1
|
Kovalyova Y, De Leon C, Krasowska-Zoladek A, Suon S, Wong J, Young S, Lee Heberling J, Price L, Berger R, Magliaro B, Cheng YS, Peier A, Rothman DM, Walji A, Smith S, Marcus J, Han X, Usenovic M. Promoting Secretion of Pathological Tau Species Using an Induced Proximity Platform That Engages the Autophagy Pathway. ACS Chem Neurosci 2025. [PMID: 40344401 DOI: 10.1021/acschemneuro.5c00161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
Abstract
Intracellular accumulation of aberrantly phosphorylated aggregated tau protein can contribute to neuronal dysfunction associated with many neurodegenerative diseases. Thus, removing such tau species is an attractive therapeutic hypothesis for these diseases. Targeted protein degradation (TPD) strategies leveraging the autophagy-lysosome pathway (ALP) are promising approaches to decrease protein aggregates by designating them for degradation. Here, we developed a novel heterobifunctional molecule, MRL828, combining a tau pathology-binding ligand and modified guanine moiety based on the autophagy-targeting chimaera technology to selectively designate aggregated tau proteins for clearance via the ALP. Surprisingly, the MRL828-dependent decrease in intracellular tau aggregates was dependent on the autophagosome, but not the lysosome. MRL828 treatment led to autophagosome-dependent secretion of oligomeric and phosphorylated tau species, suggesting a reduction of intracellular tau aggregates via secretory autophagy rather than degradation via the ALP. This work highlights a novel mechanism of action (MOA) of an ALP-based heterobifunctional molecule and a potential new strategy for the cellular removal of proteins of interest.
Collapse
Affiliation(s)
| | - Cesar De Leon
- Chemical Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Sokreine Suon
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jacky Wong
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Seth Young
- Chemical Biology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Laura Price
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Raphaëlle Berger
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Brian Magliaro
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yu-Shan Cheng
- Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Andrea Peier
- Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Deborah M Rothman
- Chemical Biology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Abbas Walji
- Discovery Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Sean Smith
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jacob Marcus
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Xiaoqing Han
- Chemical Biology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Marija Usenovic
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
2
|
Singh A, Kaakinen M, Elamaa H, Kiviniemi V, Eklund L. The glycosaminoglycan chains of perlecan regulate the perivascular fluid transport. Fluids Barriers CNS 2025; 22:48. [PMID: 40340918 PMCID: PMC12063283 DOI: 10.1186/s12987-025-00648-7] [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: 01/29/2025] [Accepted: 03/28/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND The perivascular conduct pathway that connects the cerebrospinal fluid spaces with the interstitial fluid in the parenchyma are of importance for solute clearance from the brain. In this pathway, the relatively wide perivascular space (PVS) surrounding the pial arteries provides a low-resistant passage while around the perforating arteries, the solute movement is along the basement membrane (BM), that prevents the free exchange of interstitial fluids and solutes. We hypothesize that this selectivity involves specific components of the vascular BM, which is mainly composed of type IV collagen (Col IV) and laminin networks interconnected by nidogens and heparan sulphate proteoglycans (HSPGs). Perlecan is the major HSPG in the BM that binds to Col IV and laminin via glycosaminoglycan (GAG) chains to form a molecular sieve. GAGs may also provide the charge selectivity required for filtration, and also a scaffold for amyloid-β (Aβ) aggregation. The purpose of this study was the functional characterization of perivascular fluid transport and brain clearance in mice lacking perlecan GAG chains. METHODS We generated a novel mouse line (Hspg2∆3∆91) lacking perlecan GAG side chains and investigated perivascular flow and brain clearance in these mice using intravital multiphoton and fluorescence recovery after photobleaching techniques, and functional assays with various tracers. Potentially deleterious effects on brain homeostasis were investigated using transcriptomic, proteomic and immunohistochemical methods. The Hspg2∆3∆91 mice were crossed with a 5xFAD line to examine the importance of GAGs in Aβ aggregation. RESULTS We observed a delayed inflow of CSF tracer into the Hspg2∆3∆91 brain with no changes in the clearance of parenchymal injected tracers. Quantification of the Aβ plaques revealed fewer and smaller plaques in the walls of the pial arteries at six months of age, but not in the brain parenchyma. Surprisingly, perlecan GAG deficiency had no severe deleterious effects on brain homeostasis in transcriptomic and proteomic analyses. CONCLUSIONS Potential brain clearance mechanisms are dependent on the flow through special ECM structures. BM is mainly known for its barrier function, whereas very little is known about how passage along the perivascular ECM is established. This study shows that the GAG composition of the BM affects the solute dynamics and Aβ deposition in the periarterial space.
Collapse
Affiliation(s)
- Abhishek Singh
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Mika Kaakinen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Harri Elamaa
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Vesa Kiviniemi
- Research Unit of Health Sciences and Technology (HST), Faculty of Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
- Oulu Functional Neuroimaging (OFNI), Diagnostic Imaging, Medical Research Center (MRC), Oulu University Hospital, Oulu, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
3
|
Guo J, Zhang Z, Meng X, Jing J, Hu Y, Yao Y, Ding L, Zheng L, Zhao X. Atrial fibrillation catheter ablation, brain glymphatic function, and cognitive performance. Eur Heart J 2025; 46:1733-1743. [PMID: 39981927 DOI: 10.1093/eurheartj/ehaf036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/21/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND AND AIMS It remains unknown whether the brain glymphatic system, which is driven by the heartbeat-driven pulsation of arteries and is responsible for cerebral waste clearance, is impaired in atrial fibrillation (AF) and mediates cognitive dysfunction related to AF. The aim of this study was to assess brain glymphatic alterations in AF, their role in cognitive function, and whether catheter ablation can improve glymphatic activity. METHODS In this case-control and prospective before-and-after study, patients with AF and healthy controls (HCs) were enrolled. Participants underwent brain magnetic resonance imaging and a comprehensive neuropsychological battery. Glymphatic activity was quantified by diffusion tensor image analysis along the perivascular space (DTI-ALPS) index. Magnetic resonance imaging was repeated after surgery in patients who underwent ablation. RESULTS Overall, 87 patients with AF and 44 HCs were enrolled. Compared with HCs, patients with AF had a lower ALPS index (P = .016). Nonparoxysmal AF patients showed lower ALPS index than both HCs (P = .002) and paroxysmal AF patients (P = .044). A lower ALPS index was associated with worse scores of Trail Making Test, Digit Symbol Substitution Test, Digit Span Test, and Stroop Colour and Word Test (all P < .05). Mediation analyses revealed that glymphatic activity was a mediator between AF and cognitive decline. Among the 50 patients who underwent ablation therapy, DTI-ALPS index was improved after surgery (P = .015). CONCLUSIONS Brain glymphatic function measured by DTI-ALPS index was impaired in patients with AF, mediates the association between AF and cognitive decline, and was improved after ablation therapy.
Collapse
Affiliation(s)
- Jiahuan Guo
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
| | - Zhe Zhang
- Tiantan Neuroimaging Center of Excellence, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
| | - Xu Meng
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Xicheng District, 100037 Beijing, China
| | - Jing Jing
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
- Tiantan Neuroimaging Center of Excellence, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
| | - Yiran Hu
- Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Xicheng District, 100037 Beijing, China
- Department of Cardiology and Macrovascular Disease, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
| | - Yan Yao
- Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Xicheng District, 100037 Beijing, China
| | - Ligang Ding
- Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Xicheng District, 100037 Beijing, China
| | - Lihui Zheng
- Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 North Lishi Road, Xicheng District, 100037 Beijing, China
| | - Xingquan Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, 119 South Fourth Ring West Road, Fengtai District, 100070 Beijing, China
| |
Collapse
|
4
|
Yang L, Gao Y, Lopes LS, Lian J, Fu W, Tan H, Yang S, Xie Z, Huang Y, Zhang J, Lu Y, Tang H, Xiong B, Wei X, Xie L, Peng Y, Liu X, Han H. Accelerated Molecular Transportation in the Brain Extracellular Space with 755-nm Light Attenuates Post-Stroke Cognitive Impairment in Rats. CYBORG AND BIONIC SYSTEMS 2025; 6:0262. [PMID: 40330544 PMCID: PMC12053100 DOI: 10.34133/cbsystems.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 05/08/2025] Open
Abstract
Ischemic stroke exacts a heavy toll in death and disability worldwide. After ischemic stroke, the accumulation of pathobiomolecules in the brain extracellular space (ECS) will exacerbate neurological damage and cognitive impairment. Photobiomodulation (PBM) has been demonstrated to improve cognitive function in Alzheimer's disease mouse models by accelerating molecular transportation in the brain ECS. This suggests that PBM may have a potential role in the accumulation of pathobiomolecules in the brain ECS following ischemic stroke. In this study, we developed a PBM therapy apparatus with custom parameters. By evaluating the treatment effect, we identified that 755 nm was the optimal light wavelength for ischemic stroke in rats with transient middle cerebral artery occlusion/reperfusion. Extracellular diffusion and interstitial fluid (ISF) drainage were measured using a tracer-based magnetic resonance imaging method. Our results showed that PBM accelerated molecular transportation in the brain ECS and ISF drainage, promoting the clearance of pro-inflammatory cytokines and reducing the deposition of pathological proteins. Consequently, the infarct volume decreased and neurological cognitive function was improved. Besides, the acceleration of ISF drainage was achieved by reducing expression and restoring polarization of aquaporin 4 (AQP4) in the peri-infarct area. In summary, we demonstrated that PBM could alleviate ischemia-reperfusion injury and prevent post-stroke cognitive impairment by accelerating molecular transportation in the brain ECS, paving a pathway for ischemic stroke treatment via the light-ECS interaction.
Collapse
Affiliation(s)
- Liu Yang
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Yajuan Gao
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Leonor Serrano Lopes
- Department of Nuclear Medicine, Inselspital, Bern University Hospital,
University of Bern, Bern 3010, Switzerland
- Graduate School for Cellular and Biomedical Sciences,
University of Bern, Bern 3012, Switzerland
| | - Jingge Lian
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Wanyi Fu
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
- Department of Electronic Engineering,
Tsinghua University, Beijing 100084, China
| | - Hanbo Tan
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Shuangfeng Yang
- Department of Radiology, Beijing Children’s Hospital,
Capital Medical University, National Center for Children’ Health, Beijing 100045, China
| | - Zhaoheng Xie
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Yixing Huang
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Jicong Zhang
- School of Biological Science and Medical Engineering,
Beihang University, Beijing 100191, China
| | - Yanye Lu
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Hao Tang
- School of Computer Science,
Peking University, Beijing 100871, China
| | - Bo Xiong
- School of Computer Science,
Peking University, Beijing 100871, China
| | - Xunbin Wei
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Lide Xie
- Chengde Medical University, Chengde, Hebei 067000, China
| | - Yun Peng
- Department of Radiology, Beijing Children’s Hospital,
Capital Medical University, National Center for Children’ Health, Beijing 100045, China
| | - Xinyu Liu
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Hongbin Han
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
- Chengde Medical University, Chengde, Hebei 067000, China
| |
Collapse
|
5
|
Sun Q, Liu J, Yang Z, Lei J, Li H, Wang Y. Glymphatic system dysfunction and its impact on seizure severity, cognitive function, and affective symptoms in patients with generalized tonic-clonic seizures alone. J Neurol Sci 2025; 473:123515. [PMID: 40349507 DOI: 10.1016/j.jns.2025.123515] [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: 11/08/2024] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Generalized tonic-clonic seizures alone (GTCS alone) represent a distinct idiopathic epilepsy syndrome. Glymphatic system (GS) dysfunction-a brain-wide perivascular clearance pathway-has been proposed as a contributing mechanism in epilepsy and its related comorbidities. OBJECTIVE To assess GS function in patients with GTCS alone using the Analysis Along the Perivascular Space (ALPS) index derived from diffusion spectrum imaging (DSI), and to explore its associations with clinical, cognitive, and emotional measures. METHODS A total of 101 patients with GTCS alone and 76 demographically matched healthy controls underwent DSI. The ALPS index was calculated and correlated with scores from standardized assessments, including the National Hospital Seizure Severity Scale (NHS3), Epileptic Discharge Index (EDI), Montreal Cognitive Assessment (MoCA), Kilifi Stigma Scale for Epilepsy (KSSE), and Hamilton Depression Rating Scale (HAMD). RESULTS Patients with GTCS alone had lower ALPS indices compared to healthy controls (1.43 vs. 1.52, p < 0.01). Among patients, the ALPS index positively correlated with MoCA scores (r = 0.30, p = 0.002) and negatively correlated with age (r = -0.22, p = 0.030), NHS3 (r = -0.27, p = 0.007), KSSE (r = -0.21, p = 0.038), and HAMD (r = -0.20, p = 0.042). The ALPS index was lower in patients with higher antiseizure medication loads (1.36 vs. 1.44, p = 0.042) and elevated EDI values (1.39 vs. 1.45, p = 0.039). CONCLUSIONS Glymphatic function is impaired in patients with GTCS alone, as indicated by reduced ALPS indices. These indices are associated with seizure severity, cognitive impairment, depressive symptoms, and perceived stigma. The ALPS index may serve as a noninvasive imaging biomarker of disease burden and a novel tool for understanding pathophysiological mechanisms in GTCS alone.
Collapse
Affiliation(s)
- Qibing Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinshuai Liu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zifan Yang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianxiang Lei
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hanli Li
- Department of Clinical Medicine, Anhui Medical College, Hefei, China
| | - Yu Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
6
|
Mrzyglod A, Mebrouk A, Bartkiewicz J, El Hachami H, Ryniejska M, de Tisi J, Coras R, Blumcke I, Xiao F, Miserocchi A, McEvoy A, Koepp M, Liu J, Thom M. Patterns of phosphorylated tau accumulation in a spectrum of acquired and developmental brain lesions associated with refractory epilepsy. Epilepsia 2025. [PMID: 40299318 DOI: 10.1111/epi.18418] [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: 12/17/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVE Phosphorylated tau (pTau) has been reported in surgical resections in refractory epilepsy. It is unclear whether this is activity-driven physiological pTau or signifies the advent of neurodegenerative cascades, relevant to memory decline. To date, primarily hippocampal sclerosis and focal cortical dysplasia (FCD) type II have been studied. We aimed to explore pTau in a range of acquired and developmental epileptogenic pathologies to assess its prevalence and identify potential drivers. METHOD A total of 104 cases were studied representing FCD IA (n = 11), FCD IIIA (n = 5), FCD IIIB (n = 6), cavernoma (n = 11), Sturge-Weber leptomeningeal angiomatosis (n = 10), meningioangiomatosis (n = 4), perinatal infarcts (n = 9), Rasmussen encephalitis (RE; n = 6), gray matter heterotopia (n = 6), old scars (n = 10), and temporal lobe encephaloceles (n = 7); we also included focal microinjuries following prior stereoelectroencephalography at different ages (n = 19; four in lesion-negative cases). pTau was evaluated with AT8 immunohistochemistry, with further multiplex panels of AT8 with other established pTau markers (AT100, AT180, PHF1, CP13), pS6, glial fibrillary acidic protein, reelin, calbindin, and Tbr1 in selected cases. Labeling in the lesion was compared with adjacent cortex and clinical factors such as epilepsy duration. RESULTS pTau was identified in low to moderate levels in 60% overall, mainly localized to the epileptogenic lesion and more frequent in vascular malformations (74%-100%). pTau was noted in the superficial cortex across pathologies including encephaloceles, associated with superficial gliosis. In perinatal infarcts, distinct pTau patterns were noted in the superficial ulegyric cortex and heterotopic neuronal islands. Glial pTau was rare, and FCD IA, FCD IIIA/B, and microinjuries were negative. Variable regional expression of AT8 and mTOR activation markers (pS6) was noted, including in one RE case. Higher pTau expression was associated with older age at surgery and at onset of epilepsy, suggesting additional age-related vulnerability. SIGNIFICANCE Our findings highlight localized and distinct patterns of pTau in some epilepsy pathologies. Plausible pathomechanisms include local vascular insufficiency, neuronal dysmaturation, and aging as well as seizure activity and provide direction for future exploration.
Collapse
Affiliation(s)
- Alicja Mrzyglod
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Anya Mebrouk
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Joanna Bartkiewicz
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Hanaa El Hachami
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Maritchka Ryniejska
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Jane de Tisi
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Roland Coras
- Institute of Neuropathology, University Hospitals Erlangen, Erlangen, Germany
| | - Ingmar Blumcke
- Institute of Neuropathology, University Hospitals Erlangen, Erlangen, Germany
| | - Fenglai Xiao
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Anna Miserocchi
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Andrew McEvoy
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Matthias Koepp
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Joan Liu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- School of Life Sciences, University of Westminster, London, UK
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- Division of Neuropathology, University college London Queen Square Institute of Neurology, London, UK
| |
Collapse
|
7
|
Berlind JE, Lai JD, Lie C, Vicente J, Lam K, Guo S, Chang J, Yu V, Ichida JK. KCTD20 suppression mitigates excitotoxicity in tauopathy patient organoids. Neuron 2025; 113:1169-1189.e7. [PMID: 40049159 PMCID: PMC12005969 DOI: 10.1016/j.neuron.2025.02.001] [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: 05/07/2024] [Revised: 12/13/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025]
Abstract
Excitotoxicity is a major pathologic mechanism in patients with tauopathy and other neurodegenerative diseases. However, the key neurotoxic drivers and the most effective strategies for mitigating these degenerative processes are unclear. Here, we show that glutamate treatment of induced pluripotent stem cell (iPSC)-derived cerebral organoids induces tau oligomerization and neurodegeneration and that these phenotypes are enhanced in organoids derived from tauopathy patients. Using a genome-wide CRISPR interference (CRISPRi) screen, we find that the suppression of KCTD20 potently ameliorates tau pathology and neurodegeneration in glutamate-treated organoids and mice, as well as in transgenic mice overexpressing mutant human tau. KCTD20 suppression reduces oligomeric tau and improves neuron survival by activating lysosomal exocytosis, which clears pathological tau. Our results show that glutamate signaling can induce neuronal tau pathology and identify KCTD20 suppression and lysosomal exocytosis as effective strategies for clearing neurotoxic tau species.
Collapse
Affiliation(s)
- Joshua E Berlind
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Jesse D Lai
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA; Department of Neuroscience, Amgen Inc., Cambridge, MA, USA; Neurological & Rare Diseases, Dewpoint Therapeutics, Boston, MA, USA.
| | - Cecilia Lie
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Jokabeth Vicente
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Kelsey Lam
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Sheron Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Jonathan Chang
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Violeta Yu
- Neurological & Rare Diseases, Dewpoint Therapeutics, Boston, MA, USA
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Bao C, Luo H, Wang J, Liu X, Li Y, Yang J, Chen C, Yang R, Ba W, Lian X, Dunk M, Liu J, Xu W. Poor glymphatic function is associated with mild cognitive impairment and its progression to Alzheimer's disease: A DTI-ALPS study. J Prev Alzheimers Dis 2025:100156. [PMID: 40221240 DOI: 10.1016/j.tjpad.2025.100156] [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: 12/03/2024] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND We aimed to explore the association between ALPS index and both risks of MCI from cognitively normal (CN) and incident AD progressed from MCI, as well as potential mediating factors. METHODS This study included 519 adults including 253 (48.75 %) CN and 266 (51.25 %) MCI participants from Alzheimer's Disease Neuroimaging Initiative. Glymphatic function (assessed by along the perivascular space [ALPS] index) was measured by diffusion tensor image at baseline. Neurobiomarkers (Aβ and tau from CSF, plasma and PET) and cognitive functions were served as mediators. Data were analyzed using Cox and Laplace regression and mediation analysis. RESULTS During follow-up (median 3.6 years, interquartile range [IQR]: 2.0-4.9 years), 30 (11.86 %) participants developed MCI in the CN cohort and 73 (27.4 %) participants progressed to AD in the MCI cohort. The hazard ratios (95 % confidence intervals [CIs]) of the higher ALPS index was 0.605 (0.386-0.948) for MCI and 0.501 (0.356-0.706) for AD. In addition, participants with high ALPS index had 3.837 and 3.466 years prolonged onset of MCI and AD, separately. Aβ in choroid plexus (17.1 %), tau in cortex [Inferiortemporal (21.1 %), Middletemporal (AV1451:17.0 %, FTP:15.5 %), Superiortemporal(7.7 %), Meta_temporal (AV1451:17.5 %, FTP:16.6 %)], and executive function (14.1 %) mediated the association between ALPS and MCI-AD progression. CONCLUSION High ALPS index decreases MCI risk and delays MCI progression to AD by approximately 3.5 years. Aβ in choroid plexus, tau in cortex, and executive function may partially mediate the MCI-AD progression in relation to ALPS index.
Collapse
Affiliation(s)
- Cuiping Bao
- Department of Radiology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin 300121, China
| | - Hongbin Luo
- Tianjin Jinnan District Center for Disease Control and Prevention, 28 Highway Road, Jinnan District, Tianjin 300350, China
| | - Jiao Wang
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China; Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Skockholm, Sweden
| | - Xuehuan Liu
- Department of Radiology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin 300121, China
| | - Yiming Li
- Department of Radiology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin 300121, China
| | - Jun Yang
- Department of Radiology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin 300121, China
| | - Chong Chen
- Department of Clinical Laboratory, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin 300121, China
| | - Rongrong Yang
- Public Health Science and Engineering College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Weili Ba
- Department of Radiology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin 300121, China
| | - Xinying Lian
- Tianjin Fourth Central Hospital, The Affiliated Hospital of Tianjin Medical University, Zhongshan Rord 1st Tianjin 300140, China
| | - Michelle Dunk
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Skockholm, Sweden
| | - Jun Liu
- Tianjin Fourth Central Hospital, The Affiliated Hospital of Tianjin Medical University, Zhongshan Rord 1st Tianjin 300140, China; Tianjin Fourth Central Hospital, Tianjin University, Zhongshan Rord 1st, Tianjin 300140, China; The Institute of Translational Medicine, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, 300121, China.
| | - Weili Xu
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Skockholm, Sweden
| |
Collapse
|
9
|
Wu C, Yuan J, Tian Y, Wang Y, He X, Zhao K, Huang J, Jiang R. Tauopathy after long-term cervical lymphadenectomy. Alzheimers Dement 2025; 21:e70136. [PMID: 40189841 PMCID: PMC11973124 DOI: 10.1002/alz.70136] [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: 11/18/2024] [Revised: 02/13/2025] [Accepted: 03/04/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION This study examined the effects of long-term cervical lymphadenectomy (cLE) on cognitive and Alzheimer's disease (AD)-like tauopathy changes. METHODS Male C57BL/6 mice were used to assess cLE impacts on sleep, brain pathways, and pathologies. RNA sequencing and proteomics analyzed gene/protein changes, with results verified by western blotting and immunofluorescence. RESULTS CLE led to sleep and psychiatric disorders, linked to mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) pathway activation. Activation of ERK may interfere with autophagy and is associated with phosphorylated tau accumulation. Peripheral blood analysis shows decreased brain waste in the peripheral blood post-cLE, implicating impaired lymphatic drainage and brain waste build-up. DISCUSSION These findings suggest a potential connection between cLE and AD-like tauopathy, potentially influencing surgical decisions. HIGHLIGHTS Cervical lymphadenectomy (cLE) is the cornerstone of head and neck cancers, affecting millions of people each year. We provide the first evidence of mildly impaired cognitive functioning with significant anxiety-depressive disorders in mice after long-term cLE. Long-term cLE not only directly impairs brain wastes (amyloid beta, phosphorylated tau [p-tau]) drainage, but also activates the Erk1/2 signaling pathway leading to attenuation of autophagy. We found for the first time that long-term cLE accelerated the deposition of p-tau in young mice. Patients after clinical cervical lymph node dissection showed reduced brain waste in peripheral blood consistent with mouse models. This study suggests the need for further evaluation of the neurologic effects of cervical lymph node dissection, a procedure that affects millions of people each year.
Collapse
Affiliation(s)
- Chenrui Wu
- Department of NeurosurgeryTianjin Neurological InstituteState Key Laboratory of Experimental HematologyLaboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of EducationTianjin Medical University General HospitalTianjinChina
- Department of NeurosurgerySichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Jiangyuan Yuan
- Department of NeurosurgeryTianjin Neurological InstituteState Key Laboratory of Experimental HematologyLaboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of EducationTianjin Medical University General HospitalTianjinChina
| | - Yu Tian
- Department of NeurosurgeryTianjin Neurological InstituteState Key Laboratory of Experimental HematologyLaboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of EducationTianjin Medical University General HospitalTianjinChina
| | - Youlin Wang
- Department of General SurgeryTianjin Medical University General HospitalTianjinChina
| | - Xianghui He
- Department of General SurgeryTianjin Medical University General HospitalTianjinChina
| | - Ke Zhao
- Department of General SurgeryTianjin Medical University General HospitalTianjinChina
| | - Jinhao Huang
- Department of NeurosurgeryTianjin Neurological InstituteState Key Laboratory of Experimental HematologyLaboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of EducationTianjin Medical University General HospitalTianjinChina
| | - Rongcai Jiang
- Department of NeurosurgeryTianjin Neurological InstituteState Key Laboratory of Experimental HematologyLaboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of EducationTianjin Medical University General HospitalTianjinChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
10
|
Cunningham HA, Dovek L, Recoder N, Bryant-Ekstrand MD, Ligman BR, Piantino J, Lim MM, Elliott JE. Heart rate variability impairment during sleep in Veterans with rapid eye movement sleep behaviour disorder, traumatic brain injury, and post-traumatic stress disorder: an early potential window into autonomic dysfunction? J Sleep Res 2025:e70004. [PMID: 40065628 DOI: 10.1111/jsr.70004] [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: 11/04/2024] [Revised: 12/16/2024] [Accepted: 01/29/2025] [Indexed: 03/15/2025]
Abstract
Individuals with comorbid rapid eye movement (REM) sleep behaviour disorder (RBD) and neurotrauma (NT; defined by traumatic brain injury and post-traumatic stress disorder) have an earlier age of RBD symptom onset, increased RBD-related symptom severity and more neurological features indicative of prodromal synucleinopathy compared to RBD only. An early sign of neurodegenerative condition is autonomic dysfunction, which we sought to evaluate by examining heart rate variability during sleep. Participants with overnight polysomnography were recruited from the Veterans Affairs Portland Health Care System. Veterans without NT or RBD (controls, n = 19), with RBD only (RBD, n = 14), and with RBD and NT (RBD+NT, n = 19) were evaluated. Eligible 5-min non-REM (NREM) and REM epochs without apneas/hypopneas, microarousals, and ectopic beats were analysed for frequency and time domain (e.g., low-frequency [LF] power; high-frequency [HF] power; root mean square of successive R-R intervals [RMSSD]; percentage of R-R intervals that vary ≥50 ms [pNN50]) heart rate variability outcomes. Heart rate did not significantly differ between groups in any sleep stage. Time domain and frequency domain variables (e.g., LF power, HF power, RMSSD, and pNN50) were significantly reduced in the RBD+NT group compared to the controls and RBD-only group during NREM sleep. There were no group differences detected during REM sleep. These data suggest significant reductions in heart rate variability during NREM sleep in RBD+NT participants, suggesting greater autonomic dysfunction compared to controls or RBD alone. Heart rate variability during sleep may be an early, promising biomarker, yielding mechanistic insight for diagnosis and prognosis of early neurodegeneration in this vulnerable population.
Collapse
Affiliation(s)
- Hannah A Cunningham
- VA Portland Health Care System, Research Service, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Laura Dovek
- VA Portland Health Care System, Research Service, Portland, Oregon, USA
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, USA
| | - Natasha Recoder
- VA Portland Health Care System, Research Service, Portland, Oregon, USA
| | | | - Brittany R Ligman
- VA Portland Health Care System, Research Service, Portland, Oregon, USA
| | - Juan Piantino
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, USA
| | - Miranda M Lim
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, USA
- VA Portland Health Care System, VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), Portland, Oregon, USA
- VA Portland Health Care System, National Center for Rehabilitative Auditory Research (NCRAR) and Neurology, Portland, Oregon, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, Oregon, USA
| | - Jonathan E Elliott
- VA Portland Health Care System, Research Service, Portland, Oregon, USA
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, USA
- VA Portland Health Care System, VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), Portland, Oregon, USA
| |
Collapse
|
11
|
Ragsdale SM, Radovich JM, Coiduras II, McCall WV, Grant SC, Lee C, Wilber A. Dual orexin receptor antagonists as promising therapeutics for Alzheimer's disease. NPJ BIOLOGICAL TIMING AND SLEEP 2025; 2:11. [PMID: 40066297 PMCID: PMC11890173 DOI: 10.1038/s44323-025-00025-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/23/2025] [Indexed: 03/15/2025]
Abstract
We examine the relationship between sleep, glymphatics and Alzheimer's disease (AD), and recent work questioning glymphatic clearance during sleep. We highlight a need for understanding glymphatic and/or other mechanism of clearance during sleep, and review glymphatic flow measurement methods. Further, we explore dual orexin receptor antagonists (DORAs) potential to mitigate AD sleep disturbances and enhance clearance. Further research could elucidate a linkage between DORAs, improved sleep and reducing AD pathophysiology.
Collapse
Affiliation(s)
- S. M. Ragsdale
- Department of Psychology; Program in Neuroscience; Florida State University, Tallahassee, FL USA
| | - J. M. Radovich
- Department of Chemical & Biochemical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL USA
- CIMAR, National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL USA
| | - I. I. Coiduras
- Department of Psychology; Program in Neuroscience; Florida State University, Tallahassee, FL USA
| | - W. V. McCall
- Department of Psychiatry and Health Behavior; Medical College of Georgia; Augusta University, Augusta, GA USA
| | - S. C. Grant
- Department of Chemical & Biochemical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL USA
- CIMAR, National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL USA
| | - C. Lee
- Department of Biomedical Sciences; Program in Neuroscience; College of Medicine, Florida State University, Tallahassee, FL USA
| | - A. Wilber
- Department of Psychology; Program in Neuroscience; Florida State University, Tallahassee, FL USA
| |
Collapse
|
12
|
Kritsilis M, Vanherle L, Rosenholm M, in ‘t Zandt R, Yao Y, Swanberg KM, Weikop P, Gottschalk M, Shanbhag NC, Luo J, Boster K, Nedergaard M, Meissner A, Lundgaard I. Loss of glymphatic homeostasis in heart failure. Brain 2025; 148:985-1000. [PMID: 39693238 PMCID: PMC11884761 DOI: 10.1093/brain/awae411] [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: 08/20/2024] [Revised: 11/04/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Heart failure is associated with progressive reduction in cerebral blood flow and neurodegenerative changes leading to cognitive decline. The glymphatic system is crucial for the brain's waste removal, and its dysfunction is linked to neurodegeneration. In this study, we used a mouse model of heart failure, induced by myocardial infarction, to investigate the effects of heart failure with reduced ejection fraction on the brain's glymphatic function. Using dynamic contrast-enhanced MRI and high-resolution fluorescence microscopy, we found increased solute influx from the CSF spaces to the brain, i.e. glymphatic influx, at 12 weeks post-myocardial infarction. Two-photon microscopy revealed that cerebral arterial pulsatility, a major driver of the glymphatic system, was potentiated at this time point, and could explain this increase in glymphatic influx. However, clearance of proteins from the brain parenchyma did not increase proportionately with influx, while a relative increase in brain parenchyma volume was found at 12 weeks post-myocardial infarction, suggesting dysregulation of brain fluid dynamics. Additionally, our results showed a correlation between brain clearance and cerebral blood flow. These findings highlight the role of cerebral blood flow as a key regulator of the glymphatic system, suggesting its involvement in the development of brain disorders associated with reduced cerebral blood flow. This study paves the way for future investigations into the effects of cardiovascular diseases on the brain's clearance mechanisms, which may provide novel insights into the prevention and treatment of cognitive decline.
Collapse
Affiliation(s)
- Marios Kritsilis
- Department of Experimental Medical Science, Lund University, 22362 Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Lotte Vanherle
- Department of Experimental Medical Science, Lund University, 22362 Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, University of Augsburg, 86159 Augsburg, Germany
| | - Marko Rosenholm
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - René in ‘t Zandt
- Lund University Bioimaging Centre, Lund University, 22184 Lund, Sweden
| | - Yuan Yao
- Department of Computer Science, University of Rochester, Rochester, NY 14627, USA
| | - Kelley M Swanberg
- Department of Experimental Medical Science, Lund University, 22362 Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Nagesh C Shanbhag
- Department of Experimental Medical Science, Lund University, 22362 Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Jiebo Luo
- Department of Computer Science, University of Rochester, Rochester, NY 14627, USA
| | - Kimberly Boster
- Department of Mechanical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, NY 14642, USA
| | - Anja Meissner
- Department of Experimental Medical Science, Lund University, 22362 Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, University of Augsburg, 86159 Augsburg, Germany
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, 22362 Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
| |
Collapse
|
13
|
Liu L, Zeng Q, Luo X, Hong H, Fang Y, Xie L, Zhang Y, Lin M, Wang S, Li K, Liu X, Zhang R, Chen Y, Yang Y, Huang P. Association Between Single Nucleotide Polymorphisms in the Aquaporin-4 Gene and Longitudinal Changes in White Matter Free Water and Cognitive Function in Non-Demented Older Adults. Hum Brain Mapp 2025; 46:e70171. [PMID: 40016624 PMCID: PMC11867789 DOI: 10.1002/hbm.70171] [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: 11/24/2024] [Revised: 01/19/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
We investigated whether aquaporin-4 (AQP4) single-nucleotide polymorphisms (SNPs) influence Alzheimer's disease (AD) progression through changes in the glymphatic system. We included 242 non-dementia participants and chose six SNPs previously shown to be related to AD. We analyzed the associations between AQP4 SNPs and glymphatic markers, including enlarged perivascular spaces (PVS), white matter free water (FW), and diffusion tensor image analysis along the perivascular space (DTI-ALPS), in both cross-sectional and longitudinal data. We investigated whether AQP4-related glymphatic markers are associated with AD pathology progression and cognitive impairment, and whether they mediate the relationship between AQP4 SNPs and AD progression. There was no association between AQP4 SNPs and glymphatic markers at baseline. Carriers of the AQP4 SNP rs72878794 minor allele status exhibited slower FW increase in the amyloid-positive group (SNP*time: β = -0.0040, t(46.25) = -2.062, p = 0.045, 95% CI = -0.0078 ~ -0.0001), whereas the rs9951307 minor allele carrier showed a faster FW increase in the amyloid-negative group (SNP*time: β =0.0033, t(81.19) = 2.245, p = 0.027, 95% CI = 0.0004 ~ 0.0062). The higher FW was associated with faster cognitive decline at follow-ups. AQP4 SNPs influence interstitial fluid accumulation, contributing to cognitive decline but not amyloid deposition in AD. Further studies are needed to clarify the pathways linking AQP4 SNPs and AD progression.
Collapse
Affiliation(s)
- Lingyun Liu
- Department of RadiologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Qingze Zeng
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiao Luo
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Hui Hong
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Yi Fang
- Department of NeurologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Linyun Xie
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Yao Zhang
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Miao Lin
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Shuyue Wang
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Kaicheng Li
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiaocao Liu
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Ruiting Zhang
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Yanxing Chen
- Department of NeurologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Yunjun Yang
- Department of RadiologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Peiyu Huang
- Department of RadiologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of RadiologyThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
14
|
Li H, Zhao H, Chen L, Zhang W, Zhang W, Cheng X. Rapid screening of acetylcholinesterase inhibitors in Qi-Fu-Yin using magnetic metal-organic frameworks immobilized with acetylcholinesterase. Bioorg Chem 2025; 156:108183. [PMID: 39827651 DOI: 10.1016/j.bioorg.2025.108183] [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: 12/09/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Current immobilization approaches for ligand fishing often experience challenges such as limited protein loading capacity and difficulties in the recycling process. To overcome these challenges, we synthesized a magnetic metal-organic frameworks (MMOFs) composite, which can be rapidly separated and has a large specific surface area, and employed it to immobilize acetylcholinesterase (AChE). The synthesized MMOFs@AChE composite exhibited a high immobilization yield (129.7 mg/g) and excellent relative activity recovery (88.1 %). Furthermore, immobilized AChE can improve its resistance to alkaline environments and high temperatures. After being stored at 4 °C for a month, the immobilized enzyme maintained 91.4 % of its original activity, significantly higher than the free enzyme (77.6 %). Furthermore, it preserved more than 80 % of its initial activity after five cycles and 68.7 % after eight cycles. The composite MMOFs@AChE was then incubated with Qi-Fu-Yin extract to fish for ligands binding to AChE. Notably, Qi-Fu-Yin can alleviate Alzheimer's disease (AD) symptoms by modulating the AChE pathway, while active compounds remain unclear. Sixteen potential AChE inhibitors were identified through UHPLC-Q-Exactive-Orbitrap-MS/MS. The results of ligand fishing were validated through molecular docking studies, molecular dynamics simulation, surface plasmon resonance and AChE inhibitory activity assays. The screened compounds may exert inhibitory effects on AChE by altering the spatial configuration of the catalytic site or by influencing the binding of the substrate to the catalytic site, catalytic anionic site and peripheral anionic site regions. The MMOFs@AChE-based ligand fishing platform offers an efficient, effective, and convenient approach for enzymatic inhibitors discovery from natural products.
Collapse
Affiliation(s)
- Hengyu Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078 China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China.
| | - Hongwei Zhao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China
| | - Lingxiao Chen
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China
| | - Wenting Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China
| | - Wei Zhang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078 China.
| | - Xiaorui Cheng
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078 China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China.
| |
Collapse
|
15
|
Beltran-Velasco AI. Brain Glycogen-Its Metabolic Role in Neuronal Health and Neurological Disorders-An Extensive Narrative Review. Metabolites 2025; 15:128. [PMID: 39997753 PMCID: PMC11857135 DOI: 10.3390/metabo15020128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Brain glycogen is imperative for neuronal health, as it supports energy demands and metabolic processes. This review examines the pathways involved in glycogen storage and utilization in the central nervous system, emphasizing their role in both physiology and pathology. It explores how alterations in glycogen metabolism contribute to neurological disorders, including neurodegenerative diseases, epilepsy, and metabolic conditions while highlighting the bidirectional interaction between neurons and glia in maintaining brain homeostasis. Methods: A comprehensive search of articles published between 2015 and 2025 was conducted using the following databases: ScienceDirect, Scopus, Wiley, Web of Science, Medline, and PubMed. The selection of relevant studies was based on their focus on brain glycogen metabolism and its role in neurological conditions, with studies that did not meet the inclusion criteria being excluded. Results: The metabolic processes of brain glycogen are subject to rigorous regulation by astrocyte-neuron interactions, thereby ensuring metabolic homeostasis and energy availability. The dysregulation of glycogen storage and mobilization has been implicated in the development of synaptic dysfunction, excitotoxicity, and neurodegeneration in a variety of disorders. For instance, aberrant glycogen accumulation in diseases such as Lafora disease has been associated with severe neurodegeneration, while impaired glycogen mobilization has been shown to exacerbate energy deficits in Alzheimer's and epilepsy. Conclusions: Targeting brain glycogen metabolism represents a promising approach for therapeutic intervention in neurological disorders. However, the translation of these strategies to human models remains challenging, particularly with regard to the long-term safety and specificity of glycogen-targeted therapies.
Collapse
|
16
|
Hauglund NL, Andersen M, Tokarska K, Radovanovic T, Kjaerby C, Sørensen FL, Bojarowska Z, Untiet V, Ballestero SB, Kolmos MG, Weikop P, Hirase H, Nedergaard M. Norepinephrine-mediated slow vasomotion drives glymphatic clearance during sleep. Cell 2025; 188:606-622.e17. [PMID: 39788123 DOI: 10.1016/j.cell.2024.11.027] [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: 12/01/2023] [Revised: 08/29/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025]
Abstract
As the brain transitions from wakefulness to sleep, processing of external information diminishes while restorative processes, such as glymphatic removal of waste products, are activated. Yet, it is not known what drives brain clearance during sleep. We here employed an array of technologies and identified tightly synchronized oscillations in norepinephrine, cerebral blood volume, and cerebrospinal fluid (CSF) as the strongest predictors of glymphatic clearance during NREM sleep. Optogenetic stimulation of the locus coeruleus induced anti-correlated changes in vasomotion and CSF signal. Furthermore, stimulation of arterial oscillations enhanced CSF inflow, demonstrating that vasomotion acts as a pump driving CSF into the brain. On the contrary, the sleep aid zolpidem suppressed norepinephrine oscillations and glymphatic flow, highlighting the critical role of norepinephrine-driven vascular dynamics in brain clearance. Thus, the micro-architectural organization of NREM sleep, driven by norepinephrine fluctuations and vascular dynamics, is a key determinant for glymphatic clearance.
Collapse
Affiliation(s)
- Natalie L Hauglund
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; Danish Center for Sleep Medicine, Department of Clinical Neurophysiology, Rigshospitalet, 2600 Glostrup, Denmark
| | - Mie Andersen
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Klaudia Tokarska
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tessa Radovanovic
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Celia Kjaerby
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Frederikke L Sørensen
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Zuzanna Bojarowska
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Verena Untiet
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Sheyla B Ballestero
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Mie G Kolmos
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Hajime Hirase
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester, Rochester, NY 14627, USA.
| |
Collapse
|
17
|
Canet G, Da Gama Monteiro F, Rocaboy E, Diego-Diaz S, Khelaifia B, Godbout K, Lachhab A, Kim J, Valencia DI, Yin A, Wu HT, Howell J, Blank E, Laliberté F, Fortin N, Boscher E, Fereydouni-Forouzandeh P, Champagne S, Guisle I, Hébert SS, Pernet V, Liu H, Lu W, Debure L, Rapoport DM, Ayappa I, Varga AW, Parekh A, Osorio RS, Lacroix S, Burns MP, Lucey BP, Blessing EM, Planel E. Sleep-wake variation in body temperature regulates tau secretion and correlates with CSF and plasma tau. J Clin Invest 2025; 135:e182931. [PMID: 39903530 PMCID: PMC11957704 DOI: 10.1172/jci182931] [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: 07/02/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
Sleep disturbance is bidirectionally associated with an increased risk of Alzheimer's disease and other tauopathies. While the sleep-wake cycle regulates interstitial and cerebrospinal fluid (CSF) tau levels, the underlying mechanisms remain unknown. Understanding these mechanisms is crucial, given the evidence that tau pathology spreads through neuron-to-neuron transfer, involving the secretion and internalization of pathological tau forms. Here, we combined in vitro, in vivo, and clinical methods to reveal a pathway by which changes in body temperature (BT) over the sleep-wake cycle modulate extracellular tau levels. In mice, a higher BT during wakefulness and sleep deprivation increased CSF and plasma tau levels, while also upregulating unconventional protein secretion pathway I (UPS-I) events including (a) intracellular tau dephosphorylation, (b) caspase 3-mediated cleavage of tau (TauC3), and (c) membrane translocation of tau through binding to phosphatidylinositol 4,5-bisphosphate (PIP2) and syndecan 3. In humans, the increase in CSF and plasma tau levels observed after wakefulness correlated with BT increases during wakefulness. By demonstrating that sleep-wake variation in BT regulates extracellular tau levels, our findings highlight the importance of thermoregulation in linking sleep disturbances to tau-mediated neurodegeneration and the preventative potential of thermal interventions.
Collapse
Affiliation(s)
- Geoffrey Canet
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Felipe Da Gama Monteiro
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
| | - Emma Rocaboy
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Sofia Diego-Diaz
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Boutheyna Khelaifia
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Kelly Godbout
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Aymane Lachhab
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Jessica Kim
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Daphne I. Valencia
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Audrey Yin
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Hau-Tieng Wu
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Jordan Howell
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Emily Blank
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Francis Laliberté
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
| | - Nadia Fortin
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
| | - Emmanuelle Boscher
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | | | - Stéphanie Champagne
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Isabelle Guisle
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Sébastien S. Hébert
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Vincent Pernet
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
- Department of Neurology, Inselspital, and
- Center for Experimental Neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - William Lu
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ludovic Debure
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - David M. Rapoport
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Indu Ayappa
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew W. Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ankit Parekh
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ricardo S. Osorio
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Steve Lacroix
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
| | - Mark P. Burns
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Brendan P. Lucey
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Esther M. Blessing
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Emmanuel Planel
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| |
Collapse
|
18
|
Al-Diwani A, Provine NM, Murchison A, Laban R, Swann OJ, Koychev I, Sheerin F, Da Mesquita S, Heslegrave A, Zetterberg H, Klenerman P, Irani SR. Neurodegenerative fluid biomarkers are enriched in human cervical lymph nodes. Brain 2025; 148:394-400. [PMID: 39432679 PMCID: PMC11788199 DOI: 10.1093/brain/awae329] [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: 04/30/2024] [Revised: 08/06/2024] [Accepted: 09/07/2024] [Indexed: 10/23/2024] Open
Abstract
In animal models, brain neurodegeneration biomarkers drain into cervical lymph nodes (CLNs), and this drainage function is reduced with ageing. If this occurred in humans, CLNs may provide a readily accessible measure of this aspect of protein clearance. We tested this hypothesis in people using ultrasound-guided fine needle aspiration. We measured amyloid-beta 40 and 42, phosphorylated tau 181 (pTau181), glial fibrillary acidic protein and neurofilament light using single molecule array in CLN aspirates and plasma from: (i) a discovery cohort of 25 autoimmune patients; and (ii) plasma, CLNs and capillary blood in four healthy volunteers, an optimization cohort. Ultrasound-guided fine needle aspiration was well-tolerated by all participants. In both cohorts, all biomarkers were detected in all plasma and CLN samples, other than neurofilament light (8/17 of discovery cohort). CLN biomarker concentrations were significantly greater than plasma concentrations for all except neurofilament light, most markedly for pTau181 (266-fold; P < 0.02), whose CLN concentrations decreased with age (Spearman r = -0.66, P = 0.001). This study presents the first evidence that neurodegenerative biomarkers are detectable in human CLNs. Raised CLN:plasma biomarker ratios suggest their concentration in CLNs may offer a distinct compartment for minimally-invasive measurement of brain clearance and lymphatic drainage, with potential applicability to study of ageing and future clinical trials.
Collapse
Affiliation(s)
- Adam Al-Diwani
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
| | - Nicholas M Provine
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Andrew Murchison
- Department of Radiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Rhiannon Laban
- Fluid Biomarker Laboratory, UK Dementia Research Institute at UCL, London W1T 7NF, UK
| | - Owen J Swann
- Fluid Biomarker Laboratory, UK Dementia Research Institute at UCL, London W1T 7NF, UK
| | - Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
| | - Fintan Sheerin
- Department of Radiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | | | - Amanda Heslegrave
- Fluid Biomarker Laboratory, UK Dementia Research Institute at UCL, London W1T 7NF, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Henrik Zetterberg
- Fluid Biomarker Laboratory, UK Dementia Research Institute at UCL, London W1T 7NF, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 39, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 39, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Paul Klenerman
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, UK
| | - Sarosh R Irani
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
19
|
Jiao F, Wang Q, Zhong J, Lin H, Lu J, Wang L, Wang M, Liu F, Jiang J, Zuo C. Relationships Between Glymphatic System Activity and Tau Burden, Dopaminergic Impairment, Abnormal Glucose Metabolism in Progressive Supranuclear Palsy. CNS Neurosci Ther 2025; 31:e70284. [PMID: 39963843 PMCID: PMC11833299 DOI: 10.1111/cns.70284] [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: 10/14/2024] [Revised: 01/23/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is a primary tauopathy characterized by dopaminergic impairment and abnormal glucose metabolism. The glymphatic system can promote the elimination of tau protein. The association between glymphatic function and pathological hallmark in neuroimaging remains unknown. METHODS Diffusion tensor imaging (DTI) and positron emission tomography (PET) scanning with 18F-Florzolotau, 18F-FPCIT, and 18F-FDG were performed in PSP patients. DTI analysis along the perivascular space (ALPS) index was computed to assess glymphatic function, while the semi-quantitative value was employed to measure tau burden and dopaminergic impairment. The PSP-related pattern (PSPRP) served as an indicator of abnormal metabolic brain network activity. RESULTS PSP patients exhibited changes in ALPS index and tau deposition. ALPS index, tau deposition, and PSPRP expression showed significant correlations with clinical scores. Additionally, ALPS index was correlated with tau deposition and PSPRP expression. However, neither ALPS index nor the clinical scores were correlated with striatum dysfunction. Finally, tau deposition in subcortical regions and PSPRP expression exhibited mediating effects between ALPS index and clinical scores. CONCLUSION The glymphatic dysfunction is associated with tau deposition and abnormal metabolic brain network activity and is independent of dopaminergic impairment in PSP.
Collapse
Affiliation(s)
- Fangyang Jiao
- Department of Nuclear Medicine & PET Center, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Qingmin Wang
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Jiayi Zhong
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Huamei Lin
- Department of Nuclear Medicine & PET Center, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Jiaying Lu
- Department of Nuclear Medicine & PET Center, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Luyao Wang
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Min Wang
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Fengtao Liu
- Department of Neurology, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Jiehui Jiang
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Chuantao Zuo
- Department of Nuclear Medicine & PET Center, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
20
|
Park JM, Tsai LH. Innovations in noninvasive sensory stimulation treatments to combat Alzheimer's disease. PLoS Biol 2025; 23:e3003046. [PMID: 40019895 PMCID: PMC11870349 DOI: 10.1371/journal.pbio.3003046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting millions worldwide. There is no known cure for AD, highlighting an urgent need for new, innovative treatments. Recent studies have shed light on a promising, noninvasive approach using sensory stimulation as a potential therapy for AD. Exposing patients to light and sound pulses at a frequency of 40 hertz induces brain rhythms in the gamma frequency range that are important for healthy brain activity. Using this treatment in animal models, we are now beginning to understand the molecular, cellular, and circuit-level changes that underlie improvements in disease pathology, cognition, and behavior. A mechanistic understanding of the basic biology that underlies the 40-hertz treatment will inform ongoing clinical trials that offer a promising avenue of treatment without the side effects and high costs typically associated with pharmacological interventions. Concurrent advancements in neurotechnology that can also noninvasively stimulate healthy brain rhythms are illuminating new possibilities for alternative therapies. Altogether, these noninvasive approaches could herald a new era in treating AD, making them a beacon of hope for patients, families, and caregivers facing the challenges of this debilitating condition.
Collapse
Affiliation(s)
- Jung M. Park
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
21
|
Chen K, Du X, Chao MA, Xie Z, Yang G. Surgery impairs glymphatic activity and cognitive function in aged mice. Mol Brain 2025; 18:7. [PMID: 39856767 PMCID: PMC11763125 DOI: 10.1186/s13041-025-01177-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: 11/15/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
Delirium is a common complication in elderly surgical patients and is associated with an increased risk of dementia. Although advanced age is a major risk factor, the mechanisms underlying postoperative delirium remain poorly understood. The glymphatic system, a brain-wide network of perivascular pathways, facilitates cerebrospinal fluid (CSF) flow and supports the clearance of metabolic waste. Impairments in glymphatic function have been observed in aging brains and various neurodegenerative conditions. Using in vivo two-photon imaging, we examined the effects of surgery (laparotomy) on glymphatic function in adult (6 months) and aged (18 months) mice 24 h post-surgery. In adult mice, CSF tracer entry into the brain parenchyma along periarteriolar spaces occurred rapidly following intracisternal tracer injection, with no significant differences between sham and surgery groups. In contrast, aged mice exhibited delayed tracer influx, with further impairments observed in the surgery group compared to sham controls. This glymphatic dysfunction correlated with poorer T-maze performance in aged mice. These findings suggest that surgery exacerbates glymphatic impairment in aging brains, potentially hindering brain waste clearance and contributing to postoperative delirium.
Collapse
Affiliation(s)
- Kai Chen
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Xingyu Du
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Melissa A Chao
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
22
|
Ran L, Fang Y, Cheng C, He Y, Shao Z, Kong Y, Huang H, Xu S, Luo X, Wang W, Hao X, Wang M. Genome-wide and phenome-wide studies provided insights into brain glymphatic system function and its clinical associations. SCIENCE ADVANCES 2025; 11:eadr4606. [PMID: 39823331 PMCID: PMC11740961 DOI: 10.1126/sciadv.adr4606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025]
Abstract
We applied an MRI technique diffusion tensor imaging along the perivascular space (DTI-ALPS) for assessing glymphatic system (GS) in a genome-wide association study (GWAS) and phenome-wide association study (PheWAS) of 40,486 European individuals. Exploratory analysis revealed 17 genetic loci significantly associating with the regional DTI-ALPS index. We found 58 genes, including SPPL2C and EFCAB5, which prioritized in the DTI-ALPS index subtypes and associated with neurodegenerative diseases. PheWAS of 241 traits suggested that body mass index and blood pressure phenotypes closely related to GS function. Moreover, we detected disrupted GS function in 44 of 625 predefined disease conditions. Notably, Mendelian randomization and mediation analysis indicated that lower DTI-ALPS index was a risk factor for ischemic stroke (odds ratio = 1.56, P = 0.028) by partly mediating the risk factor of obesity. Results provide insights into the genetic architecture and mechanism for the DIT-ALPS index and highlight its great clinical value, especially in cerebral stroke.
Collapse
Affiliation(s)
- Lusen Ran
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Fang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Cheng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqin He
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhonghe Shao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Kong
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shabei Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Huang SY, Ge YJ, Ren P, Wu BS, Gong W, Du J, Chen SD, Kang JJ, Ma Q, Bokde ALW, Desrivières S, Garavan H, Grigis A, Lemaitre H, Smolka MN, Hohmann S, Feng JF, Zhang YR, Cheng W, Yu JT. Genome-wide association study unravels mechanisms of brain glymphatic activity. Nat Commun 2025; 16:626. [PMID: 39805841 PMCID: PMC11730627 DOI: 10.1038/s41467-024-55706-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Brain glymphatic activity, as indicated by diffusion-tensor imaging analysis along the perivascular space (ALPS) index, is involved in developmental neuropsychiatric and neurodegenerative diseases, but its genetic architecture is poorly understood. Here, we identified 17 unique genome-wide significant loci and 161 candidate genes linked to the ALPS-indexes in a discovery sample of 31,021 individuals from the UK Biobank. Seven loci were replicated in two independent datasets. Genetic signals located at the 2p23.3 locus yielded significantly concordant effects in both young and aging cohorts. Genetic correlation and polygenic overlap analyses indicate a common underlying genetic mechanism between the ALPS-index, ventricular volumes, and cerebrospinal fluid tau levels, with GMNC (3q28) and C16orf95 (16q24.2) as the shared genetic basis. Our findings enhance the understanding of the genetics of the ALPS-index and provide insight for further research into the neurobiological mechanisms of glymphatic clearance activity across the lifespan and its relation to neuropsychiatric phenotypes.
Collapse
Affiliation(s)
- Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Ren
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weikang Gong
- School of Data Science, Fudan University, Shanghai, China
| | - Jing Du
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, UNSW, Sydney, Australia
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College, London, UK
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, 05405, Burlington, VT, USA
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Herve Lemaitre
- NeuroSpin, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, 33076, Bordeaux, France
| | - Michael N Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer 79 Center, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Wu W, Zhao Y, Cheng X, Xie X, Zeng Y, Tao Q, Yang Y, Xiao C, Zhang Z, Pang J, Jin J, He H, Lin Y, Li B, Ma J, Ye X, Lin WJ. Modulation of glymphatic system by visual circuit activation alleviates memory impairment and apathy in a mouse model of Alzheimer's disease. Nat Commun 2025; 16:63. [PMID: 39747869 PMCID: PMC11696061 DOI: 10.1038/s41467-024-55678-w] [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: 04/13/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Alzheimer's disease is characterized by progressive amyloid deposition and cognitive decline, yet the pathological mechanisms and treatments remain elusive. Here we report the therapeutic potential of low-intensity 40 hertz blue light exposure in a 5xFAD mouse model of Alzheimer's disease. Our findings reveal that light treatment prevents memory decline in 4-month-old 5xFAD mice and motivation loss in 14-month-old 5xFAD mice, accompanied by restoration of glial water channel aquaporin-4 polarity, improved brain drainage efficiency, and a reduction in hippocampal lipid accumulation. We further demonstrate the beneficial effects of 40 hertz blue light are mediated through the activation of the vLGN/IGL-Re visual circuit. Notably, concomitant use of anti-Aβ antibody with 40 hertz blue light demonstrates improved soluble Aβ clearance and cognitive performance in 5xFAD mice. These findings offer functional evidence on the therapeutic effects of 40 hertz blue light in Aβ-related pathologies and suggest its potential as a supplementary strategy to augment the efficacy of antibody-based therapy.
Collapse
Affiliation(s)
- Wen Wu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yubai Zhao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Clinical and Rehabilitation Medicine, Guiyang Healthcare Vocational University, Guizhou, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoru Xie
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China
| | - Yixiu Zeng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Quan Tao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yishuai Yang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chuan Xiao
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China
| | - Zhan Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China
| | - Jiahui Pang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Jin
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongbo He
- Guangdong Mental Health Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yangyang Lin
- Department of Rehabilitation Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Rehabilitation Medicine, Guangzhou, China
- Biomedical Innovation Center, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Boxing Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Junxian Ma
- Tianfu Xinglong Lake Laboratory, Chengdu, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Wei-Jye Lin
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China.
| |
Collapse
|
25
|
Satyanarayanan SK, Han Z, Xiao J, Yuan Q, Yung WH, Ke Y, Chang RCC, Zhu MH, Su H, Su KP, Qin D, Lee SMY. Frontiers of Neurodegenerative Disease Treatment: Targeting Immune Cells in Brain Border Regions. Brain Behav Immun 2025; 123:483-499. [PMID: 39378973 DOI: 10.1016/j.bbi.2024.10.007] [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: 05/03/2024] [Revised: 09/15/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024] Open
Abstract
Neurodegenerative diseases (NDs) demonstrate a complex interaction with the immune system, challenging the traditional view of the brain as an "immune-privileged" organ. Microglia were once considered the sole guardians of the brain's immune response. However, recent research has revealed the critical role of peripheral immune cells located in key brain regions like the meninges, choroid plexus, and perivascular spaces. These previously overlooked cells are now recognized as contributors to the development and progression of NDs. This newfound understanding opens doors for pioneering therapeutic strategies. By targeting these peripheral immune cells, we may be able to modulate the brain's immune environment, offering an alternative approach to treat NDs and circumvent the challenges posed by the blood-brain barrier. This comprehensive review will scrutinize the latest findings on the complex interactions between these peripheral immune cells and NDs. It will also critically assess the prospects of targeting these cells as a ground-breaking therapeutic avenue for these debilitating disorders.
Collapse
Affiliation(s)
- Senthil Kumaran Satyanarayanan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Zixu Han
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Jingwei Xiao
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing Ho Yung
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Ya Ke
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Faculty of Medicine Building, Hong Kong, China
| | - Maria Huachen Zhu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, Hong Kong, China
| | - Kuan-Pin Su
- An-Nan Hospital, China Medical University, Tainan, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan; Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Suki Man Yan Lee
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
26
|
Liu H, Tan AYS, Mehrabi NF, Turner CP, Curtis MA, Faull RLM, Dragunow M, Singh-Bains MK, Smith AM. Astrocytic proteins involved in regulation of the extracellular environment are increased in the Alzheimer's disease middle temporal gyrus. Neurobiol Dis 2025; 204:106749. [PMID: 39603277 DOI: 10.1016/j.nbd.2024.106749] [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: 05/28/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Alzheimer's disease (AD) has complex pathophysiology involving numerous cell types and brain processes. Astrocyte involvement in AD is gaining increased attention, however a complete characterisation of astrocytic changes in the AD human brain is warranted. Astrocytes perform important homeostatic functions including regulation of the extracellular microenvironment, critical for the health of all brain cells. We have investigated changes to key astrocyte proteins involved in the regulation of CNS extracellular environment in the human AD middle temporal gyrus (MTG): aquaporin-4 (AQP-4), glutamate transporter-1 (GLT-1) and inwardly-rectifying potassium channel 4.1 (Kir4.1). We have used a high-throughput human brain tissue microarray platform with automated quantitative image analysis to measure protein changes in a large cohort of neurological control and AD cases. We found increased astrocytic glial acidic fibrillary protein (GFAP), AQP-4, GLT-1 and Kir4.1 expression that correlates with advancing Braak stage, increasing amyloid pathology and, to a greater extent, the degree of tau pathology. We confirmed that Kir4.1 immunostaining is predominantly found in astrocytes and revealed a novel redistribution of Kir4.1 protein expression into astrocytic processes in the AD MTG. Our study presents novel and potentially modifiable glial changes in the AD human brain that are critical to our understanding of disease pathogenesis.
Collapse
Affiliation(s)
- Henry Liu
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
| | - Adelie Y S Tan
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Nasim F Mehrabi
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
| | - Clinton P Turner
- Department of Anatomical Pathology, Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
| | - Malvindar K Singh-Bains
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Amy M Smith
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand.
| |
Collapse
|
27
|
Van den Bulcke L, Davidoff H, Heremans E, Potts Y, Vansteelandt K, De Vos M, Christiaens D, Emsell L, Jacobson LH, Hoyer D, Buyse B, Vandenbulcke M, Testelmans D, Van Den Bossche M. Acoustic Stimulation to Improve Slow-Wave Sleep in Alzheimer's Disease: A Multiple Night At-Home Intervention. Am J Geriatr Psychiatry 2025; 33:73-84. [PMID: 39048400 DOI: 10.1016/j.jagp.2024.07.002] [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: 05/14/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVES To investigate the efficacy of closed-loop acoustic stimulation (CLAS) during slow-wave sleep (SWS) to enhance slow-wave activity (SWA) and SWS in patients with Alzheimer's disease (AD) across multiple nights and to explore associations between stimulation, participant characteristics, and individuals' SWS response. DESIGN A 2-week, open-label at-home intervention study utilizing the DREEM2 headband to record sleep data and administer CLAS during SWS. SETTING AND PARTICIPANTS Fifteen older patients with AD (6 women, mean age: 76.27 [SD = 6.06], mean MOCA-score: 16.07 [SD = 6.94]), living at home with their partner, completed the trial. INTERVENTION Patients first wore the device for two baseline nights, followed by 14 nights during which the device was programmed to randomly either deliver acoustic stimulations of 50 ms pink noise (± 40 dB) targeted to the slow-wave up-phase during SWS or only mark the wave (sham). RESULTS On a group level, stimulation significantly enhanced SWA and SWS with consistent SWS enhancement throughout the intervention. However, substantial variability existed in individual responses to stimulation. Individuals received more stimulations on nights with increased SWS compared to baseline than on nights with no change or a decrease. In individuals, having lower baseline SWS correlated with receiving fewer stimulations on average during the intervention. CONCLUSION CLAS during SWS is a promising nonpharmacological method to enhance SWA and SWS in AD. However, patients with lower baseline SWS received fewer stimulations during the intervention, possibly resulting in less SWS enhancement. Individual variability in response to stimulation underscores the need to address personalized stimulation parameters in future research and therapy development.
Collapse
Affiliation(s)
- Laura Van den Bulcke
- Geriatric Psychiatry (LVDB, KV, LE, MV, MVDB), University Psychiatric Center KU Leuven, Leuven 3000, Belgium; Neuropsychiatry (LVDB, KV, LE, MV, MVDB), Research Group Psychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium
| | - Hannah Davidoff
- Department of Electrical Engineering (ESAT) (HD, EH, MDV, DC), KU Leuven, Heverlee 3001, Belgium; CSH (Circuits and Systems for Health) - imec (HD), Heverlee 3001, Belgium
| | - Elisabeth Heremans
- Department of Electrical Engineering (ESAT) (HD, EH, MDV, DC), KU Leuven, Heverlee 3001, Belgium
| | - Yasmin Potts
- Florey Institute of Neuroscience and Mental Health (YP, LHJ, DH), Parkville, Victoria 3010, Australia
| | - Kristof Vansteelandt
- Geriatric Psychiatry (LVDB, KV, LE, MV, MVDB), University Psychiatric Center KU Leuven, Leuven 3000, Belgium; Neuropsychiatry (LVDB, KV, LE, MV, MVDB), Research Group Psychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium
| | - Maarten De Vos
- Department of Electrical Engineering (ESAT) (HD, EH, MDV, DC), KU Leuven, Heverlee 3001, Belgium; Department of Development and Regeneration (MDV), Faculty of Medicine, KU Leuven, Leuven 3000, Belgium
| | - Daan Christiaens
- Department of Electrical Engineering (ESAT) (HD, EH, MDV, DC), KU Leuven, Heverlee 3001, Belgium; Translational MRI (LE), Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Louise Emsell
- Geriatric Psychiatry (LVDB, KV, LE, MV, MVDB), University Psychiatric Center KU Leuven, Leuven 3000, Belgium; Neuropsychiatry (LVDB, KV, LE, MV, MVDB), Research Group Psychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium; Translational MRI (LE), Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Laura H Jacobson
- Florey Institute of Neuroscience and Mental Health (YP, LHJ, DH), Parkville, Victoria 3010, Australia; Department of Biochemistry and Pharmacology (LHJ, DH), School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Daniël Hoyer
- Florey Institute of Neuroscience and Mental Health (YP, LHJ, DH), Parkville, Victoria 3010, Australia; Department of Biochemistry and Pharmacology (LHJ, DH), School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia.; Department of Molecular Medicine (DH), The Scripps Research Institute, La Jolla, California 92037, USA
| | - Bertien Buyse
- Department of Pneumology (BB, DT), Leuven University Center for Sleep and Wake disorders, University Hospitals Leuven, Leuven 3000, Belgium; Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE) (BB, DT), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven 3000, Belgium
| | - Mathieu Vandenbulcke
- Geriatric Psychiatry (LVDB, KV, LE, MV, MVDB), University Psychiatric Center KU Leuven, Leuven 3000, Belgium; Neuropsychiatry (LVDB, KV, LE, MV, MVDB), Research Group Psychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium
| | - Dries Testelmans
- Department of Pneumology (BB, DT), Leuven University Center for Sleep and Wake disorders, University Hospitals Leuven, Leuven 3000, Belgium; Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE) (BB, DT), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven 3000, Belgium
| | - Maarten Van Den Bossche
- Geriatric Psychiatry (LVDB, KV, LE, MV, MVDB), University Psychiatric Center KU Leuven, Leuven 3000, Belgium; Neuropsychiatry (LVDB, KV, LE, MV, MVDB), Research Group Psychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium.
| |
Collapse
|
28
|
Sasaki K, Fujita H, Sato T, Kato S, Takahashi Y, Takeshita Y, Kanda T, Saito T, Saido TC, Hattori S, Hozumi Y, Yamada Y, Waki H. GLP-1 receptor signaling restores aquaporin 4 subcellular polarization in reactive astrocytes and promotes amyloid β clearance in a mouse model of Alzheimer's disease. Biochem Biophys Res Commun 2024; 741:151016. [PMID: 39577079 DOI: 10.1016/j.bbrc.2024.151016] [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: 11/09/2024] [Accepted: 11/16/2024] [Indexed: 11/24/2024]
Abstract
The physiological actions of a gut hormone, glucagon-like peptide-1 (GLP-1), in Alzheimer's disease (AD) brain remain poorly understood, although GLP-1 receptor (GLP-1R) expression in this organ has been shown in several experimental studies. Therefore, we explored whether the GLP-1R signaling promotes the clearance of amyloid β (Aβ) (1-42) which is a core pathological hallmark of AD, focusing on the water channel protein aquaporin 4 (AQP4) localized to astrocyte endfeet perivascular membranes in intact brain. First, we confirmed that Glp1r mRNA is predominantly expressed at perivascular site of astrocytes in normal mouse cerebral cortex through in situ hybridization analysis. Next, we observed that 20-week subcutaneous administration of a GLP-1R agonist (GLP-1RA) liraglutide significantly reduced Aβ (1-42) accumulation in the cerebral cortex and improved spatial working memory in an AD mouse model, AppNL-G-F/NL-G-F mice. Furthermore, our current data revealed that the 4-week liraglutide treatment relocalized subcellular AQP4 in morphologically injured reactive astrocytes of AppNL-G-F/NL-G-F mice to the cell surface perivascular site through PKA-mediated AQP4 phosphorylation. Such translocation of phosphorylated AQP4 to astrocyte cell surface following incubation with liraglutide was observed also in the present in vitro study using the cell line in which AQP4 cDNA was introduced into immortalized human astrocyte. These results suggest that enhanced intracerebral GLP-1R signaling following peripheral administration of GLP-1RA restores AQP4 subcellular polarization in reactive astrocytes and would promote Aβ excretion possibly through increasing AQP4-mediated intracerebral water flux in the brain in AD.
Collapse
Affiliation(s)
- Kana Sasaki
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroki Fujita
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan.
| | - Takehiro Sato
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan
| | - Shunske Kato
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan; Center for Medical Education and Training, Akita University Hospital, Akita, Japan
| | - Yuya Takahashi
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yukio Takeshita
- Blood-Brain Barrier Research Center, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Takashi Kanda
- Department of Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan; Neuromuscular Center Yoshimizu Hospital, Shimonoseki, Yamaguchi, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan; Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takamori C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Satoko Hattori
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, Japan; Research Creation Support Center, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yasukazu Hozumi
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuichiro Yamada
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan; Center for Diabetes, Endocrinology and Metabolism, Kansai Electric Power Hospital, Osaka, Japan; Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto, Japan.
| | - Hironori Waki
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan.
| |
Collapse
|
29
|
Shang Y, Yu L, Xing H, Chang Y, Dong K, Xiao Y, Liu Y, Feng M, Qin Y, Dai H. Diffusion Tensor Imaging Analysis Along the Perivascular Space (DTI-ALPS) Demonstrates That Sleep Disorders Exacerbate Glymphatic Circulatory Impairment and Cognitive Impairment in Patients with Alzheimer's Disease. Nat Sci Sleep 2024; 16:2205-2215. [PMID: 39735385 PMCID: PMC11675307 DOI: 10.2147/nss.s496607] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/15/2024] [Indexed: 12/31/2024] Open
Abstract
Objective Sleep disorders are common in Alzheimer's disease (AD) patients and can impair the glymphatic system, leading to cognitive decline. This study aimed to investigate whether AD patients with sleep disorders exhibit worse glymphatic function and more severe cognitive impairment compared to those without sleep disorders and to explore the underlying molecular imaging mechanisms. Methods This study included 40 AD patients with sleep disorders (ADSD), 39 cognitively matched AD patients without sleep disorders (ADNSD), and 25 healthy middle-aged and elderly controls (NC). Participants underwent functional magnetic resonance imaging (fMRI), and cognitive and sleep assessments. The ALPS (Along the Perivascular Space) index was calculated, followed by intergroup comparisons, correlation analyses, and mediation analyses. The diagnostic utility of the ALPS index was assessed using a receiver operating characteristic (ROC) curve. Results The ALPS index was lower in the ADNSD and ADSD groups compared to the NC group. In the ADSD group, PSQI scores were negatively correlated with MMSE scores. The ALPS index was positively correlated with MMSE scores and negatively with PSQI scores. Mediation analyses indicated that the ALPS index partially mediated the effect of sleep disturbances on cognitive impairment (indirect effect = -0.134; mediation effect = 30.505%). The area under the ROC curve (AUROC) for distinguishing ADSD from ADNSD was 0.86, with a cutoff ALPS index value 1.309. Conclusion Sleep disorders worsen glymphatic function and cognitive impairment in AD patients. The ALPS index partially mediates the impact of sleep disorders on cognitive function and shows moderate accuracy in distinguishing between patients with ADSD and ADNSD.
Collapse
Affiliation(s)
- Yi Shang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Lefan Yu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Hanqi Xing
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Yue Chang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Ke Dong
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Yao Xiao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Yuanqing Liu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Mengmeng Feng
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Yiren Qin
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Hui Dai
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
- Institute of Medical Imaging, Soochow University, Suzhou, 215006, People’s Republic of China
| |
Collapse
|
30
|
Engelhardt E, Resende EDPF, Gomes KB. Physiopathological mechanisms underlying Alzheimer's disease: a narrative review. Dement Neuropsychol 2024; 18:e2024VR01. [PMID: 39697643 PMCID: PMC11654088 DOI: 10.1590/1980-5764-dn-2024-vr01] [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: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 12/20/2024] Open
Abstract
The neuropathological signature of Alzheimer's disease (AD) comprises mainly amyloid plaques, and neurofibrillary tangles, resulting in synaptic and neuronal loss. These pathological structures stem from amyloid dysfunctional metabolism according to the amyloid cascade hypothesis, leading to the formation of plaques, and apparently inducing the initiation of the abnormal tau pathway, with phosphorylation and aggregation of these proteins, ultimately causing the formation of tangles. In this narrative review, the existing hypothesis related to the pathophysiology of AD were compiled, and biological pathways were highlighted in order to identify the molecules that could represent biological markers of the disease, necessary to establish early diagnosis, as well as the selection of patients for therapeutical interventional strategies.
Collapse
Affiliation(s)
- Eliasz Engelhardt
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo Horizonte MG, Brazil
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte MG, Brazil
| | - Karina Braga Gomes
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Belo Horizonte MG, Brazil
| |
Collapse
|
31
|
Regalbuto S, Zangaglia R, Valentino F, Todisco M, Pacchetti C, Cotta Ramusino M, Mazzacane F, Picascia M, Arceri S, Malomo G, Capriglia E, Spelta L, Rubino A, Pisani A, Terzaghi M. Clinical correlates of obstructive sleep apnoea in idiopathic normal pressure hydrocephalus. Eur J Neurol 2024; 31:e16448. [PMID: 39207116 PMCID: PMC11555027 DOI: 10.1111/ene.16448] [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: 05/28/2024] [Revised: 07/30/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND PURPOSE The pathogenesis of idiopathic normal pressure hydrocephalus (iNPH) remains controversial. Limited studies have indicated a high prevalence of obstructive sleep apnoea (OSA) amongst iNPH patients. The aim was to investigate the clinical correlates of OSA in iNPH patients. METHODS In this cross-sectional observational study, consecutive iNPH patients were prospectively enrolled. Evaluations included the iNPH Rating Scale, the Movement Disorder Society Unified Parkinson's Disease Rating Scale part III, the time and number of steps to walk 10 m, the Epworth Sleepiness Scale, the Pittsburgh Sleep Quality Index, a complete neuropsychological evaluation, 3-T brain MRI, full-night video-polysomnography, tap test and cerebrospinal fluid (CSF) neurodegeneration biomarkers. RESULTS Fifty-one patients were screened, of whom 38 met the inclusion criteria. Amongst the recruited patients, 19/38 (50%) exhibited OSA, with 12/19 (63.2%) presenting moderate to severe disorder. OSA+ iNPH patients required more time (p = 0.02) and more steps (p = 0.04) to complete the 10-m walking test, had lower scores on the gait subitem of the iNPH Rating Scale (p = 0.04) and demonstrated poorer performance on specific neuropsychological tests (Rey Auditory Verbal Learning Test immediate recall, p = 0.03, and Rey-Osterrieth Complex Figure, p = 0.01). Additionally, OSA+ iNPH patients had higher levels of total tau (p = 0.02) and phospho-tau (p = 0.03) in their CSF but no statistically significant differences in beta-amyloid (1-42) levels compared to OSA- iNPH patients. CONCLUSION Obstructive sleep apnoea is highly prevalent in iNPH patients, particularly at moderate to severe levels. OSA is associated with worse motor and cognitive performance in iNPH. The CSF neurodegeneration biomarker profile observed in OSA+ iNPH patients may reflect OSA-induced impairment of cerebral fluid dynamics.
Collapse
Affiliation(s)
- Simone Regalbuto
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
| | | | | | | | | | - Matteo Cotta Ramusino
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Federico Mazzacane
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Marta Picascia
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
| | | | - Gaetano Malomo
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Elena Capriglia
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Laura Spelta
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
| | - Annalisa Rubino
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
| | - Antonio Pisani
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| | - Michele Terzaghi
- IRCCS Mondino FoundationNational Neurological InstitutePaviaItaly
- Department of Brain and Behavioural SciencesUniversity of PaviaPaviaItaly
| |
Collapse
|
32
|
Han F, Lee J, Chen X, Ziontz J, Ward T, Landau SM, Baker SL, Harrison TM, Jagust WJ. Global brain activity and its coupling with cerebrospinal fluid flow is related to tau pathology. Alzheimers Dement 2024; 20:8541-8555. [PMID: 39508716 PMCID: PMC11667553 DOI: 10.1002/alz.14296] [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: 04/19/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024]
Abstract
INTRODUCTION Factors responsible for the deposition of pathological tau in the brain are incompletely understood. This study links macroscale tau deposition in the human brain to cerebrospinal fluid (CSF) flow dynamics using resting-state functional magnetic resonance imaging (rsfMRI). METHODS Low-frequency (< 0.1 Hz) resting-state global brain activity is coupled with CSF flow and potentially reflects CSF dynamics-related clearance. We examined the correlation between rsfMRI measures of CSF inflow and global activity (gBOLD-CSF coupling) as a predictor, interacting with amyloid beta (Aβ), of tau and cortical thickness (dependent variables) across Alzheimer's Disease Neuroimaging Initiative (ADNI) participants from cognitively unimpaired through mild cognitive impairment (MCI) and Alzheimer's disease (AD). RESULTS Tau deposition in Aβ+ participants, accompanied by cortical thinning and cognitive decline, is associated with decreased gBOLD-CSF coupling. Tau mediates the relationship between coupling and thickness. DISCUSSION Findings suggest that resting-state global brain activity and CSF movements comodulate Alzheimer's tau deposition, presumably related to CSF clearance. HIGHLIGHTS A non-invasive functional magnetic resonance imaging (fMRI) assessment of a CSF clearance-related process is carried out. Global brain activity is coupled with CSF inflow in human fMRI during resting state. Global fMRI-CSF coupling is correlated with tau in Alzheimer's disease (AD). This coupling measure is also associated with cortical thickness, mediated by tau.
Collapse
Affiliation(s)
- Feng Han
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - JiaQie Lee
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Xi Chen
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
- Department of Cellular and Tissue ImagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Jacob Ziontz
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Tyler Ward
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Susan M. Landau
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Suzanne L. Baker
- Department of Cellular and Tissue ImagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | | | - William J. Jagust
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
- Department of Cellular and Tissue ImagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | | |
Collapse
|
33
|
Eisenbaum M, Bachmeier C. Contribution of astrocytes to the neurovascular elimination of tau. Neural Regen Res 2024; 19:2559-2560. [PMID: 38808980 PMCID: PMC11168527 DOI: 10.4103/nrr.nrr-d-23-01705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 05/30/2024] Open
Affiliation(s)
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA
- Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
34
|
Rane Levendovszky S, Flores J, Peskind ER, Václavů L, van Osch MJP, Iliff J. Preliminary investigations into human neurofluid transport using multiple novel non-contrast MRI methods. J Cereb Blood Flow Metab 2024; 44:1580-1592. [PMID: 39053490 PMCID: PMC11572104 DOI: 10.1177/0271678x241264407] [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: 07/10/2023] [Revised: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 07/27/2024]
Abstract
We discuss two potential non-invasive MRI methods to study phenomena related to subarachnoid cerebrospinal fluid (CSF) motion and perivascular fluid transport, and their association with sleep and aging. We apply diffusion-based intravoxel incoherent motion (IVIM) imaging to evaluate pseudodiffusion coefficient, D*, or CSF movement across large spaces like the subarachnoid space (SAS). We also performed perfusion-based multi-echo, Hadamard encoded arterial spin labeling (ASL) to evaluate whole brain cortical cerebral blood flow (CBF) and trans-endothelial exchange (Tex) of water from the vasculature into the perivascular space and parenchyma. Both methods were used in young adults (N = 9, 6 F, 23 ± 3 years old) in the setting of sleep and sleep deprivation. To study aging, 10 older adults (6 F, 67 ± 3 years old) were imaged after a night of normal sleep and compared with the young adults. D* in SAS was significantly (p < 0.05) reduced with sleep deprivation (0.016 ± 0.001 mm2/s) compared to normal sleep (0.018 ± 0.001 mm2/s) and marginally reduced with aging (0.017 ± 0.001 mm2/s, p = 0.029). Cortical CBF and Tex were unchanged with sleep deprivation but significantly lower in older adults (37 ± 3 ml/100 g/min, 578 ± 61 ms) than in young adults (42 ± 2 ml/100 g/min, 696 ± 62 ms). IVIM was sensitive to sleep physiology and aging, and multi-echo, multi-delay ASL was sensitive to aging.
Collapse
Affiliation(s)
| | - Jaqueline Flores
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Elaine R Peskind
- VISN 20 Mental Illness Research, Education, and Clinical Center, Veterans Affairs Puget Sound Healthcare System, Seattle, WA, USA
| | - Lena Václavů
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias JP van Osch
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeffrey Iliff
- VISN 20 Mental Illness Research, Education, and Clinical Center, Veterans Affairs Puget Sound Healthcare System, Seattle, WA, USA
- Department of Psychiatry and Behavioral Science, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
35
|
Singh V, Rochakim N, Ferraresso F, Choudhury A, Kastrup CJ, Ahn HJ. Caveolin-1 and Aquaporin-4 as Mediators of Fibrinogen-Driven Cerebrovascular Pathology in Hereditary Cerebral Amyloid Angiopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.623066. [PMID: 39605467 PMCID: PMC11601418 DOI: 10.1101/2024.11.11.623066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hereditary Cerebral Amyloid Angiopathy (HCAA) is a rare inherited form of CAA, characterized by increased vascular deposits of amyloid peptides. HCAA provides a unique opportunity to study the pathogenic mechanisms linked to CAA, as it is associated with severe cerebrovascular pathology. Some of HCAA-associated amyloid-β (Aβ) mutations significantly enhance the interaction between fibrinogen and Aβ, resulting in altered fibrin structure and co-deposition with Aβ in the perivascular space. However, the mechanisms underlying perivascular fibrinogen deposition and the associated cerebrovascular pathology in HCAA remain unclear. To investigate this, we analyzed TgSwDI transgenic mice carrying HCAA-associated mutations and observed a significant age-dependent increase in fibrin(ogen) extravasation and fibrin(ogen)-Aβ colocalization in the perivascular space. Moreover, Caveolin-1, a protein involved in non-specific transcytosis across the endothelium, significantly increased with age in TgSwDI mice and correlated with fibrin(ogen) extravasation. Additionally, we noted significant aquaporin-4 (AQP4) depolarization in the CAA-laden blood vessels of TgSwDI mice, which also correlated with fibrin(ogen) extravasation and fibrin(ogen)-Aβ colocalization. Given that AQP4 plays a crucial role in Aβ clearance via the glymphatic pathway, its depolarization may disrupt this critical clearance mechanism, thereby exacerbating CAA pathology. To further explore the relationship between fibrin(ogen) and these factors, we depleted fibrinogen in TgSwDI mice using siRNA against fibrinogen. This intervention resulted in decreased CAA, reduced caveolin-1 levels, attenuated microglial activation, restored polarized expression of AQP4, and improved spatial memory in fibrinogen-depleted TgSwDI mice. These findings suggest that targeting fibrinogen could be a promising strategy for mitigating CAA pathology and its associated cerebrovascular pathology. Significance Statement Our study reveals the mechanism by which fibrin(ogen)-Aβ colocalization could exacerbates CAA pathology. Our findings highlight that the age-dependent increase of endothelial caveolin-1 could facilitate fibrin(ogen) extravasation, which binds with Aβ in the perivascular space inducing microglial neuroinflammation and AQP4 depolarization, thus exacerbating CAA pathology. Furthermore, fibrinogen depletion could mitigate CAA severity, reduce microglial activation, restore AQP4 polarization and memory impairment. These results suggest that targeting fibrinogen and caveolin-1-mediated transcytosis may offer new strategies to address CAA-associated cerebrovascular pathology.
Collapse
|
36
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [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: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
37
|
Kato D, Kameda H, Kinota N, Fujii T, Xiawei B, Simi Z, Takai Y, Chau S, Miyasaka Y, Mashimo T, Abe Y, Yasui M, Minowa K, Kudo K. Loss of aquaporin-4 impairs cerebrospinal fluid solute clearance through cerebrospinal fluid drainage pathways. Sci Rep 2024; 14:27982. [PMID: 39543281 PMCID: PMC11564557 DOI: 10.1038/s41598-024-79147-y] [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: 08/07/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
The aquaporin-4 (AQP4) water channel is essential in neurofluid dynamics. AQP4 loss impairs solute exchange between the cerebrospinal fluid (CSF) and interstitial fluid (ISF). However, whether AQP4 expression affects solute clearance from the CSF space to the extracranial space remains unclear. This study aimed to investigate this using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) following the intrathecal administration of gadolinium-based contrast agents (GBCAs) to AQP4 knockout (KO) rats. AQP4 KO rats showed reduced efflux of intrathecal GBCAs to the extracranial spaces through CSF drainage pathways and increased retention of intrathecal GBCAs in the CSF space compared with the controls. These results suggest that AQP4 loss impairs solute clearance from the CSF space to the extracranial spaces via the CSF drainage pathways. This study revealed a close relationship between AQP4 expression and CSF solute clearance, contributing to a better understanding of the function of AQP4 in neurofluid dynamics.
Collapse
Affiliation(s)
- Daisuke Kato
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Kameda
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan.
- Department of Radiology, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan.
| | - Naoya Kinota
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takaaki Fujii
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Bai Xiawei
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Zhou Simi
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshiki Takai
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Pathology, Tohoku University Hospital, Sendai, Japan
| | - Simon Chau
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiki Miyasaka
- Laboratory of Reproductive Engineering, Institute of Experimental Animal Sciences, Osaka University Medical School, Suita, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan.
| | - Masato Yasui
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyuki Minowa
- Department of Radiology, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Kohsuke Kudo
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
38
|
Studart-Neto A, Barbosa BJAP, Coutinho AM, de Souza LC, Schilling LP, da Silva MNM, Castilhos RM, Bertolucci PHF, Borelli WV, Gomes HR, Fernandes GBP, Barbosa MT, Balthazar MLF, Frota NAF, Forlenza OV, Smid J, Brucki SMD, Caramelli P, Nitrini R, Engelhardt E, Resende EDPF. Guidelines for the use and interpretation of Alzheimer's disease biomarkers in clinical practice in Brazil: recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2024; 18:e2024C001. [PMID: 39534442 PMCID: PMC11556292 DOI: 10.1590/1980-5764-dn-2024-c001] [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: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, the diagnostic accuracy of Alzheimer's disease has been enhanced by the development of different types of biomarkers that indicate the presence of neuropathological processes. In addition to improving patient selection for clinical trials, biomarkers can assess the effects of new treatments on pathological processes. However, there is concern about the indiscriminate and poorly supported use of biomarkers, especially in asymptomatic individuals or those with subjective cognitive decline. Difficulties interpreting these tests, high costs, and unequal access make this scenario even more challenging in healthcare. This article presents the recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology (Departamento Científico de Neurologia Cognitiva e Envelhecimento da Academia Brasileira de Neurologia) regarding the rational use and interpretation of Alzheimer's disease biomarkers in clinical practice. The clinical diagnosis of cognitive-behavioral syndrome is recommended as the initial step to guide the request for biomarkers.
Collapse
Affiliation(s)
- Adalberto Studart-Neto
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Breno José Alencar Pires Barbosa
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Pernambuco, Hospital das Clínicas, Recife, Centro de Ciências Médicas, Recife PE, Brazil
- Universidade Federal de Pernambuco, Empresa Brasileira de Serviços Hospitalares, Hospital das Clínicas, Departamento de Neurologia, Recife PE, Brazil
| | - Artur Martins Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Radiologia, Centro de Medicina Nuclear, Laboratório de Investigação Médica (LIM 43), São Paulo SP, Brazil
- Hospital Sírio-Libanês, Medicina Nuclear e Serviço de PET-CT, São Paulo SP, Brazil
| | - Leonardo Cruz de Souza
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Lucas Porcello Schilling
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil
| | - Mari Nilva Maia da Silva
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Nina Rodrigues, Serviço de Neuropsiquiatria, São Luís MA, Brazil
| | - Raphael Machado Castilhos
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Centro de Neurologia Cognitiva e Comportamental, Porto Alegre RS, Brazil
| | - Paulo Henrique Ferreira Bertolucci
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| | - Wyllians Vendramini Borelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Ciências Morfológicas, Porto Alegre RS, Brazil
| | - Hélio Rodrigues Gomes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Laboratório de Líquido Cefalorraquidiano, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Laboratório de Investigação Médica (LIM 15), São Paulo SP, Brazil
- Departamento Científico de Líquido Cefalorraquiano, Academia Brasileira de Neurologia, São Paulo SP, Brazil
| | | | - Maira Tonidandel Barbosa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Marcio Luiz Figueredo Balthazar
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil
| | - Norberto Anízio Ferreira Frota
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil
- Universidade de Fortaleza, Fortaleza, CE, Brazil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brazil
| | - Jerusa Smid
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Sonia Maria Dozzi Brucki
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Paulo Caramelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Ricardo Nitrini
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Eliasz Engelhardt
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Psiquiatria, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| |
Collapse
|
39
|
Leipp F, Vialaret J, Mohaupt P, Coppens S, Jaffuel A, Niehoff AC, Lehmann S, Hirtz C. Glial fibrillary acidic protein in Alzheimer's disease: a narrative review. Brain Commun 2024; 6:fcae396. [PMID: 39554381 PMCID: PMC11568389 DOI: 10.1093/braincomms/fcae396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/10/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Astrocytes are fundamental in neural functioning and homeostasis in the central nervous system. These cells respond to injuries and pathological conditions through astrogliosis, a reactive process associated with neurodegenerative diseases such as Alzheimer's disease. This process is thought to begin in the early stages of these conditions. Glial fibrillary acidic protein (GFAP), a type III intermediate filament protein predominantly expressed in astrocytes, has emerged as a key biomarker for monitoring this response. During astrogliosis, GFAP is released into biofluids, making it a candidate for non-invasive diagnosis and tracking of neurodegenerative diseases. Growing evidence positions GFAP as a biomarker for Alzheimer's disease with specificity and disease-correlation characteristics comparable to established clinical markers, such as Aβ peptides and phosphorylated tau protein. To improve diagnostic accuracy, particularly in the presence of confounders and comorbidities, incorporating a panel of biomarkers may be advantageous. This review will explore the potential of GFAP within such a panel, examining its role in early diagnosis, disease progression monitoring and its integration into clinical practice for Alzheimer's disease management.
Collapse
Affiliation(s)
- Florine Leipp
- Shimadzu France SAS France, Noisiel, France
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Jérôme Vialaret
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Pablo Mohaupt
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Salomé Coppens
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | | | | | - Sylvain Lehmann
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Christophe Hirtz
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| |
Collapse
|
40
|
Parra Bravo C, Naguib SA, Gan L. Cellular and pathological functions of tau. Nat Rev Mol Cell Biol 2024; 25:845-864. [PMID: 39014245 DOI: 10.1038/s41580-024-00753-9] [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: 06/10/2024] [Indexed: 07/18/2024]
Abstract
Tau protein is involved in various cellular processes, including having a canonical role in binding and stabilization of microtubules in neurons. Tauopathies are neurodegenerative diseases marked by the abnormal accumulation of tau protein aggregates in neurons, as seen, for example, in conditions such as frontotemporal dementia and Alzheimer disease. Mutations in tau coding regions or that disrupt tau mRNA splicing, tau post-translational modifications and cellular stress factors (such as oxidative stress and inflammation) increase the tendency of tau to aggregate and interfere with its clearance. Pathological tau is strongly implicated in the progression of neurodegenerative diseases, and the propagation of tau aggregates is associated with disease severity. Recent technological advancements, including cryo-electron microscopy and disease models derived from human induced pluripotent stem cells, have increased our understanding of tau-related pathology in neurodegenerative conditions. Substantial progress has been made in deciphering tau aggregate structures and the molecular mechanisms that underlie protein aggregation and toxicity. In this Review, we discuss recent insights into the diverse cellular functions of tau and the pathology of tau inclusions and explore the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sarah A Naguib
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
41
|
Wang Y, Lei K, Zhao L, Zhang Y. Clinical glycoproteomics: methods and diseases. MedComm (Beijing) 2024; 5:e760. [PMID: 39372389 PMCID: PMC11450256 DOI: 10.1002/mco2.760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Glycoproteins, representing a significant proportion of posttranslational products, play pivotal roles in various biological processes, such as signal transduction and immune response. Abnormal glycosylation may lead to structural and functional changes of glycoprotein, which is closely related to the occurrence and development of various diseases. Consequently, exploring protein glycosylation can shed light on the mechanisms behind disease manifestation and pave the way for innovative diagnostic and therapeutic strategies. Nonetheless, the study of clinical glycoproteomics is fraught with challenges due to the low abundance and intricate structures of glycosylation. Recent advancements in mass spectrometry-based clinical glycoproteomics have improved our ability to identify abnormal glycoproteins in clinical samples. In this review, we aim to provide a comprehensive overview of the foundational principles and recent advancements in clinical glycoproteomic methodologies and applications. Furthermore, we discussed the typical characteristics, underlying functions, and mechanisms of glycoproteins in various diseases, such as brain diseases, cardiovascular diseases, cancers, kidney diseases, and metabolic diseases. Additionally, we highlighted potential avenues for future development in clinical glycoproteomics. These insights provided in this review will enhance the comprehension of clinical glycoproteomic methods and diseases and promote the elucidation of pathogenesis and the discovery of novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yujia Wang
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Kaixin Lei
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Lijun Zhao
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Yong Zhang
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
42
|
Sun M, Zhang M, Di F, Bai W, Sun J, Zhang M, Sun J, Li M, Liang X. Polystyrene nanoplastics induced learning and memory impairments in mice by damaging the glymphatic system. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116874. [PMID: 39153278 DOI: 10.1016/j.ecoenv.2024.116874] [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: 04/25/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
The excessive usage of nanoplastics (NPs) has posed a serious threat to the ecological environment and human health, which can enter the brain and then result in neurotoxicity. However, research on the neurotoxic effects of NPs based on different exposure routes and modifications of functional groups is lacking. In this study, the neurotoxicity induced by NPs was studied using polystyrene nanoplastics (PS-NPs) of different modifications (PS, PS-COOH, and PS-NH2). It was found that PS-NH2 through intranasal administration (INA) exposure route exhibited the greatest accumulation in the mice brain after exposure for 7 days. After the mice were exposed to PS-NH2 by INA means for 28 days, the exploratory ability and spatial learning ability were obviously damaged in a dose-dependent manner. Further analysis indicated that these damages induced by PS-NH2 were closely related to the decreased ability of glymphatic system to clear β-amyloid (Aβ) and phosphorylated Tau (P-Tau) proteins, which was ascribed to the loss of aquaporin-4 (AQP4) polarization in the astrocytic endfeet. Moreover, the loss of AQP4 polarization might be regulated by the NF-κB pathway. Our current study establishes the connection between the neurotoxicity induced by PS-NPs and the glymphatic system dysfunction for the first time, which will contribute to future research on the neurotoxicity of NPs.
Collapse
Affiliation(s)
- Meng Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China; School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Min Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fanglin Di
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Weijie Bai
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Jikui Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Mingkun Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Jinlong Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Meng Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China.
| | - Xue Liang
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
43
|
Yuan Y, Peng W, Lei J, Zhao Y, Zhao B, Li Y, Wang J, Qu Q. AQP4 Endocytosis-Lysosome Degradation Mediated by MMP-9/β-DG Involved in Diabetes Cognitive Impairment. Mol Neurobiol 2024; 61:8438-8453. [PMID: 38512439 DOI: 10.1007/s12035-024-04085-9] [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: 11/23/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024]
Abstract
Cognitive impairment is considered to be one of the important comorbidities of diabetes, but the underlying mechanisms are widely unknown. Aquaporin-4 (AQP4) is the most abundant water channel in the central nervous system, which plays a neuroprotective role in various neurological diseases by maintaining the function of glymphatic system and synaptic plasticity. However, whether AQP4 is involved in diabetes-related cognitive impairment remains unknown. β-dystroglycan (β-DG), a key molecule for anchoring AQP4 on the plasma membrane of astrocytes and avoiding its targeting to lysosomes for degradation, can be cleaved by matrix metalloproteinase-9 (MMP-9). β-DG deficiency can cause a decline in AQP4 via regulating its endocytosis. However, whether cleavage of β-DG can affect the expression of AQP4 remains unreported. In this study, we observed that diabetes mice displayed cognitive disorder accompanied by reduction of AQP4 in prefrontal cortex. And we found that bafilomycin A1, a widely used lysosome inhibitor, could reverse the downregulation of AQP4 in diabetes, further demonstrating that the reduction of AQP4 in diabetes is a result of more endocytosis-lysosome degradation. In further experiments, we found diabetes caused the excessive activation of MMP-9/β-DG which leaded to the loss of connection between AQP4 and β-DG, further inducing the endocytosis of AQP4. Moreover, inhibition of MMP-9/β-DG restored the endocytosis-lysosome degradation of AQP4 and partially alleviated cognitive dysfunction in diabetes. Our study sheds new light on the role of AQP4 in diabetes-associated cognitive disorder. And we provide a promising therapeutic target to reverse the endocytosis-lysosome degradation of AQP4 in diabetes, such as MMP-9/β-DG.
Collapse
Affiliation(s)
- Ye Yuan
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Wei Peng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Jingna Lei
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Yi Zhao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Beiyu Zhao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Yan Li
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China.
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China.
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
44
|
Burgos AF, Olson PA, Vgontzas A. The Glymphatic System and its Relationship to Migraine. Curr Neurol Neurosci Rep 2024; 24:517-525. [PMID: 39150650 DOI: 10.1007/s11910-024-01368-5] [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] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE OF REVIEW We aim to critically review animal and human studies of the glymphatic system in migraine and propose a model for how the glymphatic system may function in migraine, based on the available evidence. RECENT FINDINGS Early studies in animal models report migraine attacks temporarily disrupt glymphatic flow. Human imaging studies suggest chronic migraine may be associated with alterations in glymphatic system function, albeit with conflicting results. Presently, it remains unknown whether repetitive migraine attacks or frequent nights of insomnia impair glymphatic system function over time in those with migraine, and whether alterations in glymphatic function could contribute to worsening migraine disability or risk for cognitive disease. Longitudinal studies of glymphatic function in patients with migraine and insomnia, with inclusion of cognitive assessments, may be informative.
Collapse
Affiliation(s)
| | - Patricia A Olson
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Angeliki Vgontzas
- Graham Headache Center, Department of Neurology, Brigham and Women's Faulkner Hospital, Harvard Medical School, 1153 Centre Street Suite 4H, Boston, MA, 02130, USA.
| |
Collapse
|
45
|
Cunningham HA, Dovek L, Recoder N, Bryant-Ekstrand MD, Ligman BR, Piantino J, Lim MM, Elliott JE. Heart rate variability impairment during sleep in Veterans with REM sleep behavior disorder, traumatic brain injury, and posttraumatic stress disorder: An early potential window into autonomic dysfunction? BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.20.614142. [PMID: 39386663 PMCID: PMC11463592 DOI: 10.1101/2024.09.20.614142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Individuals with comorbid REM sleep behavior disorder (RBD) and neurotrauma (defined by traumatic brain injury and post-traumatic stress disorder) have an earlier age of RBD symptom onset, increased RBD-related symptom severity and more neurological features indicative of prodromal synucleinopathy compared to RBD only. An early sign of neurodegenerative condition is autonomic dysfunction, which we sought to evaluate by examining heart rate variability during sleep. Participants with overnight polysomnography were recruited from the VA Portland Health Care System. Veterans without neurotrauma or RBD (controls; n=19), with RBD only (RBD, n=14), and with RBD and neurotrauma (RBD+NT, n=19) were evaluated. Eligible 5-minute NREM and REM epochs without apneas/hypopneas, microarousals, and ectopic beats were analyzed for frequency and time domain (e.g. low frequency power, LF; high frequency power, HF; root mean square of successive RR intervals, RMSSD; % of RR intervals that vary ≥50 ms, pNN50) heart rate variability outcomes. Heart rate did not significantly differ between groups in any sleep stage. Time domain and frequency domain variables (e.g., LF power, HF power, RMSSD, and pNN50) were significantly reduced in the RBD and RBD+NT groups compared to controls and RBD only during NREM sleep. There were no group differences detected during REM sleep. These data suggest significant reductions in heart rate variability during NREM sleep in RBD+NT participants, suggesting greater autonomic dysfunction compared to controls or RBD alone. Heart rate variability during sleep may be an early, promising biomarker, yielding mechanistic insight for diagnosis and prognosis of early neurodegeneration in this vulnerable population. STATEMENT OF SIGNIFICANCE Comorbid REM sleep behavior disorder (RBD) and neurotrauma (NT, traumatic brain injury + post-traumatic stress disorder; RBD+NT) is associated with increased neurodegenerative symptom burden and worsened health. Sleep and autonomic function are integrally and bidirectionally related to neurodegenerative processes. In the current study, we sought to determine if early signs of autonomic dysfunction, measured via heart rate variability (HRV), were present during sleep in comorbid RBD+NT compared to RBD only and controls. Our data show reduced time and frequency domain HRV during NREM sleep in RBD+NT Veterans compared to RBD only and controls. These data contribute evidence that participants with RBD and comorbid NT demonstrate significantly worse autonomic dysfunction compared to age/sex matched participants with RBD alone.
Collapse
|
46
|
Xiao D, Li J, Ren Z, Dai M, Jiang Y, Qiu T, Zhang H, Chen Y, Zhang Y, Zhang Y, Palaniyappan L. Association of cortical morphology, white matter hyperintensity, and glymphatic function in frontotemporal dementia variants. Alzheimers Dement 2024; 20:6045-6059. [PMID: 39129270 PMCID: PMC11497707 DOI: 10.1002/alz.14158] [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: 01/17/2024] [Revised: 04/13/2024] [Accepted: 05/25/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Frontotemporal dementia (FTD) can be phenotypically divided into behavioral variant FTD (bvFTD), nonfluent variant primary progressive aphasia (nfvPPA), and semantic variant PPA (svPPA). However, the neural underpinnings of this phenotypic heterogeneity remain elusive. METHODS Cortical morphology, white matter hyperintensities (WMH), diffusion tensor image analysis along the perivascular space (DTI-ALPS), and their interrelationships were assessed in subtypes of FTD. Neuroimaging-transcriptional analyses on the regional cortical morphological deviances among subtypes were also performed. RESULTS Changes in cortical thickness, surface area, gyrification, WMH, and DTI-ALPS were subtype-specific in FTD. The three morphologic indices are related to whole-brain WMH volume and cognitive performance, while cortical thickness is related to DTI-ALPS. Neuroimaging-transcriptional analyses identified key biological pathways linked to the formation and/or spread of TDP-43/tau pathologies. DISCUSSION We found subtype-specific changes in cortical morphology, WMH, and glymphatic function in FTD. Our findings have the potential to contribute to the development of personalized predictions and treatment strategies for this disorder. HIGHLIGHTS Cortical morphologic changes, white matter hyperintensities (WMH), and glymphatic dysfunction are subtype-specific. Cortical morphologic changes, WMH, and glymphatic dysfunction are inter-correlated. Cortical morphologic changes and WMH burden contribute to cognitive impairments.
Collapse
Affiliation(s)
- Die Xiao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Jianyu Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Zhanbing Ren
- College of Physical Education, Shenzhen UniversityShenzhenP. R. China
| | - Minghui Dai
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Yihan Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Ting Qiu
- Douglas Mental Health University InstituteMcGill UniversityMontrealCanada
| | - Huixiong Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Yifan Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Youming Zhang
- Department of RadiologyXiangya HospitalCentral South UniversityChangshaP. R. China
- National Clinical Research Center for Geriatric DiseasesXiangya HospitalCentral South UniversityChangshaHunanP. R. China
| | - Yuanchao Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Lena Palaniyappan
- Douglas Mental Health University InstituteMcGill UniversityMontrealCanada
| | | |
Collapse
|
47
|
Guo Y, Wu L, Liu J, Liu J, Sun Z. Correlation between glymphatic dysfunction and cranial defect in severe traumatic brain injury: a retrospective case-control study based on a diffusion tensor image analysis along the perivascular space (DTI-ALPS) investigation. Quant Imaging Med Surg 2024; 14:6756-6766. [PMID: 39281142 PMCID: PMC11400707 DOI: 10.21037/qims-24-348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/17/2024] [Indexed: 09/18/2024]
Abstract
Background To date, limited research has been conducted on the functionality of the glymphatic system during the recovery phase of severe traumatic brain injury (sTBI). This study aimed to use a diffusion tensor image analysis along the perivascular space (DTI-ALPS) to evaluate glymphatic system function in patients recovering from sTBI who underwent unilateral decompressive craniectomy, and to examine the correlation between the ALPS index and the size of the cranial defect. We hypothesized that assessments would reveal ongoing impairments in glymphatic system function among sTBI patients during the recovery phase. Methods A total of 23 patients with a history of sTBI who had previously undergone unilateral decompressive craniectomy at Xiangya Hospital of Central South University from January 2020 to December 2020 were enrolled in the study, along with 33 healthy control (HC) subjects. All the subjects underwent magnetic resonance imaging (MRI) with DTI scans, and the ALPS index was subsequently calculated to assess glymphatic system functionality. Additionally, the circumference and sectional area of the cranial defect were measured for each patient. An analysis of variance (ANOVA) was used to compare the ALPS index values between the sTBI patients and HC subjects, while a Pearson correlation analysis was used to examine the correlation between the ALPS index and cranial defect characteristics. Results The ALPS index values of both the craniectomy side (t=-9.08, P<0.001) and non-craniectomy side (t=-5.06, P<0.001) of the sTBI group were significantly lower than those of the HC group. However, no statistically significant differences were observed between the ALPS index values of the craniectomy and non-craniectomy sides. Additionally, no significant differences were observed in the ALPS index values of both the craniectomy and non-craniectomy sides among the early, intermediate, and late recovery phases. In the sTBI patients, a moderately strong negative correlation was found between the circumference of the cranial defect and the ALPS index of the craniectomy side (r=-0.62, P=0.002), and a moderately negative correlation was found between the sectional area of the cranial defect and the ALPS index of the craniectomy side (r=-0.56, P=0.005). Conclusions The non-invasive DTI-ALPS technique revealed significantly reduced ALPS index values during the recovery phase of sTBI, indicating persistent impairment in glymphatic system function. A significant negative correlation was found between the ALPS index value of the craniectomy side and the size of the cranial defect. These findings suggest that the ALPS index may serve as a valuable prognostic factor in the recovery phase of sTBI.
Collapse
Affiliation(s)
- Yong Guo
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Lin Wu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jiacheng Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jinfang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhongyi Sun
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
48
|
Yue Y, Zhang X, Lv W, Lai HY, Shen T. Interplay between the glymphatic system and neurotoxic proteins in Parkinson's disease and related disorders: current knowledge and future directions. Neural Regen Res 2024; 19:1973-1980. [PMID: 38227524 DOI: 10.4103/1673-5374.390970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/26/2023] [Indexed: 01/17/2024] Open
Abstract
Parkinson's disease is a common neurodegenerative disorder that is associated with abnormal aggregation and accumulation of neurotoxic proteins, including α-synuclein, amyloid-β, and tau, in addition to the impaired elimination of these neurotoxic protein. Atypical parkinsonism, which has the same clinical presentation and neuropathology as Parkinson's disease, expands the disease landscape within the continuum of Parkinson's disease and related disorders. The glymphatic system is a waste clearance system in the brain, which is responsible for eliminating the neurotoxic proteins from the interstitial fluid. Impairment of the glymphatic system has been proposed as a significant contributor to the development and progression of neurodegenerative disease, as it exacerbates the aggregation of neurotoxic proteins and deteriorates neuronal damage. Therefore, impairment of the glymphatic system could be considered as the final common pathway to neurodegeneration. Previous evidence has provided initial insights into the potential effect of the impaired glymphatic system on Parkinson's disease and related disorders; however, many unanswered questions remain. This review aims to provide a comprehensive summary of the growing literature on the glymphatic system in Parkinson's disease and related disorders. The focus of this review is on identifying the manifestations and mechanisms of interplay between the glymphatic system and neurotoxic proteins, including loss of polarization of aquaporin-4 in astrocytic endfeet, sleep and circadian rhythms, neuroinflammation, astrogliosis, and gliosis. This review further delves into the underlying pathophysiology of the glymphatic system in Parkinson's disease and related disorders, and the potential implications of targeting the glymphatic system as a novel and promising therapeutic strategy.
Collapse
Affiliation(s)
- Yumei Yue
- Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xiaodan Zhang
- Department of Emergency Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wen Lv
- Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hsin-Yi Lai
- Department of Neurology of the Second Affiliated Hospital and School of Brain Science and Brain Medicine, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang Province, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ting Shen
- Department of Neurology of the Second Affiliated Hospital and School of Brain Science and Brain Medicine, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
49
|
Nishida I, Yamada K, Sakamoto A, Wakabayashi T, Iwatsubo T. Chronic Neuronal Hyperexcitation Exacerbates Tau Propagation in a Mouse Model of Tauopathy. Int J Mol Sci 2024; 25:9004. [PMID: 39201689 PMCID: PMC11354494 DOI: 10.3390/ijms25169004] [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: 07/18/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
The intracerebral spread of tau is a critical mechanism associated with functional decline in Alzheimer's disease (AD) and other tauopathies. Recently, a hypothesis has emerged suggesting that tau propagation is linked to functional neuronal connections, specifically driven by neuronal hyperactivity. However, experimental validation of this hypothesis remains limited. In this study, we investigated how tau propagation from the entorhinal cortex to the hippocampus, the neuronal circuit most susceptible to tau pathology in AD, is affected by the selective stimulation of neuronal activity along this circuit. Using a mouse model of seed-induced propagation combined with optogenetics, we found that the chronic stimulation of this neuronal connection over a 4-week period resulted in a significant increase in insoluble tau accumulation in both the entorhinal cortex and hippocampus. Importantly, the ratio of tau accumulation in the hippocampus relative to that in the entorhinal cortex, serving as an indicator of transcellular spreading, was significantly higher in mice subjected to chronic stimulation. These results support the notion that abnormal neuronal activity promotes tau propagation, thereby implicating it in the progression of tauopathy.
Collapse
Affiliation(s)
- Itaru Nishida
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Asami Sakamoto
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Tomoko Wakabayashi
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
- Department of Pathophysiology, Meiji Pharmaceutical University, Tokyo 2040004, Japan
| | - Takeshi Iwatsubo
- National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 1878551, Japan;
| |
Collapse
|
50
|
Liu L, Tu L, Shen Q, Bao Y, Xu F, Zhang D, Xu Y. Meta-analysis of the relationship between the number and location of perivascular spaces in the brain and cognitive function. Neurol Sci 2024; 45:3743-3755. [PMID: 38459400 DOI: 10.1007/s10072-024-07438-3] [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: 10/23/2023] [Accepted: 03/01/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Cerebral perivascular spaces are part of the cerebral microvascular structure and play a role in lymphatic drainage and the removal of waste products from the brain. Relationships of the number and location of such spaces with cognition are unclear. OBJECTIVE To meta-analyze available data on potential associations of severity and location of perivascular spaces with cognitive performance. METHODS We searched PubMed, EMBASE, Web of Science and the Cochrane Central Registry of Controlled Trials for relevant studies published between January 2000 and July 2023. Performance on different cognitive domains was compared to the severity of perivascular spaces in different brain regions using comprehensive meta-analysis. When studies report unadjusted and adjusted means, we use adjusted means for meta-analysis. The study protocol is registered in the PROSPERO database (CRD42023443460). RESULTS We meta-analyzed data from 26 cross-sectional studies and two longitudinal studies involving 7908 participants. In most studies perivascular spaces was using a visual rating scale. A higher number of basal ganglia perivascular spaces was linked to lower general intelligence and attention. Moreover, increased centrum semiovale perivascular spaces were associated with worse general intelligence, executive function, language, and memory. Conversely, higher hippocampus perivascular spaces were associated with enhanced memory and executive function. Subgroup analyses revealed variations in associations among different disease conditions. CONCLUSIONS A higher quantity of perivascular spaces in the brain is correlated with impaired cognitive function. The location of these perivascular spaces and the underlying disease conditions may influence the specific cognitive domains that are affected. SYSTEMATIC REVIEW REGISTRATION The study protocol has been registered in the PROSPERO database (CRD42023443460).
Collapse
Affiliation(s)
- Ling Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liangdan Tu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiuyan Shen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Bao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fang Xu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|