus, perturbing the synchrony of the regular circadian clock functions led to chronic psychiatric and neurological disorders and poor immunological response in several COVID-19 survivors. Understanding the links between the host immune system and viral replication machinery from a clock-infection biology perspective promises novel avenues of intervention. Behavioral improvements in our daily lifestyle can reduce the severity and expedite the convalescent stage of COVID-19 by maintaining consistent eating, sleep, and physical activity schedules. Including dietary supplements and nutraceuticals with prophylactic value aids in combating COVID-19, as their deficiency can lead to a higher risk of infection, vulnerability, and severity of COVID-19. Thus, besides developing therapeutic measures, perpetual healthy practices could also contribute to combating the upcoming pandemics. This review highlights the impact of the COVID-19 pandemic on biological rhythms, sleep-wake cycles, physical activities, and eating patterns and how those disruptions possibly contribute to the response, severity, and outcome of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sandip Das
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India
| | - Rajni Khan
- National Institute of Pharmaceutical Education and Research (NIPER) - Hajipur, Vaishali, Hajipur, 844102, Bihar, India
| | - Srishti Banerjee
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, 845401, India.
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Sandipan Ray
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India.
| |
Collapse
|
45
|
Day GS. Diagnosing Alzheimer Disease. Continuum (Minneap Minn) 2024; 30:1584-1613. [PMID: 39620836 DOI: 10.1212/con.0000000000001507] [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: 12/21/2024]
Abstract
OBJECTIVE This article reviews the current understanding of Alzheimer disease (AD), including the natural history, common risk factors, and expected progression of AD neuropathologic change so that neurologists can apply this knowledge to identify patients with symptoms, signs, and findings on common diagnostic tests consistent with AD. LATEST DEVELOPMENTS The advent of potential disease-modifying therapies emphasizes the need to develop and deploy a practical and efficient approach to diagnose patients with cognitive impairment due to AD. ESSENTIAL POINTS The accumulation and spread of cerebral amyloid plaques and tau tangles in patients with AD leads to synaptic dysfunction, neuronal loss, and the eventual emergence and progression of cognitive impairment. A pragmatic and organized approach is needed to recognize patients with symptomatic AD in clinical practice, stage the level of impairment, confirm the clinical diagnosis, and apply this information to advance therapeutic decision making.
Collapse
|
46
|
Ichim AM, Barzan H, Moca VV, Nagy-Dabacan A, Ciuparu A, Hapca A, Vervaeke K, Muresan RC. The gamma rhythm as a guardian of brain health. eLife 2024; 13:e100238. [PMID: 39565646 DOI: 10.7554/elife.100238] [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: 05/30/2024] [Accepted: 11/09/2024] [Indexed: 11/21/2024] Open
Abstract
Gamma oscillations in brain activity (30-150 Hz) have been studied for over 80 years. Although in the past three decades significant progress has been made to try to understand their functional role, a definitive answer regarding their causal implication in perception, cognition, and behavior still lies ahead of us. Here, we first review the basic neural mechanisms that give rise to gamma oscillations and then focus on two main pillars of exploration. The first pillar examines the major theories regarding their functional role in information processing in the brain, also highlighting critical viewpoints. The second pillar reviews a novel research direction that proposes a therapeutic role for gamma oscillations, namely the gamma entrainment using sensory stimulation (GENUS). We extensively discuss both the positive findings and the issues regarding reproducibility of GENUS. Going beyond the functional and therapeutic role of gamma, we propose a third pillar of exploration, where gamma, generated endogenously by cortical circuits, is essential for maintenance of healthy circuit function. We propose that four classes of interneurons, namely those expressing parvalbumin (PV), vasointestinal peptide (VIP), somatostatin (SST), and nitric oxide synthase (NOS) take advantage of endogenous gamma to perform active vasomotor control that maintains homeostasis in the neuronal tissue. According to this hypothesis, which we call GAMER (GAmma MEdiated ciRcuit maintenance), gamma oscillations act as a 'servicing' rhythm that enables efficient translation of neural activity into vascular responses that are essential for optimal neurometabolic processes. GAMER is an extension of GENUS, where endogenous rather than entrained gamma plays a fundamental role. Finally, we propose several critical experiments to test the GAMER hypothesis.
Collapse
Grants
- RO-NO-2019-0504 Unitatea Executiva pentru Finantarea Invatamantului Superior, a Cercetarii, Dezvoltarii si Inovarii
- ERA-NET-FLAG-ERA-ModelDXConsciousness Unitatea Executiva pentru Finantarea Invatamantului Superior, a Cercetarii, Dezvoltarii si Inovarii
- ERANET-NEURON-2-UnscrAMBLY Unitatea Executiva pentru Finantarea Invatamantului Superior, a Cercetarii, Dezvoltarii si Inovarii
- ERANET-FLAG-ERA-MONAD Unitatea Executiva pentru Finantarea Invatamantului Superior, a Cercetarii, Dezvoltarii si Inovarii
- ERANET-NEURON-2-IBRAA Unitatea Executiva pentru Finantarea Invatamantului Superior, a Cercetarii, Dezvoltarii si Inovarii
- ERANET-NEURON-2-RESIST-D Unitatea Executiva pentru Finantarea Invatamantului Superior, a Cercetarii, Dezvoltarii si Inovarii
- PN-IV-P8-8.1-PRE-HE-ORG-2024-0185 Unitatea Executiva pentru Finantarea Invatamantului Superior, a Cercetarii, Dezvoltarii si Inovarii
- 952096 NEUROTWIN European Commission
- INSPIRE POC 488/1/1/2014+/127725 Ministerul Investițiilor și Proiectelor Europene
Collapse
Affiliation(s)
- Ana Maria Ichim
- Transylvanian Institute of Neuroscience, Department of Experimental and Theoretical Neuroscience, Cluj-Napoca, Romania
- Preclinical MRI Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babeș-Bolyai University, Cluj-Napoca, Romania
| | - Harald Barzan
- Transylvanian Institute of Neuroscience, Department of Experimental and Theoretical Neuroscience, Cluj-Napoca, Romania
| | - Vasile Vlad Moca
- Transylvanian Institute of Neuroscience, Department of Experimental and Theoretical Neuroscience, Cluj-Napoca, Romania
| | - Adriana Nagy-Dabacan
- Transylvanian Institute of Neuroscience, Department of Experimental and Theoretical Neuroscience, Cluj-Napoca, Romania
| | - Andrei Ciuparu
- Transylvanian Institute of Neuroscience, Department of Experimental and Theoretical Neuroscience, Cluj-Napoca, Romania
| | - Adela Hapca
- Transylvanian Institute of Neuroscience, Department of Experimental and Theoretical Neuroscience, Cluj-Napoca, Romania
- Faculty of Biology and Geology, Babeș-Bolyai University, Cluj-Napoca, Romania
| | - Koen Vervaeke
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Raul Cristian Muresan
- Transylvanian Institute of Neuroscience, Department of Experimental and Theoretical Neuroscience, Cluj-Napoca, Romania
- STAR-UBB Institute, Babeș-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
47
|
Huang Y, Sun J, Li S, Shi Y, Yu L, Wu A, Wang X. Isoliquiritigenin mitigates intervertebral disc degeneration induced by oxidative stress and mitochondrial impairment through a PPARγ-dependent pathway. Free Radic Biol Med 2024; 225:98-111. [PMID: 39366471 DOI: 10.1016/j.freeradbiomed.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
OBJECTIVES Oxidative stress, mitochondrial dysfunction, and apoptosis play significant roles in the degradation of extracellular matrix (ECM) in nucleus pulposus cells (NPCs), ultimately contributing to intervertebral disc degeneration (IVDD). This study investigates the potential of isoliquiritigenin (ISL), a natural extract known for its antioxidant, anti-inflammatory, and anti-atherosclerotic properties, to alleviate IVDD. METHODS The viability of NPCs treated with ISL and tert-butyl hydroperoxide (TBHP) was assessed using the CCK-8 assay. Various techniques, including Western blot, qRT-PCR, immunofluorescence (IF), and immunohistochemistry, were employed to measure the expression of ECM components, oxidative stress markers, and apoptosis-related proteins. Mitochondrial function was evaluated through Western blot and IF analyses. Network pharmacology predicted ISL targets, and the expression levels of PPARγ were assessed using the aforementioned methods. The role of PPARγ in the therapeutic effects of ISL on IVDD was examined through siRNA knockdown. The therapeutic impact of ISL on puncture-induced IVDD in rats was evaluated using X-ray, MRI, and histological staining techniques. RESULTS In vitro, ISL reduced oxidative stress in NPCs, restored mitochondrial function, inhibited apoptosis, and improved the ECM phenotype. In vivo, ISL slowed the progression of IVDD in a rat model. Further analysis revealed that ISL enhances PPARγ activity and promotes its expression by direct binding, contributing to the delay of IVDD progression. CONCLUSION This study demonstrates that ISL effectively treats puncture-induced IVDD in rats by inhibiting oxidative stress, restoring mitochondrial function, and reducing NPC apoptosis through a PPARγ-dependent mechanism. By balancing ECM synthesis and degradation, ISL presents a novel therapeutic approach for IVDD and identifies a promising target for treatment.
Collapse
Affiliation(s)
- Yeheng Huang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Jing Sun
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Sunlong Li
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yifeng Shi
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Lianggao Yu
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Aimin Wu
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xiangyang Wang
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
48
|
Yao W, Hou X, Zhou H, You S, Lv T, Chen H, Yang Z, Chen C, Bai F. Associations between the multitrajectory neuroplasticity of neuronavigated rTMS-mediated angular gyrus networks and brain gene expression in AD spectrum patients with sleep disorders. Alzheimers Dement 2024; 20:7885-7901. [PMID: 39324544 PMCID: PMC11567849 DOI: 10.1002/alz.14255] [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: 04/24/2024] [Accepted: 08/18/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION The multifactorial influence of repetitive transcranial magnetic stimulation (rTMS) on neuroplasticity in neural networks is associated with improvements in cognitive dysfunction and sleep disorders. The mechanisms of rTMS and the transcriptional-neuronal correlation in Alzheimer's disease (AD) patients with sleep disorders have not been fully elucidated. METHODS Forty-six elderly participants with cognitive impairment (23 patients with low sleep quality and 23 patients with high sleep quality) underwent 4-week periods of neuronavigated rTMS of the angular gyrus and neuroimaging tests, and gene expression data for six post mortem brains were collected from another database. Transcription-neuroimaging association analysis was used to evaluate the effects on cognitive dysfunction and the underlying biological mechanisms involved. RESULTS Distinct variable neuroplasticity in the anterior and posterior angular gyrus networks was detected in the low sleep quality group. These interactions were associated with multiple gene pathways, and the comprehensive effects were associated with improvements in episodic memory. DISCUSSION Multitrajectory neuroplasticity is associated with complex biological mechanisms in AD-spectrum patients with sleep disorders. HIGHLIGHTS This was the first transcription-neuroimaging study to demonstrate that multitrajectory neuroplasticity in neural circuits was induced via neuronavigated rTMS, which was associated with complex gene expression in AD-spectrum patients with sleep disorders. The interactions between sleep quality and neuronavigated rTMS were coupled with multiple gene pathways and improvements in episodic memory. The present strategy for integrating neuroimaging, rTMS intervention, and genetic data provide a new approach to comprehending the biological mechanisms involved in AD.
Collapse
Affiliation(s)
- Weina Yao
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Xinle Hou
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Huijuan Zhou
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Shengqi You
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Tingyu Lv
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Haifeng Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Zhiyuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Chang Chen
- School of Elderly Care Services and ManagementNanjing University of Chinese MedicineNanjingChina
| | - Feng Bai
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Geriatric Medicine Center, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Institute of Geriatric MedicineMedical School of Nanjing UniversityNanjingChina
| |
Collapse
|
49
|
Tonon AC, Nexha A, Mendonça da Silva M, Gomes FA, Hidalgo MP, Frey BN. Sleep and circadian disruption in bipolar disorders: From psychopathology to digital phenotyping in clinical practice. Psychiatry Clin Neurosci 2024; 78:654-666. [PMID: 39210713 PMCID: PMC11804932 DOI: 10.1111/pcn.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Sleep and biological rhythms are integral to mood regulation across the lifespan, particularly in bipolar disorder (BD), where alterations in sleep phase, structure, and duration occur in all mood states. These disruptions are linked to poorer quality of life, heightened suicide risk, impaired cognitive function, and increased relapse rates. This review highlights the pathophysiology of sleep disturbances in BD and aims to consolidate understanding and clinical applications of these phenomena. It also summarizes the evolution of sleep and biological rhythms assessment methods, including ecological momentary assessment (EMA) and digital phenotyping. It underscores the importance of recognizing circadian rhythm involvement in mood regulation, suggesting potential therapeutic targets. Future research directions include elucidating circadian clock gene mechanisms, understanding environmental impacts on circadian rhythms, and investigating the bidirectional relationship between sleep disturbances and mood regulation in BD. Standardizing assessment methods and addressing privacy concerns related to EMA technology and digital phenotyping are essential for advancing research. Collaborative efforts are crucial for enhancing clinical applicability and understanding the broader implications of biological rhythms in BD diagnosis and treatment. Overall, recognizing the significance of sleep and biological rhythms in BD offers promise for improved outcomes through targeted interventions and a deeper understanding of the disorder's underlying mechanisms.
Collapse
Affiliation(s)
- André C. Tonon
- Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare HamiltonHamiltonOntarioCanada
- Department of Psychiatry and Behavioural NeurosciencesMcMaster UniversityHamiltonOntarioCanada
| | - Adile Nexha
- Department of Psychiatry and Behavioural NeurosciencesMcMaster UniversityHamiltonOntarioCanada
| | - Mariana Mendonça da Silva
- Laboratório de Cronobiologia e SonoPorto Alegre Clinicas Hospital, Federal University of Rio Grande do Sul (UFRGS)Porto AlegreBrazil
| | - Fabiano A. Gomes
- Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare HamiltonHamiltonOntarioCanada
- Department of Psychiatry and Behavioural NeurosciencesMcMaster UniversityHamiltonOntarioCanada
| | - Maria Paz Hidalgo
- Laboratório de Cronobiologia e SonoPorto Alegre Clinicas Hospital, Federal University of Rio Grande do Sul (UFRGS)Porto AlegreBrazil
- Graduate Program in Psychiatry and Behavioral SciencesFederal University of Rio Grande do Sul (UFRGS)Porto AlegreBrazil
| | - Benicio N. Frey
- Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare HamiltonHamiltonOntarioCanada
- Department of Psychiatry and Behavioural NeurosciencesMcMaster UniversityHamiltonOntarioCanada
| |
Collapse
|
50
|
Zhou Z, Zhang R, Zhang Y, Xu Y, Wang R, Chen S, Lv Y, Chen Y, Ren Y, Luo P, Cheng Q, Xu H, Weng S, Zuo A, Ba Y, Liu S, Han X, Liu Z. Circadian disruption in cancer hallmarks: Novel insight into the molecular mechanisms of tumorigenesis and cancer treatment. Cancer Lett 2024; 604:217273. [PMID: 39306230 DOI: 10.1016/j.canlet.2024.217273] [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: 06/27/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Circadian rhythms are 24-h rhythms governing temporal organization of behavior and physiology generated by molecular clocks composed of autoregulatory transcription-translation feedback loops (TTFLs). Disruption of circadian rhythms leads to a spectrum of pathologies, including cancer by triggering or being involved in different hallmarks. Clock control of phenotypic plasticity involved in tumorigenesis operates in aberrant dedifferentiating to progenitor-like cell states, generation of cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT) events. Circadian rhythms might act as candidates for regulatory mechanisms of cellular senescent and functional determinants of senescence-associated secretory phenotype (SASP). Reciprocal control between clock and epigenetics sheds light on post-transcriptional regulation of circadian rhythms and opens avenues for novel anti-cancer strategies. Additionally, disrupting circadian rhythms influences microbiota communities that could be associated with altered homeostasis contributing to cancer development. Herein, we summarize recent advances in support of the nexus between disruptions of circadian rhythms and cancer hallmarks of new dimensions, thus providing novel perspectives on potentially effective treatment approaches for cancer management.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruiqi Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruizhi Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yingying Lv
- Department of Pediatrics, The First Affliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Pediatrics, The Third Affliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yifeng Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|