1
|
Ma Y, Han Y. Targeting the brain's glymphatic pathway: A novel therapeutic approach for cerebral small vessel disease. Neural Regen Res 2026; 21:433-442. [PMID: 39688573 DOI: 10.4103/nrr.nrr-d-24-00821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Cerebral small vessel disease encompasses a group of neurological disorders characterized by injury to small blood vessels, often leading to stroke and dementia. Due to its diverse etiologies and complex pathological mechanisms, preventing and treating cerebral small vessel vasculopathy is challenging. Recent studies have shown that the glymphatic system plays a crucial role in interstitial solute clearance and the maintenance of brain homeostasis. Increasing evidence also suggests that dysfunction in glymphatic clearance is a key factor in the progression of cerebral small vessel disease. This review begins with a comprehensive introduction to the structure, function, and driving factors of the glymphatic system, highlighting its essential role in brain waste clearance. Afterwards, cerebral small vessel disease was reviewed from the perspective of the glymphatic system, after which the mechanisms underlying their correlation were summarized. Glymphatic dysfunction may lead to the accumulation of metabolic waste in the brain, thereby exacerbating the pathological processes associated with cerebral small vessel disease. The review also discussed the direct evidence of glymphatic dysfunction in patients and animal models exhibiting two subtypes of cerebral small vessel disease: arteriolosclerosis-related cerebral small vessel disease and amyloid-related cerebral small vessel disease. Diffusion tensor image analysis along the perivascular space is an important non-invasive tool for assessing the clearance function of the glymphatic system. However, the effectiveness of its parameters needs to be enhanced. Among various nervous system diseases, including cerebral small vessel disease, glymphatic failure may be a common final pathway toward dementia. Overall, this review summarizes prevention and treatment strategies that target glymphatic drainage and will offer valuable insight for developing novel treatments for cerebral small vessel disease.
Collapse
Affiliation(s)
- Yuhui Ma
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | |
Collapse
|
2
|
Ayyappan K, Unger L, Kitchen P, Bill RM, Salman MM. Measuring glymphatic function: Assessing the toolkit. Neural Regen Res 2026; 21:534-541. [PMID: 40145955 DOI: 10.4103/nrr.nrr-d-24-01013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/27/2025] [Indexed: 03/28/2025] Open
Abstract
Glymphatic flow has been proposed to clear brain waste while we sleep. Cerebrospinal fluid moves from periarterial to perivenous spaces through the parenchyma, with subsequent cerebrospinal fluid drainage to dural lymphatics. Glymphatic disruption is associated with neurological conditions such as Alzheimer's disease and traumatic brain injury. Therefore, investigating its structure and function may improve understanding of pathophysiology. The recent controversy on whether glymphatic flow increases or decreases during sleep demonstrates that the glymphatic hypothesis remains contentious. However, discrepancies between different studies could be due to limitations of the specific techniques used and confounding factors. Here, we review the methods used to study glymphatic function and provide a toolkit from which researchers can choose. We conclude that tracer analysis has been useful, ex vivo techniques are unreliable, and in vivo imaging is still limited. Finally, we explore the potential for future methods and highlight the need for in vitro models, such as microfluidic devices, which may address technique limitations and enable progression of the field.
Collapse
Affiliation(s)
- Koushikk Ayyappan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Lucas Unger
- College of Health and Life Sciences, Aston University, Birmingham, UK
- Aston Institute for Membrane Excellence and the School of Biosciences, Aston University, Birmingham, UK
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Birmingham, UK
- Aston Institute for Membrane Excellence and the School of Biosciences, Aston University, Birmingham, UK
| | - Roslyn M Bill
- College of Health and Life Sciences, Aston University, Birmingham, UK
- Aston Institute for Membrane Excellence and the School of Biosciences, Aston University, Birmingham, UK
| | - Mootaz M Salman
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- BHF Oxford Centre of Research Excellence, University of Oxford, Oxford, UK
- Kavli Institute for NanoScience Discovery, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Ghaderi S, Mohammadi S, Fatehi F. Glymphatic pathway dysfunction in severe obstructive sleep apnea: A meta-analysis. Sleep Med 2025; 131:106528. [PMID: 40267528 DOI: 10.1016/j.sleep.2025.106528] [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: 01/21/2025] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Obstructive sleep apnea (OSA), a sleep disorder, is associated with cognitive decline and is potentially linked to glymphatic system dysfunction. This meta-analysis investigates glymphatic function in severe OSA (apnea-hypopnea index ≥30) using the Diffusion Tensor Imaging Analysis along the Perivascular Space (DTI-ALPS) index. METHODS This study followed PRISMA guidelines for systematic reviews and meta-analyses. A comprehensive search of PubMed, Web of Science, Scopus, and Embase was conducted from inception to January 20, 2024. Studies investigating the ALPS index in OSA using DTI were included. Analyses included a random-effects meta-analysis, sensitivity analysis, meta-regression, publication bias evaluation (funnel plot, Egger's test, and Begg's test), and risk of bias assessment. RESULTS Systematic review identified four studies (137 patients with severe OSA and 170 healthy controls (HCs)). Pooled analysis revealed a significant reduction in the DTI-ALPS index in severe OSA patients compared to HCs (standardized mean difference: -0.95, 95 % CI: -1.46 to -0.44, p < 0.001), indicating impaired glymphatic function. Heterogeneity was moderate to high (I2 = 76.07 %), but sensitivity analyses confirmed robustness. Meta-regression analyses identified the sources of heterogeneity as the apnea-hypopnea index (β = -0.039, p = 0.009) and the Epworth Sleepiness Scale (β = -0.150, p = 0.032), with no effects observed for age or male ratio. Qualitative (funnel plot) and quantitative publication bias assessments (Egger's and Begg's tests) showed no significant bias, and risk of bias evaluations using the Newcastle-Ottawa Scale indicated high methodological quality across studies. CONCLUSIONS These findings suggest that severe OSA disrupts glymphatic activity. The DTI-ALPS index emerges as a promising tool for assessing glymphatic dysfunction in OSA.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Custer RM, Lynch KM, Barisano G, Herting MM, Åkerstedt T, Nilsonne G, Ahmadi H, Choupan J. Effects of one-night partial sleep deprivation on perivascular space volume fraction: Findings from the Stockholm Sleepy Brain Study. Sleep Med 2025; 131:106537. [PMID: 40300399 DOI: 10.1016/j.sleep.2025.106537] [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: 12/05/2024] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
Increased waste clearance in the brain is thought to occur most readily during deep sleep (stage N3). Sleep deprivation disrupts time spent in deeper sleep stages, fragmenting the clearance process. Here, we have utilized the publicly available Stockholm Sleepy Brain Study to investigate whether various sleep-related measures are associated with changes in perivascular space (PVS) volume fraction following a late-night short-sleep experiment. The study sample consisted of 60 participants divided into old (65-75 years) and young (20-30 years) age groups. We found that partial sleep deprivation was not significantly associated with major PVS changes. In our centrum semiovale models, we observed an interaction between percentage of total sleep time spent in N3 and sleep deprivation status on PVS volume fraction. In our basal ganglia models, we saw an interaction between N2 (both percentage of total sleep time and absolute time in minutes) and sleep deprivation status. However, the significance of these findings did not survive multiple comparisons corrections. This work highlights the need for future longitudinal studies of PVS and sleep, allowing for quantification of within-subject morphological changes occurring in PVS due to patterns of poor sleep. Our findings here provide insight on the impact that a single night of late-night short-sleep has on the perivascular waste clearance system.
Collapse
Affiliation(s)
- Rachel M Custer
- Laboratory of Neuro Imaging (LONI), Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kirsten M Lynch
- Laboratory of Neuro Imaging (LONI), Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Megan M Herting
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Torbjörn Åkerstedt
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Psychology, Stockholm University, Stockholm, Sweden
| | - Gustav Nilsonne
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Psychology, Stockholm University, Stockholm, Sweden
| | - Hedyeh Ahmadi
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Jeiran Choupan
- Laboratory of Neuro Imaging (LONI), Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; NeuroScope Inc., Scarsdale, NY, USA.
| |
Collapse
|
5
|
Copie RG, Blomqvist K, Farzaneh Kari M, Kurkela M, Niemi M, Rauhala PV, Lohela TJ, Rosenholm M, Lilius TO. Modulation of spinal morphine pharmacokinetics and antinociception by α 2-adrenergic agonists in the male rat. Neuropharmacology 2025; 270:110369. [PMID: 39956316 DOI: 10.1016/j.neuropharm.2025.110369] [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: 12/12/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
The synergistic antinociceptive effects of α2-adrenergic agonists and intrathecal (i.t.) opioids were initially linked to pharmacodynamics. However, the α2-agonist dexmedetomidine also enhances brain delivery of CSF-administered drugs by increasing glymphatic influx. Here, fadolmidine, a hydrophilic α2-agonist designed for spinal analgesia, was studied for its sedative, antinociceptive, and pharmacokinetic effects with co-administered lumbar intrathecal morphine. Subcutaneous and i.t. dexmedetomidine served as comparators. Forty-eight male Sprague-Dawley rats received i.t. lumbar catheters. Sedative effects of i.t. fadolmidine (1-10 μg) and i.t. dexmedetomidine (1-10 μg) were assessed by open field and rotarod tests. The antinociceptive effects of morphine alone (1.5 μg i.t.) and co-administered with i.t. fadolmidine (3 and 10 μg) were evaluated using the tail-flick test. Effects of i.t. fadolmidine and subcutaneous dexmedetomidine (0.2 mg/kg) on morphine concentration within CNS were assessed by liquid chromatography-tandem mass spectrometry at 60 min. While i.t. dexmedetomidine was sedating, i.t fadolmidine was not. The antinociceptive effects of other treatment regimens weaned at latest after 90 min, whereas the combination of fadolmidine 10 μg i.t. and morphine 1.5 μg i.t. provided antinociception until the end of the measurement period (%maximum possible effect of 77.5 ± 11.5 vs saline 10.6 ± 11.1, p = 0.0002 at 120 min). Subcutaneous dexmedetomidine effectively targeted lumbar morphine towards the injection site resulting in a 3335-fold (95% CI: 929-11978) lower brain-to-injection site ratio, versus a 355-fold (95% CI: 196-641) difference with saline. By improving spinal opioid targeting, α2-adrenergic agonists dexmedetomidine and fadolmidine may reduce supraspinal side effects, enabling safe and efficacious intrathecal analgesia.
Collapse
Affiliation(s)
- Radu G Copie
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Kim Blomqvist
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Melina Farzaneh Kari
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mika Kurkela
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pekka V Rauhala
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Terhi J Lohela
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Department of Anaesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Marko Rosenholm
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tuomas O Lilius
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Emergency Medicine and Services, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
Pinho-Correia LM, McCullough SJC, Ghanizada H, Nedergaard M, Rustenhoven J, Da Mesquita S. CSF transport at the brain-meningeal border: effects on neurological health and disease. Lancet Neurol 2025; 24:535-547. [PMID: 40409317 DOI: 10.1016/s1474-4422(25)00115-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 05/25/2025]
Abstract
The existence of specialised structures that allow a continuous exchange of CSF between different anatomical compartments at the brain-meningeal border is challenging conventional notions around molecular transport within the brain. Experimental findings highlight the conduits and cellular structures controlling the transport of CSF and immune cells between the brain parenchyma (via the glymphatic system), the subarachnoid space (enclosed by the meningeal pia and arachnoid layers), and the outmost meningeal dural layer and calvaria (via the so-called arachnoid cuff exit points). Studies in both rodent models and humans show new mechanisms of brain glymphatic molecular transport, meningeal lymphatic vascular drainage, and immune surveillance at the brain-draining skull bone marrow and cervical lymph nodes. Pathological alterations at the brain-meningeal border have been implicated in disorders of diverse causes, from traumatic brain injury to Alzheimer's disease.
Collapse
Affiliation(s)
| | | | - Hashmat Ghanizada
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, NY, USA
| | - Justin Rustenhoven
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
7
|
Duyvesteyn E, Vizcarra VS, Waight E, Balbuena E, Hablitz LM. Biological Fluid Flows: Signaling Mediums for Circadian Timing. J Biol Rhythms 2025; 40:234-248. [PMID: 40145493 DOI: 10.1177/07487304251323318] [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] [Indexed: 03/28/2025]
Abstract
While there is extensive literature on both the neuronal circuitry of rhythms and the intracellular molecular clock, there is a large component of signaling that has been understudied: interstitial fluid (ISF)-fluid that surrounds the cells in the extracellular space of tissue. In this review, we highlight evidence in the circadian literature supporting ISF signaling as key to circadian synchronization and entrainment and propose new mechanisms of how fluid movement between the brain and periphery may act as zeitgebers by examining the main ISF pathways of the body, focusing on circadian regulation of the glymphatic and lymphatic systems. We identify key pieces of circadian research that point to ISF as an important timing medium, expand on the basics of cerebrospinal fluid (CSF) and ISF production, and outline the basic structure and function of the glymphatic and lymphatic systems.
Collapse
Affiliation(s)
- Evalien Duyvesteyn
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Velia S Vizcarra
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Emma Waight
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Estephanie Balbuena
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
8
|
Hua L, Zeng X, Zhang K, Zhao Z, Yuan Z. Reduced glymphatic clearance in early psychosis. Mol Psychiatry 2025:10.1038/s41380-025-03058-1. [PMID: 40389626 DOI: 10.1038/s41380-025-03058-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 05/02/2025] [Accepted: 05/13/2025] [Indexed: 05/21/2025]
Abstract
Psychosis involves neuroinflammation and oxidative stress, both affecting the glymphatic system, the lymphatic-like, fluid-transport system in the brain. However, it is unclear whether early psychosis is related to impairments in glymphatic functions. In resting-state fMRI, it has been recently established in a number of neurodegenerative diseases that the coupling relationship between cortical blood-oxygen-level-dependent (BOLD) signal and ventricular cerebrospinal fluid (CSF) flow is associated with brain waste clearance, a key glymphatic function that has not been examined in psychosis or any other psychiatric populations. In a large dataset (total n = 137, age = 23.86 ± 4.16), we demonstrated that glymphatic clearance marked by BOLD-CSF coupling was weaker and more delayed in patients with early psychosis compared to healthy controls. BOLD-CSF coupling also varied between the non-affective and affective psychosis groups with group differences most prominent in high-order but not low-order cortical regions. Finally, reduced global BOLD-CSF coupling was associated with cognitive decline and more severe psychotic symptoms. We provided novel evidence highlighting dysregulated coupling between cortical activity and macroscopic CSF flow as a biomarker for early psychosis. Similar to recent observations in neurodegenerative disorders, the association between reduced BOLD-CSF coupling and psychotic symptoms suggested that waste clearance is disrupted in psychosis which shed light on the pathophysiology of this disease from a glymphatic point of view.
Collapse
Affiliation(s)
- Lin Hua
- Faculty of Health Sciences, University of Macau, Macau SAR, China
- Center for Cognitive and Brain Sciences, University of Macau, Macau SAR, China
- Brainstem Imaging Laboratory, Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Xinglin Zeng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
- Center for Cognitive and Brain Sciences, University of Macau, Macau SAR, China
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kaixi Zhang
- Center for Cognitive and Brain Sciences, University of Macau, Macau SAR, China
- Center for Addiction and Mental Health, Toronto, ON, Canada
| | - Zhiying Zhao
- Center for Cognitive and Brain Sciences, University of Macau, Macau SAR, China.
| | - Zhen Yuan
- Faculty of Health Sciences, University of Macau, Macau SAR, China.
- Center for Cognitive and Brain Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
9
|
Khandayataray P, Murthy MK. Exploring the nexus: Sleep disorders, circadian dysregulation, and Alzheimer's disease. Neuroscience 2025; 574:21-41. [PMID: 40189132 DOI: 10.1016/j.neuroscience.2025.03.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
We reviewed the connections among Alzheimer's disease (AD), sleep deprivation, and circadian rhythm disorders. Evidence is mounting that disrupted sleep and abnormal circadian rhythms are not merely symptoms of AD, but are also involved in accelerating the disease. Amyloid-beta (Aβ) accumulates, a feature of AD, and worsens with sleep deprivation because glymphatic withdrawal is required to clear toxic proteins from the brain. In addition, disturbances in circadian rhythm can contribute to the induction of neuroinflammation and oxidative stress, thereby accelerating neurodegenerative processes. While these interactions are bidirectional, Alzheimer's pathology further disrupts sleep and circadian function in a vicious cycle that worsens cognitive decline, which is emphasized in the review. The evidence that targeting sleep and circadian mechanisms may serve as therapeutic strategies for AD was strengthened by this study through the analysis of the molecular and physiological pathways. Further work on this nexus could help unravel the neurobiological mechanisms common to the onset of Alzheimer's and disrupted sleep and circadian regulation, which could result in earlier intervention to slow or prevent the onset of the disease.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India.
| |
Collapse
|
10
|
van Hattem T, Verkaar L, Krugliakova E, Adelhöfer N, Zeising M, Drinkenburg WHIM, Claassen JAHR, Bódizs R, Dresler M, Rosenblum Y. Targeting Sleep Physiology to Modulate Glymphatic Brain Clearance. Physiology (Bethesda) 2025; 40:0. [PMID: 39601891 DOI: 10.1152/physiol.00019.2024] [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: 04/05/2024] [Revised: 09/12/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
Sleep has been postulated to play an important role in the removal of potentially neurotoxic molecules, such as amyloid-β, from the brain via the glymphatic system. Disturbed sleep, on the other hand, may contribute to the accumulation of neurotoxins in brain tissue, eventually leading to neuronal death. A bidirectional relationship has been proposed between impaired sleep and neurodegenerative processes, which start years before the onset of clinical symptoms associated with conditions like Alzheimer's and Parkinson's diseases. Given the heavy burden these conditions place on society, it is imperative to develop interventions that promote efficient brain clearance, thereby potentially aiding in the prevention or slowing of neurodegeneration. In this review, we explore whether the metabolic clearance function of sleep can be enhanced through sensory (e.g., auditory, vestibular) or transcranial (e.g., magnetic, ultrasound, infrared light) stimulation, as well as pharmacological (e.g., antiepileptics) and behavioral (e.g., sleeping position, physical exercise, cognitive intervention) modulation of sleep physiology. A particular focus is placed on strategies to enhance slow-wave activity during nonrapid eye movement sleep as a driver of glymphatic brain clearance. Overall, this review provides a comprehensive overview on the potential preventative and therapeutic applications of sleep interventions in combating neurodegeneration, cognitive decline, and dementia.
Collapse
Affiliation(s)
- Timo van Hattem
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lieuwe Verkaar
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elena Krugliakova
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nico Adelhöfer
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel Zeising
- Klinikum Ingolstadt, Centre of Mental Health, Ingolstadt, Germany
| | - Wilhelmus H I M Drinkenburg
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen, Groningen, The Netherlands
| | - Jurgen A H R Claassen
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Róbert Bódizs
- Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary
| | - Martin Dresler
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yevgenia Rosenblum
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Toft‐Bertelsen TL, Edelbo BL, Steffensen AB, Lolansen SD, Wardman JH, Jensen DB, MacAulay N. The Cerebrospinal Fluid Secretion Rate Increases in Awake and Freely Moving Rats but Differs With Experimental Methodology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412469. [PMID: 40071749 PMCID: PMC12061303 DOI: 10.1002/advs.202412469] [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] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/12/2025] [Indexed: 05/10/2025]
Abstract
Cerebrospinal fluid (CSF) dynamics hold implications for neurological health. Despite its importance, accurate quantification of the CSF secretion rate remains a challenge due to methodological controversies and the influence of anesthesia. A novel technique is established to determine CSF dynamics in awake and freely moving rats, and the CSF secretion is quantified with three different methodologies. The CSF secretion rate is higher in awake rats than in anesthetized rats, the latter demonstrating no requirement for mechanical ventilation for optimal CSF quantification. The CSF secretion rate is ≈10-fold lower with the "direct method" than with the ventriculo-cisternal perfusion assay, although the relative acetazolamide-mediated reduction in CSF secretion is similar across three tested methods. The findings demonstrate the importance of awake models for optimal quantification of the absolute rate of CSF secretion but highlight the versatility of methodologies for the determination of relative changes in CSF secretion associated with inhibitors, age, sex, and various pathologies.
Collapse
Affiliation(s)
| | - Beatriche L. Edelbo
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen NDK‐2200Denmark
| | - Annette B. Steffensen
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen NDK‐2200Denmark
| | - Sara D. Lolansen
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen NDK‐2200Denmark
| | - Jonathan H. Wardman
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen NDK‐2200Denmark
| | - Dennis B. Jensen
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen NDK‐2200Denmark
| | - Nanna MacAulay
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen NDK‐2200Denmark
| |
Collapse
|
12
|
Gu J, Chen Y, Tang H, Chen X, Xing S. Impaired glymphatic system is associated with secondary neuronal injury in the thalamus following cerebral cortical infarction. Brain Res Bull 2025; 224:111330. [PMID: 40180189 DOI: 10.1016/j.brainresbull.2025.111330] [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: 11/28/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Focal cerebral infarction leads to abnormal amyloid-β (Aβ) deposits, which relates to secondary neuronal injury in the ipsilateral thalamus and hinders post-stroke functional recovery. However, the mechanisms underlying Aβ-pathology in the thalamus remain unclear. This study was designed to investigate the potential associations of glymphatic system with Aβ deposits and delayed neuronal damage of the ipsilateral thalamus secondary to cerebral infarction. Cortical infarction was induced with middle cerebral artery occlusion (MCAO). Secondary neuronal damage, Aβ deposits and aquaporin 4 (AQP4) polarity in the thalamus were examined by Nissl staining, immunochemistry and immunoblotting analyses. Glymphatic function was evaluated using fluorescent tracers. The effects of glymphatic system on Aβ deposits and secondary neuronal damage were determined by shRNA-mediated AQP4 knockdown. The results showed that AQP4 polarization and the clearance of tracers were obviously decreased in the ipsilateral thalamus at seven days after MCAO when compared to the sham-operated group. In parallel, there were increases in Aβ deposits, neuronal loss and astrogliosis in the ipsilateral thalamus at seven days after MCAO. Additionally, AQP4 knockdown further reduced the degree of AQP4 polarity and efflux function of tracers, coinciding with marked increases in Aβ deposits and neuronal loss in the ipsilateral thalamus at seven days after MCAO. This effect was associated with exacerbated somatosensory and cognitive deficits. These findings suggest that impaired glymphatic system was associated with secondary neuronal damage in the thalamus after cerebral infarction by possibly suppressing Aβ clearance.
Collapse
Affiliation(s)
- Jinmin Gu
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Yuqian Chen
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Huijia Tang
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Xinran Chen
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China.
| | - Shihui Xing
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, China; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China.
| |
Collapse
|
13
|
Levendovszky SR, Meyer B. Diffusion Tensor Imaging in Neurofluids. Neuroimaging Clin N Am 2025; 35:211-222. [PMID: 40210378 PMCID: PMC11986261 DOI: 10.1016/j.nic.2024.11.001] [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] [Indexed: 04/12/2025]
Abstract
In this review article, we describe the development and application of diffusion-based MR imaging methods for studying glymphatic physiology. Fluid exchange and solute transport are the 2 key components of the glymphatic system. Here we describe the use of low b-value imaging, free water fraction imaging, and diffusion time sensitization to leverage cerebral spinal fluid, as well as interstitial fluid motion in the parenchyma. We also describe multiple b-value diffusion imaging to better delineate diffusion components within the brain. Finally, we touch upon newer approaches that use advanced models of the diffusion signal, including high b-value imaging.
Collapse
Affiliation(s)
- Swati Rane Levendovszky
- Department of Radiology, University of Washington School of Medicine, 1959 Northeast Pacific Street, Box 357223, Seattle, WA 98195, USA.
| | - Briana Meyer
- Department of Radiology, University of Washington School of Medicine, 1959 Northeast Pacific Street, Box 357223, Seattle, WA 98195, USA
| |
Collapse
|
14
|
Nenert R, Mueller C, Catiul C, Pilkington J, LeVan P, Sharma A, Szaflarski JP, Amara AW. Brain physiological pulsations are linked to sleep architecture and cognitive performance in older adults. Neuroimage 2025; 311:121187. [PMID: 40187437 DOI: 10.1016/j.neuroimage.2025.121187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The glymphatic system facilitates efficient waste clearance in the brain through the movement of cerebrospinal fluid (CSF) along perivascular spaces. Animal studies have demonstrated that glymphatic efficiency declines with age, but evidence for such decline in humans is limited. We hypothesized that reduced glymphatic efficiency in older adults may be related to age-related worsening of sleep quality, potentially contributing to cognitive impairment. METHODS 20 participants aged ≥60 years provided multi-dimensional cognitive measures, overnight polysomnography, and Magnetic Resonance Encephalography (MREG) performed the morning following the PSG. MREG is a single-shot, three-dimensional (3D) sequence employing a spherical stack-of-spirals trajectory that undersamples 3D k-space, enabling whole-brain data acquisition every 100 milliseconds to non-invasively and dynamically assess brain physiological pulsations. Spectral power and optical flow analyses quantified physiological pulsations within cardiovascular (CvB; 0.52-1.6 Hz), respiratory (RFB; 0.11-0.44 Hz), and low-frequency (LFB; 0.008-0.1 Hz) bands. These measures were correlated with cognitive test scores and sleep parameters assessed by overnight polysomnography. RESULTS Significant associations emerged between physiological pulsations, sleep, and cognitive measures. Cardiovascular pulsation strength correlated with non-rapid eye movement (NREM) stage 3 (N3) sleep percentage (peak voxel in right frontal pole; r = 0.72, p < 0.001) and language domain performance (left calcarine gyrus; r = 0.56, p = 0.01). Respiratory pulsations correlated strongly with sleep onset latency (right inferior temporal gyrus; r = 0.75, p < 0.001). Additionally, low-frequency pulsations were associated with sleep onset latency (right precentral gyrus; r = 0.67, p = 0.002). These findings suggest that glymphatic efficiency, as reflected by brain pulsations, is closely linked to sleep quality and cognitive performance in older adults, particularly involving cortical and subcortical structures relevant to cognitive and sleep regulatory functions. CONCLUSION This study uniquely demonstrates that brain physiological pulsations measured non-invasively with MREG are significantly associated with sleep architecture and cognitive performance in older adults. These findings underscore the potential of MREG to assess glymphatic function and provide important insights into the mechanisms linking sleep disturbances, cognitive decline, and aging. The identified correlations between pulsations and specific brain regions highlight potential pathways through which impaired glymphatic function could contribute to cognitive decline in older adults, suggesting promising avenues for future clinical and research applications.
Collapse
Affiliation(s)
- Rodolphe Nenert
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA.
| | - Christina Mueller
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA
| | - Corina Catiul
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA
| | - Jennifer Pilkington
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA
| | - Pierre LeVan
- Dept. of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Ayushe Sharma
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA
| | - Jerzy P Szaflarski
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA; Neurobiology, USA; Neurosurgery, USA; UAB Epilepsy Center, Birmingham, AL, USA
| | - Amy W Amara
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA; University of Colorado Anschutz Medical Campus Department of Neurology, Aurora, CO, USA
| |
Collapse
|
15
|
Sewell KR, Collins AM, Chappel-Farley MG, Jain S, Huang H, Grove G, Kramer AF, McAuley E, Burns J, Hillman C, Vidoni E, Marsland A, Kang C, Wan L, Wilckens KA, Erickson KI. Self-report and actigraphy measures of sleep and domain-specific cognitive performance in older adults. GeroScience 2025:10.1007/s11357-025-01665-y. [PMID: 40274698 DOI: 10.1007/s11357-025-01665-y] [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: 02/05/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Poor sleep is associated with worse cognitive function in older adults. However, nuanced associations between sleep and cognition might be masked by the multidimensional nature of sleep which requires multiple approaches (e.g., self-report and actigraphy) to gain meaningful insight. We investigated associations of sleep with cognition and hypothesized that the most consistent association would be between self-reported sleep duration and actigraphy-measured wake after sleep onset (WASO). We utilized baseline data from the Investigating Gains in Neurocognition in an Intervention Trial of Exercise study. Cognitively unimpaired older adults (n=589, aged 65-80) completed a comprehensive cognitive assessment with generation of five domain-specific cognitive composite scores. Sleep was measured via the Pittsburgh Sleep Quality Index (PSQI) and 24-h actigraphy (GT9X Link). Greater actigraphy WASO and shorter self-reported sleep duration were associated with poorer performance in all five cognitive domains (β[range: WASO] = -0.14 to -0.19, all p<0.05; β[range: duration] = 0.08-0.15, all p<0.05). Shorter actigraphy sleep duration was also associated with poorer EF/attentional control (β=0.09, p=0.020) and processing speed (β=0.10, p=0.013). Actigraphy and self-reported sleep were more strongly associated with episodic memory in older (74 years) and younger (66 years) individuals, respectively. Actigraphy-derived WASO was consistently and robustly associated with cognitive performance. Additionally, our results suggest that self-reported sleep duration provides insight into sleep behaviors related to brain health (e.g., long periods of still wakefulness), beyond actigraphy-measured sleep duration. Thus, both self-report and actigraphy measures of sleep provide critical and unique information for interpreting relationships with cognitive performance.
Collapse
Affiliation(s)
- Kelsey R Sewell
- Innovation Tower, AdventHealth Research Institute, 265 E Rollins St, Suite 2100, NeuroscienceOrlando, FL, 32804, USA.
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia.
| | - Audrey M Collins
- Innovation Tower, AdventHealth Research Institute, 265 E Rollins St, Suite 2100, NeuroscienceOrlando, FL, 32804, USA
| | | | - Shivangi Jain
- Innovation Tower, AdventHealth Research Institute, 265 E Rollins St, Suite 2100, NeuroscienceOrlando, FL, 32804, USA
| | - Haiqing Huang
- Innovation Tower, AdventHealth Research Institute, 265 E Rollins St, Suite 2100, NeuroscienceOrlando, FL, 32804, USA
| | - George Grove
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Arthur F Kramer
- Department of Psychology, Northeastern University, Boston, MA, 02115, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, Champaign, IL, 61801, USA
| | - Edward McAuley
- Department of Psychology, Northeastern University, Boston, MA, 02115, USA
- Department of Health and Kinesiology, University of Illinois at Urbana Champaign, Champaign, IL, 61801, USA
| | - Jeffrey Burns
- Department of Neurology, University of Kansas, Kansas City, KS, 66160, USA
| | - Charles Hillman
- Department of Psychology, Northeastern University, Boston, MA, 02115, USA
- Department of Physical Therapy, Movement, & Rehabilitation Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Eric Vidoni
- Department of Neurology, University of Kansas, Kansas City, KS, 66160, USA
| | - Anna Marsland
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Chaeryon Kang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lu Wan
- Innovation Tower, AdventHealth Research Institute, 265 E Rollins St, Suite 2100, NeuroscienceOrlando, FL, 32804, USA
| | - Kristine A Wilckens
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kirk I Erickson
- Innovation Tower, AdventHealth Research Institute, 265 E Rollins St, Suite 2100, NeuroscienceOrlando, FL, 32804, USA
| |
Collapse
|
16
|
Ibrahim A, Högl B, Stefani A. Sleep as the Foundation of Brain Health. Semin Neurol 2025. [PMID: 40139214 DOI: 10.1055/a-2566-4073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Sleep is a vital function, taking about one-third of a human lifetime, and is essential for achieving and maintaining brain health. From homeostatic neurophysiology to emotional and procedural memory processing to clearance of brain waste, sleep and circadian alignment remain paramount. Yet modern lifestyles and clinical practice often dismiss sleep, resulting in profound long-term repercussions. This chapter examines the roles of sleep and circadian rhythms in memory consolidation, synaptic plasticity, and clearance of metabolic waste, highlighting recent advances in neuroscience research. We explore how insufficient and disordered sleep-a public health concern-can impair cognition, escalate neurodegenerative risks, and compromise neurovascular integrity, thereby impacting brain health. These findings underscore the need for comprehensive screening for disturbed sleep and targeted interventions in clinical practice. Emerging interventions and AI-driven technologies may allow early detection and personalized and individualized treatments and improve outcomes. Overall, this chapter reaffirms that healthy sleep is indispensable at any level of neurological disease prevention-on par with the role of diet and exercise in cardiovascular health-and represents the foundation of brain health.
Collapse
Affiliation(s)
- Abubaker Ibrahim
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Birgit Högl
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Ambra Stefani
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
17
|
Hu M, Wu L, Wei E, Pan X, Zhu Q, Xiuyun X, Lv L, Dong X, Liu H, Liu Y. Sildenafil promotes osteogenic differentiation of human mesenchymal stem cells and inhibits bone loss by affecting the TGF-β signaling pathway. Stem Cell Res Ther 2025; 16:201. [PMID: 40264229 PMCID: PMC12016470 DOI: 10.1186/s13287-025-04320-7] [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: 12/21/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Osteoporosis, a common bone disorder, is primarily managed pharmacologically. However, existing medications are associated with non-trivial side-effects. Sildenafil, which already finds many clinical applications, promotes angiogenesis and cellular differentiation. Osteoporotic patients often exhibit a reduced intraosseous vasculature and impaired cellular differentiation; sildenafil may thus usefully treat osteoporosis. METHODS Here, the effects of sildenafil on the osteogenic differentiation of human mesenchymal stem cells (hMSCs) were explored, as were the molecular mechanisms in play. We treated hMSCs with varying concentrations of sildenafil and measured cell proliferation and osteogenic differentiation in vitro. We used a mouse model of subcutaneous ectopic osteogenesis to assess sildenafil's effect on hMSC osteogenic differentiation in vivo. We also explored the effects of sildenafil on bone loss in tail-suspended (TS) and ovariectomized (OVX) mice. Mechanistically, we employed RNA-sequencing to define potentially relevant molecular pathways. RESULTS The appropriate concentrations of sildenafil significantly enhanced osteogenic hMSC differentiation; the optimal sildenafil concentration may be 10 mg/L. Sildenafil mitigated osteoporosis in OVX and TS mice. The appropriate concentrations of sildenafil probably promoted hMSC osteogenic differentiation by acting on the transforming growth factor-β (TGF-β) signaling pathway. CONCLUSIONS In conclusion, sildenafil enhanced hMSC osteogenic differentiation and inhibited bone loss. Sildenafil may usefully treat osteoporosis. Our findings offer new insights into the physiological effects of the medicine.
Collapse
Affiliation(s)
- Menglong Hu
- Department of Prosthodontics, Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Likun Wu
- Department of Prosthodontics, Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Erfan Wei
- Department of Prosthodontics, Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Xingtong Pan
- Department of Prosthodontics, Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Qiyue Zhu
- Department of Prosthodontics, Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Xv Xiuyun
- Department of Prosthodontics, Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Letian Lv
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Xinyi Dong
- Department of Prosthodontics, Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China.
- Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, Beijing, 100081, China.
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, China.
- Peking University School and Hospital of StomatologyPeking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, China.
| |
Collapse
|
18
|
Aquino G, Palagini L, Alfì G, Feige B, Spiegelhalder K, Piarulli A, Gemignani A. The Interplay Between the Sleep Slow Oscillation and Cerebrospinal Fluid: New Vistas for Insomnia Research. J Sleep Res 2025:e70069. [PMID: 40243037 DOI: 10.1111/jsr.70069] [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: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Insomnia disorder affects about 10% of the global population, representing a major health concern. Despite the availability of evidence-based treatments, the neurobiological mechanisms underpinning this disorder remain poorly understood. Recently, the investigation of the less than 1 Hz oscillations (commonly termed slow oscillations), a hallmark of slow wave sleep, has gained increased interest in research on insomnia. In this context, an intriguing perspective arises from the association between slow oscillations and metabolic waste clearance, an impaired process in individuals suffering from insomnia disorder. Indeed, the exploration of the relationships between cerebrospinal fluid dynamics and glymphatic system functions, which relate to brain metabolic clearance, and sleep slow oscillations may represent a promising avenue for future research in this field. This narrative review examines current knowledge about the intricate interplay among these mechanisms and their implications for insomnia disorder. Particular attention is given to the role of sleep slow oscillations in the clearance of metabolic waste during sleep, their coupling with cerebrospinal fluid oscillations, and the regulatory mechanisms underlying glymphatic function. The review emphasises the relevance of investigating sleep slow oscillations-related mechanisms in insomnia, intending to provide novel insights into the neurophysiological underpinnings of the disorder and contribute to more accurate diagnostic approaches. Furthermore, a deeper understanding of these mechanisms could pave the way for the development of innovative or adjunctive therapeutic strategies targeting sleep slow oscillations-related alterations in insomnia disorder.
Collapse
Affiliation(s)
- Giulia Aquino
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Laura Palagini
- Department of Neuroscience, University of Pisa Hospital, Pisa, Italy
| | - Gaspare Alfì
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Bernd Feige
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Kai Spiegelhalder
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Andrea Piarulli
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
- Coma Science Group, GIGA Consciousness, University of Liège, Liège, Belgium
| | - Angelo Gemignani
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
- Department of Neuroscience, University of Pisa Hospital, Pisa, Italy
| |
Collapse
|
19
|
Huang NX, Zeng JY, Huang HW, Fang SY, Chen S, Li JQ, Chen HJ, Zou ZY. Association of glymphatic system disturbance with neural dysfunction in amyotrophic lateral sclerosis. Quant Imaging Med Surg 2025; 15:3445-3457. [PMID: 40235752 PMCID: PMC11994501 DOI: 10.21037/qims-24-1297] [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: 06/26/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025]
Abstract
Background Formation and aggregation of pathological proteins in the brain constitutes a critical hallmark of amyotrophic lateral sclerosis (ALS). However, the role of the glymphatic system in the clearance of pathological proteins in ALS remains unclear. The purpose of this cross-sectional study was to evaluate glymphatic system disturbance in ALS and its relation to neural function. Methods This study included 38 healthy controls (HCs) and 30 patients with ALS who underwent diffusion tensor imaging (DTI) and resting-state functional magnetic resonance imaging (rs-fMRI). The disease severity, duration, and progression rate of ALS were recorded. Glymphatic system function was indirectly evaluated by DTI analysis along the perivascular space (ALPS) surrounding the deep medullary vein. Neural activity was examined in sensorimotor-related brain areas by measuring amplitude of low-frequency fluctuation (ALFF) based on rs-fMRI. A two-sample t-test or Mann-Whitney test was used to examine between-group differences in ALPS, diffusivities measured along the x-, y-, and z-axis in the association (Dxx_association, Dyy_association, Dzz_association) and projection (Dxx_projection, Dyy_projection, Dzz_projection) fiber areas, and ALFF indices. The associations between ALPS, diffusivities, ALFF, and clinical assessments were determined via Spearman correlation analysis, and diagnostic performance was evaluated with receiver operating characteristic curve analysis. Results Patients with ALS exhibited significantly decreased ALPS and increased diffusivities (Dyy_association and Dyy_projection) as compared to HCs (all P values <0.05). Patients with ALS showed decreased ALFF in sensorimotor-related regions, including the bilateral primary motor and somatosensory areas (all P values <0.001) and left supplementary motor area (P=0.031). ALPS and diffusivities were correlated with ALFF in the sensorimotor-motor regions (all P values <0.05), and ALPS and ALFF correlated with disease severity and duration (all P values <0.05). ALPS, diffusivities, and ALFF showed moderate ability to diagnose ALS. Conclusions The glymphatic system function was impaired in ALS. This may contribute to spontaneous neural activity disturbance and could represent a mechanism for the development of sensorimotor deficits frequently observed in patients with ALS.
Collapse
Affiliation(s)
- Nao-Xin Huang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jing-Yi Zeng
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hui-Wei Huang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Si-Yuan Fang
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Sheng Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Neurology, Fujian Medical University, Fuzhou, China
| | - Jian-Qi Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Hua-Jun Chen
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhang-Yu Zou
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Neurology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
20
|
Alipour M, Rausch J, Mednick SC, Cook JD, Plante DT, Malerba P. The Space-Time Organisation of Sleep Slow Oscillations as Potential Biomarker for Hypersomnolence. J Sleep Res 2025:e70059. [PMID: 40170232 DOI: 10.1111/jsr.70059] [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: 09/16/2024] [Revised: 02/21/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
Research suggests that the spatial profile of slow wave activity (SWA) could be altered in hypersomnolence. Slow oscillations (SOs; 0.5-1.5 Hz), single waveform events contributing to SWA, can be labelled as Global, Frontal, or Local depending on their presentation on the scalp. We showed that SO space-time types differentiate in their amplitudes, coordination with sleep spindles, and propagation patterns. This study applies our data-driven analysis to the nocturnal sleep of adults with and without hypersomnolence and major depressive disorder (MDD) to explore the potential relevance of SO space-time patterns as hypersomnolence signatures in the sleep EEG. We leverage an existing dataset of nocturnal polysomnography with high-density EEG in 83 adults, organised in four groups depending on the presence/absence of hypersomnolence and on the presence/absence of MDD. Group comparisons were conducted considering either two groups (hypersomnolence status) or the four groups separately. Data shows enhanced Frontal SO activity compared with Global activity in hypersomnolence, with or without MDD, and a loss of Global SO amplitude at central regions in hypersomnolence without MDD compared to controls. As Global SOs travel fronto-parietally, we interpret these results as likely driven by a loss of coordination of Global SO activity in hypersomnolence without MDD, resulting in an overabundance of Frontal SOs. This study suggests that characteristics of Frontal SO and Global SOs may have the potential to differentiate individuals with hypersomnolence without MDD, and that the space-time organisation of SOs could be a mechanistically relevant indicator of changes in sleep brain dynamics related to hypersomnolence.
Collapse
Affiliation(s)
- Mahmoud Alipour
- Center for Biobehavioral Health, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
- The Ohio State University, College of Medicine, Columbus, Ohio, USA
| | - Joseph Rausch
- Center for Biobehavioral Health, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
- The Ohio State University, College of Medicine, Columbus, Ohio, USA
| | - Sara C Mednick
- Department of Cognitive Sciences, University of California, Irvine, California, USA
| | - Jesse D Cook
- Department of Psychiatry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Psychology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David T Plante
- Department of Psychiatry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Paola Malerba
- Center for Biobehavioral Health, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
- The Ohio State University, College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
21
|
Bonnar O, Eyre B, van Veluw SJ. Perivascular brain clearance as a therapeutic target in cerebral amyloid angiopathy and Alzheimer's disease. Neurotherapeutics 2025; 22:e00535. [PMID: 39890534 PMCID: PMC12047398 DOI: 10.1016/j.neurot.2025.e00535] [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: 10/10/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
Although distinct diseases, both cerebral amyloid angiopathy (CAA) and Alzheimer's disease (AD) are characterized by the aggregation and accumulation of amyloid-β (Aβ). This is thought to be due, in part, to impaired perivascular Aβ clearance from the brain. This shared failure in both diseases presents a common opportunity for therapeutic intervention. In this review we discuss the idea that promoting perivascular brain clearance could be an effective strategy for safely reducing Aβ levels in CAA and AD thereby improving clinical outcomes, most notably hemorrhagic stroke and cognitive decline. We will explore the evidence for the different forces that are thought to drive perivascular brain clearance, review the literature on potential strategies for potentiating these driving forces, and finally we will discuss the substantial translational challenges and considerations that would accompany such an intervention.
Collapse
Affiliation(s)
- Orla Bonnar
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Beth Eyre
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Susanne J van Veluw
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Franks NP, Wisden W. Reply to: A curious concept of CNS clearance. Nat Neurosci 2025; 28:734-736. [PMID: 40069362 DOI: 10.1038/s41593-025-01898-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/28/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Nicholas P Franks
- Department of Life Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute, Imperial College London, London, UK.
| | - William Wisden
- Department of Life Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute, Imperial College London, London, UK.
| |
Collapse
|
23
|
Chung SJ, Kang SH, Kang M, Choi Y, Park YJ, Kim H, Oh K, Koh SB, Kim JB. Impact of sleep apnea on alzheimer's disease in relation to sex: an 8-year longitudinal follow-up study of a nationwide cohort. Alzheimers Res Ther 2025; 17:65. [PMID: 40114234 PMCID: PMC11924715 DOI: 10.1186/s13195-024-01667-6] [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: 09/05/2024] [Accepted: 12/26/2024] [Indexed: 03/22/2025]
Abstract
BACKGROUND We aimed to investigate the association between sleep apnea and incident dementia (dementia of the Alzheimer type [DAT] and vascular dementia) and whether differences in the effects of sleep apnea on dementia depend on sex. Furthermore, we sought to determine whether obesity affects the sex-specific relationship between sleep apnea and dementia. METHODS We used de-identified data on patients with sleep apnea and a control group aged ≥ 50 years from the Korean National Health Insurance Service. After propensity score matching to balance age and sex between the patient and control groups, 30,111 individuals with sleep apnea (patient group) and 121,528 individuals without sleep apnea (control group) were included. To investigate the impact of sleep apnea on the development of dementia, we used Cox proportional hazards regression after controlling for potential confounders. RESULTS Sleep apnea was predictive of developing DAT in both women (hazard ratio [HR] = 1.30, 95% confidence interval [CI] 1.16-1.44, p < 0.001) and men (HR = 1.13, 95% CI 1.03-1.24, p = 0.012). The adverse effects of sleep apnea on DAT were more prominent in women than in men (p = 0.015 for sleep apnea×sex). Furthermore, obesity affected the sex-specific relationship between sleep apnea and DAT. Specifically, the adverse effects of obese sleep apnea on the DAT were more pronounced in women than in men (p = 0.002 for obese sleep apnea×sex). In contrast, there were no differences in the effects of non-obese sleep apnea on DAT between women and men (p = 0.667 for non-obese sleep apnea×sex). CONCLUSIONS Our results highlight sex differences in the adverse effects of sleep apnea on DAT. Furthermore, these results suggest that sex-specific strategies for controlling sleep apnea are necessary to prevent DAT.
Collapse
Affiliation(s)
- Su Jin Chung
- Department of Neurology, Inje University Ilsan Paik Hospital, Inje University College of Medicine, 170 Juhwa-ro, Ilsanseo-gu, Goyang-si, Gyeonggi-do, 10380, South Korea
| | - Sung Hoon Kang
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, South Korea.
| | - Minwoong Kang
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, South Korea
| | - Yunjin Choi
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, South Korea
| | - Yu Jeong Park
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, South Korea
| | - Hayom Kim
- Department of Neurology, Korea University Anam Hospital, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Kyungmi Oh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, South Korea
| | - Seong-Beom Koh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, South Korea
| | - Jung Bin Kim
- Department of Neurology, Korea University Anam Hospital, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea.
| |
Collapse
|
24
|
Sánchez-Benavides G, Iranzo A, Grau-Rivera O, Giraldo DM, Buongiorno M. Olfactory Dysfunction as a Clinical Marker of Early Glymphatic Failure in Neurodegenerative Diseases. Diagnostics (Basel) 2025; 15:719. [PMID: 40150062 PMCID: PMC11941644 DOI: 10.3390/diagnostics15060719] [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: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
An abnormal accumulation of misfolded proteins is a common feature shared by most neurodegenerative disorders. Olfactory dysfunction (OD) is common in the elderly population and is present in 90% of patients with Alzheimer's or Parkinson's disease, usually preceding the cognitive and motor symptoms onset by several years. Early Aβ, tau, and α-synuclein protein aggregates deposit in brain structures involved in odor processing (olfactory bulb and tract, piriform cortex, amygdala, entorhinal cortex, and hippocampus) and seem to underly OD. The glymphatic system is a glial-associated fluid transport system that facilitates the movement of brain fluids and removes brain waste during specific sleep stages. Notably, the glymphatic system became less functional in aging and it is impaired in several conditions, including neurodegenerative diseases. As the nasal pathway has been recently described as the main outflow exit of cerebrospinal fluid and solutes, we hypothesized that OD may indeed be a clinical marker of early glymphatic dysfunction through abnormal accumulation of pathological proteins in olfactory structures. This effect may be more pronounced in peri- and postmenopausal women due to the well-documented impact of estrogen loss on the locus coeruleus, which may disrupt multiple mechanisms involved in glymphatic clearance. If this hypothesis is confirmed, olfactory dysfunction might be considered as a clinical proxy of glymphatic failure in neurodegenerative diseases.
Collapse
Affiliation(s)
- Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (G.S.-B.); (O.G.-R.)
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28029 Madrid, Spain
| | - Alex Iranzo
- Sleep Disorders Center, Neurology Service, Hospital Clínic Universitari de Barcelona, University of Barcelona, 08036 Barcelona, Spain;
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 28031 Madrid, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (G.S.-B.); (O.G.-R.)
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28029 Madrid, Spain
| | - Darly Milena Giraldo
- Neurology Department, Vall d’Hebron University Hospital, 08035 Barcelona, Spain;
- Neurovascular Diseases Research Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| | - Mariateresa Buongiorno
- Neurology Department, Vall d’Hebron University Hospital, 08035 Barcelona, Spain;
- Neurovascular Diseases Research Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| |
Collapse
|
25
|
MacKinnon MJ, Song S, Chao THH, Hsu LM, Albert ST, Ma Y, Shnitko TA, Wang TWW, Nonneman RJ, Freeman CD, Ozarkar SS, Emir UE, Shen MD, Philpot BD, Hantman AW, Lee SH, Chang WT, Shih YYI. SORDINO for Silent, Sensitive, Specific, and Artifact-Resisting fMRI in awake behaving mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642406. [PMID: 40161795 PMCID: PMC11952411 DOI: 10.1101/2025.03.10.642406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Blood-oxygenation-level-dependent (BOLD) functional magnetic resonance imaging (fMRI) has revolutionized our understanding of the brain activity landscape, bridging circuit neuroscience in animal models with noninvasive brain mapping in humans. This immensely utilized technique, however, faces challenges such as acoustic noise, electromagnetic interference, motion artifacts, magnetic-field inhomogeneity, and limitations in sensitivity and specificity. Here, we introduce Steady-state On-the-Ramp Detection of INduction-decay with Oversampling (SORDINO), a transformative fMRI technique that addresses these challenges by maintaining a constant total gradient amplitude while acquiring data during continuously changing gradient direction. When benchmarked against conventional fMRI on a 9.4T system, SORDINO is silent, sensitive, specific, and resistant to motion and susceptibility artifacts. SORDINO offers superior compatibility with multimodal experiments and carries novel contrast mechanisms distinct from BOLD. It also enables brain-wide activity and connectivity mapping in awake, behaving mice, overcoming stress- and motion-related confounds that are among the most challenging barriers in current animal fMRI studies.
Collapse
Affiliation(s)
- Martin J. MacKinnon
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sheng Song
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tzu-Hao Harry Chao
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Li-Ming Hsu
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott T. Albert
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yuncong Ma
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tatiana A. Shnitko
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tzu-Wen Winnie Wang
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Randy J. Nonneman
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corey D. Freeman
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Siddhi S. Ozarkar
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Uzay E. Emir
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark D. Shen
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Benjamin D. Philpot
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam W. Hantman
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sung-Ho Lee
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wei-Tang Chang
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
26
|
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
|
27
|
Korann V, Panganiban KJ, Stogios N, Remington G, Graff-Guerrero A, Chintoh A, Hahn MK, Agarwal SM. The Dysregulation of the Glymphatic System in Patients with Psychosis Spectrum Disorders Minimally Exposed to Antipsychotics: La dérégulation du système glymphatique en présence de troubles psychotiques chez des patients peu exposés à des antipsychotiques. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2025; 70:260-270. [PMID: 39428987 PMCID: PMC11562879 DOI: 10.1177/07067437241290193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
OBJECTIVE The pathophysiological mechanisms influencing psychosis spectrum disorders are largely unknown. The glymphatic system, which is a brain waste clearance pathway, has recently been implicated in its pathophysiology and has also been shown to be disrupted in various neurodegenerative and vascular diseases. Initial studies examining the glymphatic system in psychosis spectrum disorders have reported disruptions, but the findings have been confounded by medication effects as they included antipsychotic-treated patients. In this study, we used diffusion tensor imaging analysis along the perivascular space (DTI-ALPS) as a technique to measure the functionality of the glymphatic system in a sample of antipsychotic-minimally exposed patients with psychosis spectrum disorders and healthy controls. METHODS The study included 13 antipsychotic-minimally exposed (2 weeks antipsychotic exposure in the past 3 months/lifetime) patients with psychosis spectrum disorders and 114 healthy controls. We quantified water diffusion metrics along the x-, y-, and z-axes in both projection and association fibres to derive the DTI-ALPS index, a proxy for glymphatic activity. Between-group differences were analyzed using two-way ANCOVA controlling for age and sex. Partial correlations were used to assess the association between the ALPS index and clinical variables. RESULTS Analyses revealed that antipsychotic-minimally exposed psychosis spectrum disorder patients had a lower DTI-ALPS index value than healthy controls in both hemispheres and the whole brain (all P < 0.005). Significant differences were also observed between the x and y projections/associations between patients and healthy controls (P < 0.001). Furthermore, we did not find any significant correlations (all P > 0.05) between the DTI-ALPS index with age, body mass index, symptomatology, and metabolic parameters. CONCLUSION This study shows that the glymphatic system is dysregulated in antipsychotic-minimally exposed patients with psychosis spectrum disorders. Understanding the mechanisms that influence the glymphatic system may help to understand the pathophysiology of psychosis spectrum disorders as proper waste clearance is needed for normal brain functioning.
Collapse
Affiliation(s)
- Vittal Korann
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Kristoffer J. Panganiban
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Nicolette Stogios
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Gary Remington
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Psychological Clinical Science, University of Toronto Scarborough, Toronto, ON, Canada
| | - Ariel Graff-Guerrero
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Psychological Clinical Science, University of Toronto Scarborough, Toronto, ON, Canada
- Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Araba Chintoh
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Margaret K. Hahn
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Sri Mahavir Agarwal
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Younes M. Evaluation of Sleep Quality in Clinical Practice. Sleep Med Clin 2025; 20:25-45. [PMID: 39894597 DOI: 10.1016/j.jsmc.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] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
A major problem in clinical sleep medicine is that the most comprehensive and sophisticated investigative tool, the polysomnography, fails to provide a reason for the patient's complaints in all but those with sleep apnea and movement disorders. The reasons why conventional metrics of sleep quality are of limited value are discussed in detail. This is followed by description of several well-established features that have not been implemented in clinical practice because of the impracticality of doing the measurements visually. Automation is pending. Finally, several automated features recently derived from spectral analysis of the electroencephalogram were described.
Collapse
Affiliation(s)
- Magdy Younes
- Department of Medicine, University of Manitoba, 1105-255 Wellington Crescent, Winnipeg, Manitoba, R3M 3V4 Canada.
| |
Collapse
|
29
|
Ji KH, Yun CH. Brain Health in Sleep Disorders. Sleep Med Clin 2025; 20:57-72. [PMID: 39894599 DOI: 10.1016/j.jsmc.2024.10.004] [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] [Indexed: 02/04/2025]
Abstract
Sleep is a critical determinant of brain health, influencing cognitive, emotional, and physiologic functions. The complex bidirectional relationship between sleep and brain health underscores the importance of sleep in maintaining cognitive function, regulating brain homeostasis, and facilitating the clearance of metabolic waste through the glymphatic system. Chronic sleep deprivation and sleep disorders such as insomnia and obstructive sleep apnea have been shown to negatively impact brain structures and functions. This review discusses the impact of sleep disorders on brain health. It also explores the implications of impaired sleep on cardiovascular health, immune function, and neuroplasticity.
Collapse
Affiliation(s)
- Ki-Hwan Ji
- Department of Neurology, Inje University Busan Paik Hospital, College of Medicine, Inje University, 75 Bokji-ro, Busanjin-gu, Busan 47392, Republic of Korea
| | - Chang-Ho Yun
- Deparment of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, Gyeonggi 13620, Republic of Korea.
| |
Collapse
|
30
|
Zamani A, Walker AK, Wright DK. Glymphatic dysfunction and neurodegeneration in ALS: Longitudinal insights from rNLS8 TDP-43 mice. Neurobiol Dis 2025; 206:106832. [PMID: 39914774 DOI: 10.1016/j.nbd.2025.106832] [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: 11/13/2024] [Revised: 12/28/2024] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Dysfunctional Tar DNA binding protein-43 (TDP-43) is found in approximately 95 % of all people with amyotrophic lateral sclerosis (ALS). Recent evidence suggests that the glymphatic system, which clears the brain of waste proteins, is impaired in ALS and may contribute to the accumulation of TDP-43. This study extends this work to investigate how glymphatic function changes over time in the rNLS8 doxycycline (Dox)-dependent TDP-43 mouse model of ALS. Motor function, advanced MRI biomarkers of neurodegeneration, and cortical glymphatic pathway gene expression were assessed together with dynamic contrast-enhanced MRI (DCE-MRI) assessment of glymphatic function at 0-, 3-, 7-, and 21-days after removing mice from Dox feed to initiate cytoplasmic human TDP-43 expression. A trend toward increased glymphatic influx was observed at 3-days post-Dox, together with MRI evidence of brain changes that occurred in the absence of hind-limb clasping and motor impairment. Glymphatic flow is facilitated by aquaporin-4 (AQP4) water channels polarized to astrocytic end feet. We found that while glymphatic function normalized to control levels at 7-days post-Dox, AQP4 expression in the cortex was significantly decreased. After 3-weeks of human TDP-43 expression, glymphatic dysfunction, weight loss, neurodegeneration, motor impairments and astrogliosis were observed. Our findings highlight early glymphatic dysfunction in ALS, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Adam K Walker
- Queensland Brain Institute, The University of Queensland, QLD 4072, Australia; Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW 2006, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
31
|
Turner KL, Brockway DF, Hossain MS, Griffith KR, Greenawalt DI, Zhang Q, Gheres KW, Crowley NA, Drew PJ. Type-I nNOS neurons orchestrate cortical neural activity and vasomotion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634042. [PMID: 39896560 PMCID: PMC11785022 DOI: 10.1101/2025.01.21.634042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
It is unknown how the brain orchestrates coordination of global neural and vascular dynamics. We sought to uncover the role of a sparse but unusual population of genetically-distinct interneurons known as type-I nNOS neurons, using a novel pharmacological strategic to unilaterally ablate these neurons from the somatosensory cortex of mice. Region-specific ablation produced changes in both neural activity and vascular dynamics, decreased power in the delta-band of the local field potential, reduced sustained vascular responses to prolonged sensory stimulation, and abolished the post-stimulus undershoot in cerebral blood volume. Coherence between the left and right somatosensory cortex gamma-band power envelope and blood volume at ultra-low frequencies was decreased, suggesting type-1 nNOS neurons integrate long-range coordination of brain signals. Lastly, we observed decreases in the amplitude of resting-state blood volume oscillations and decreased vasomotion following the ablation of type-I nNOS neurons. This demonstrates that a small population of nNOS-positive neurons are indispensable for regulating both neural and vascular dynamics in the whole brain and implicates disruption of these neurons in diseases ranging from neurodegeneration to sleep disturbances.
Collapse
Affiliation(s)
- Kevin L. Turner
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Dakota F. Brockway
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Md Shakhawat Hossain
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Keith R. Griffith
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Denver I. Greenawalt
- Graduate Program in Molecular Cellular and Integrative Biosciences, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Qingguang Zhang
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
- Department of Physiology, Michigan State University, East Lansing, MI 48824
| | - Kyle W. Gheres
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Nicole A. Crowley
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Patrick J. Drew
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Neurosurgery, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
32
|
Ko JS, Choi Y, Jeong E, Park JE, Kim HS. Hourly Variations in Glymphatic Function Based on MRI Scan Times in Cognitively Normal Individuals. Acad Radiol 2025:S1076-6332(25)00088-1. [PMID: 39934074 DOI: 10.1016/j.acra.2025.01.034] [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: 12/13/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/13/2025]
Abstract
RATIONALE AND OBJECTIVES This study evaluated glymphatic function changes according to MRI scan time over a 24-hour cycle, using diffusion tensor image analysis along the perivascular space (DTI-ALPS) to assess interstitial fluid dynamics. MATERIAL AND METHODS This single-center retrospective cohort study included cognitively normal participants between January and August 2023. Participants were grouped by MRI scan time: dawn, early morning, daytime, and evening/night. Glymphatic function was assessed via the ALPS index calculated from DTI. ALPS index values were compared among groups using one-way ANOVA with post hoc pairwise independent t-tests. Multiple linear regression analysis adjusted for age, sex, and mini-mental state examination scores was used to compare daytime ALPS index with other groups. Paired t-tests assessed ALPS index changes in participants with follow-up MRIs. RESULTS Among 539 participants (age: 70 ± 10 years, 41.4% male), the dawn group had the highest mean ALPS index, followed by early morning, evening/night, and daytime groups (P =0.024). Daytime ALPS index was significantly lower than dawn (P =0.0036) and early morning (P =0.018). Multiple linear regression confirmed lower daytime ALPS index, with the dawn group showing the largest difference (0.067, 95% CI: 0.025-0.108; P =0.002). No significant difference was observed in ALPS index for follow-up scans from dawn or evening/night to daytime (P =0.353). CONCLUSION Glymphatic function varies diurnally, with lower values during daytime. These findings emphasize the importance of circadian timing in evaluating glymphatic function using DTI-ALPS. Further studies are needed to explore intra-individual glymphatic variations.
Collapse
Affiliation(s)
- Ji Su Ko
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, Republic of Korea (J.S.K., Y.C., E.J., J.E.P., H.S.K.); Department of Radiology, Kangbuk Samsung Hospital, Seoul, Republic of Korea (J.S.K.)
| | - Yangsean Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, Republic of Korea (J.S.K., Y.C., E.J., J.E.P., H.S.K.).
| | - Eunseon Jeong
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, Republic of Korea (J.S.K., Y.C., E.J., J.E.P., H.S.K.)
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, Republic of Korea (J.S.K., Y.C., E.J., J.E.P., H.S.K.)
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, Republic of Korea (J.S.K., Y.C., E.J., J.E.P., H.S.K.)
| |
Collapse
|
33
|
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
|
34
|
Corbali O, Levey AI. Glymphatic system in neurological disorders and implications for brain health. Front Neurol 2025; 16:1543725. [PMID: 39974364 PMCID: PMC11835678 DOI: 10.3389/fneur.2025.1543725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025] Open
Affiliation(s)
- Osman Corbali
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | | |
Collapse
|
35
|
Zhang Y, Wu D, Fan Z, Leng X, Wan T, Shi X, Wang X, Wang X, Song C, Du F, Jiang W. Abnormal glymphatic system in patients with autoimmune encephalitis: Relationship with cognitive performance. Brain Res Bull 2025; 221:111232. [PMID: 39880290 DOI: 10.1016/j.brainresbull.2025.111232] [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: 11/17/2024] [Revised: 01/13/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
OBJECTIVES We aimed to explore the impact of glymphatic function in patients diagnosed with autoimmune encephalitis (AE). METHODS In this prospective longitudinal study, patients were recruited from Xijing Hospital between June 2020 and January 2024. Glymphatic function was evaluated using diffusion tensor imaging analysis along the perivascular space (DTI-ALPS). Cognitive impairment was defined as a Montreal Cognitive Assessment (MoCA) score below 26 at the 12-month follow-up. RESULTS A total of 115 individuals were enrolled, including 85 patients with AE and 30 age- and sex-matched healthy controls (HCs). After correcting for age and sex, patients with AE had a significantly lower baseline ALPS index compared to HCs (1.173, 95 % CI [1.135, 1.210] vs. 1.456, 95 % CI [1.371, 1.541]; P < 0.001). The baseline ALPS index was correlated with cognitive performance, including a positive correlation with the Mini-Mental State Examination (MMSE) score (r = 0.568, P < 0.001) and a positive correlation with the MoCA score (r = 0.645, P < 0.001). In the longitudinal study, the ALPS index gradually increased over the follow-up period (P < 0.001), and a low level of the baseline ALPS index was associated with a higher risk of long-term cognitive impairment (HR [95 % CI] = 1.70 [1.12-2.58], P = 0.013). CONCLUSION The glymphatic system is impaired in AE patients. A decreased DTI-ALPS index is associated with a decline in cognitive performance. Additionally, a low baseline ALPS index may predict an increased risk of long-term cognitive impairment in AE patients.
Collapse
Affiliation(s)
- Yingchi Zhang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China
| | - Dianwei Wu
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China
| | - Zhirong Fan
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China
| | - Xiuxiu Leng
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China
| | - Ting Wan
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China
| | - Xiaodan Shi
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China
| | - Xiaomu Wang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China
| | - Xuan Wang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China
| | - Changgeng Song
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China.
| | - Fang Du
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China.
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province 710038, China.
| |
Collapse
|
36
|
Sun YR, Lv QK, Liu JY, Wang F, Liu CF. New perspectives on the glymphatic system and the relationship between glymphatic system and neurodegenerative diseases. Neurobiol Dis 2025; 205:106791. [PMID: 39778750 DOI: 10.1016/j.nbd.2025.106791] [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: 12/13/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Neurodegenerative diseases (ND) are characterized by the accumulation of aggregated proteins. The glymphatic system, through its rapid exchange mechanisms between cerebrospinal fluid (CSF) and interstitial fluid (ISF), facilitates the movement of metabolic substances within the brain, serving functions akin to those of the peripheral lymphatic system. This emerging waste clearance mechanism offers a novel perspective on the removal of pathological substances in ND. This article elucidates recent discoveries regarding the glymphatic system and updates relevant concepts within its model. It discusses the potential roles of the glymphatic system in ND, including Alzheimer's disease (AD), Parkinson's disease (PD), and multiple system atrophy (MSA), and proposes the glymphatic system as a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Yan-Rui Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Qian-Kun Lv
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Jun-Yi Liu
- Department of Neurology, Dushu Lake hospital affilicated to Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| |
Collapse
|
37
|
Wang LJ, Ma X, Ye J. Neuronal activities drive brain waste clearance through the glymphatic system. Acta Pharm Sin B 2025; 15:1193-1195. [PMID: 40177567 PMCID: PMC11959877 DOI: 10.1016/j.apsb.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 04/05/2025] Open
Affiliation(s)
- Lin-jian Wang
- Institute of Trauma and Metabolism, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Xiwen Ma
- Institute of Trauma and Metabolism, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Jianping Ye
- Institute of Trauma and Metabolism, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
38
|
Zhu Y, Zhu J, Ni C, Chen A, Li L, Gao Y, Shoffstall AJ, Yu X. Impact of infusion conditions and anesthesia on CSF tracer dynamics in mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634133. [PMID: 39896601 PMCID: PMC11785030 DOI: 10.1101/2025.01.21.634133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Tracer imaging has been instrumental in mapping the brain's solute transport pathways facilitated by cerebrospinal fluid (CSF) flow. However, the impact of tracer infusion parameters on CSF flow remains incompletely understood. This study evaluated the influence of infusion location, rate, and anesthetic regimens on tracer transport using dynamic contrast-enhanced MRI with Gd-DTPA as a CSF tracer. Infusion rate effects were assessed by administering Gd-DTPA into the cisterna magna (ICM) at two rates under isoflurane anesthesia. Anesthetic effects were evaluated by comparing transport patterns between isoflurane and ketamine/xylazine (K/X) anesthesia at the slower rate. Gd-DTPA transport was also examined after lateral ventricle (ICV) infusion, the primary site of CSF production. The results demonstrate that, besides anesthesia, both the location and rate of infusion substantially affected solute transport within the brain. ICV infusion led to rapid, extensive transport into deep brain regions, while slower ICM infusion resulted in more pronounced transport to dorsal brain regions. Cross-correlation and hierarchical clustering analyses of region-specific Gd-DTPA signal time courses revealed that ICM infusion facilitated transport along periarterial spaces, while ICV infusion favored transport across the ventricular-parenchymal interface. These findings underscore the importance of experimental conditions in influencing tracer kinetics and spatial distribution in the brain.
Collapse
Affiliation(s)
- Yuran Zhu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Junqing Zhu
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Chenxin Ni
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Anbang Chen
- Department of Electrical, Computer, and Systems Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yue Gao
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Andrew J. Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xin Yu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
39
|
Nepozitek J, Dusek P, Sonka K. Glymphatic system, sleep, and Parkinson's disease: interconnections, research opportunities, and potential for disease modification. Sleep 2025; 48:zsae251. [PMID: 39450429 PMCID: PMC11725518 DOI: 10.1093/sleep/zsae251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Indexed: 10/26/2024] Open
Affiliation(s)
- Jiri Nepozitek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| | - Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| | - Karel Sonka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| |
Collapse
|
40
|
Meinhold L, Gennari AG, Baumann-Vogel H, Werth E, Schreiner SJ, Ineichen C, Baumann CR, O’Gorman Tuura R. T2 MRI visible perivascular spaces in Parkinson's disease: clinical significance and association with polysomnography measured sleep. Sleep 2025; 48:zsae233. [PMID: 39377177 PMCID: PMC11725513 DOI: 10.1093/sleep/zsae233] [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: 05/23/2024] [Revised: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
Poor sleep quality might contribute to the risk and progression of neurodegenerative disorders via deficient cerebral waste clearance functions during sleep. In this retrospective cross-sectional study, we explore the link between enlarged perivascular spaces (PVS), a putative marker of sleep-dependent glymphatic clearance, with sleep quality and motor symptoms in patients with Parkinson's disease (PD). T2-weighted magnetic resonance imaging (MRI) images of 20 patients and 17 healthy control participants were estimated visually for PVS in the basal ganglia (BG) and centrum semiovale (CSO). The patient group additionally underwent a single-night polysomnography. Readouts included polysomnographic sleep features and slow-wave activity (SWA), a quantitative EEG marker of sleep depth. Associations between PVS counts, PD symptoms (MDS-UPDRS scores), and sleep parameters were evaluated using correlation and regression analyses. Intra- and inter-rater reproducibility was assessed with weighted Cohen`s kappa coefficient. BG and CSO PVS counts in both patients and controls did not differ significantly between groups. In patients, PVS in both brain regions was negatively associated with SWA (1-2 Hz; BG: r(15) = -.58, padj = .015 and CSO: r(15) = -.6, padj = .015). Basal ganglia PVS counts were positively associated with motor symptoms of daily living (IRR = 1.05, CI [1.01, 1.09], p = .007, padj = .026) and antidepressant use (IRR = 1.37, CI [1.05, 1.80], p = .021, padj = .043) after controlling for age. Centrum Semiovale PVS counts in patients were positively associated with a diagnosis of REM sleep behavior disorder (IRR = 1.39, CI [1.06, 1.84], p = .018, padj = .11). These results add to evidence that sleep deterioration may play a role in impairing glymphatic clearance via altered perivascular function, potentially contributing to disease severity in PD patients.
Collapse
Affiliation(s)
- Lena Meinhold
- Center for MR Research, University Children’s Hospital, Zurich, Switzerland
- University of Zurich Sleep & Health Competence Center, Zurich, Switzerland
| | - Antonio G Gennari
- Center for MR Research, University Children’s Hospital, Zurich, Switzerland
| | - Heide Baumann-Vogel
- Zentrum für Soziale Psychiatrie, Psychiatric University Hospital Zurich, Zurich, Switzerland
| | - Esther Werth
- University of Zurich Sleep & Health Competence Center, Zurich, Switzerland
- Department of Neurology, University Hospital, Zurich, Switzerland
| | - Simon J Schreiner
- University of Zurich Sleep & Health Competence Center, Zurich, Switzerland
- Department of Neurology, University Hospital, Zurich, Switzerland
| | | | - Christian R Baumann
- University of Zurich Sleep & Health Competence Center, Zurich, Switzerland
- Department of Neurology, University Hospital, Zurich, Switzerland
| | - Ruth O’Gorman Tuura
- Center for MR Research, University Children’s Hospital, Zurich, Switzerland
- University of Zurich Sleep & Health Competence Center, Zurich, Switzerland
| |
Collapse
|
41
|
Lian X, Liu Z, Gan Z, Yan Q, Tong L, Qiu L, Liu Y, Chen JF, Li Z. Targeting the glymphatic system to promote α-synuclein clearance: a novel therapeutic strategy for Parkinson's disease. Neural Regen Res 2025; 21:01300535-990000000-00661. [PMID: 39819820 PMCID: PMC12094544 DOI: 10.4103/nrr.nrr-d-24-00764] [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: 08/15/2024] [Revised: 08/23/2024] [Accepted: 09/05/2024] [Indexed: 01/19/2025] Open
Abstract
ABSTRACT The excessive buildup of neurotoxic α-synuclein plays a pivotal role in the pathogenesis of Parkinson's disease, highlighting the urgent need for innovative therapeutic strategies to promote α-synuclein clearance, particularly given the current lack of disease-modifying treatments. The glymphatic system, a recently identified perivascular fluid transport network, is crucial for clearing neurotoxic proteins. This review aims to synthesize current knowledge on the role of the glymphatic system in α-synuclein clearance and its implications for the pathology of Parkinson's disease while emphasizing potential therapeutic strategies and areas for future research. The review begins with an overview of the glymphatic system and details its anatomical structure and physiological functions that facilitate cerebrospinal fluid circulation and waste clearance. It summarizes emerging evidence from neuroimaging and experimental studies that highlight the close correlation between the glymphatic system and clinical symptom severity in patients with Parkinson's disease, as well as the effect of glymphatic dysfunction on α-synuclein accumulation in Parkinson's disease models. Subsequently, the review summarizes the mechanisms of glymphatic system impairment in Parkinson's disease, including sleep disturbances, aquaporin-4 impairment, and mitochondrial dysfunction, all of which diminish glymphatic system efficiency. This creates a vicious cycle that exacerbates α-synuclein accumulation and worsens Parkinson's disease. The therapeutic perspectives section outlines strategies for enhancing glymphatic activity, such as improving sleep quality and pharmacologically targeting aquaporin-4 or its subcellular localization. Promising interventions include deep brain stimulation, melatonin supplementation, γ-aminobutyric acid modulation, and non-invasive methods (such as exercise and bright-light therapy), multisensory γ stimulation, and ultrasound therapy. Moreover, identifying neuroimaging biomarkers to assess glymphatic flow as an indicator of α-synuclein burden could refine Parkinson's disease diagnosis and track disease progression. In conclusion, the review highlights the critical role of the glymphatic system in α-synuclein clearance and its potential as a therapeutic target in Parkinson's disease. It advocates for further research to elucidate the specific mechanisms by which the glymphatic system clears misfolded α-synuclein and the development of imaging biomarkers to monitor glymphatic activity in patients with Parkinson's disease. Findings from this review suggest that enhancing glymphatic clearance is a promising strategy for reducing α-synuclein deposits and mitigating the progression of Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoyue Lian
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhenghao Liu
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zuobin Gan
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qingshan Yan
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Luyao Tong
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Linan Qiu
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuntao Liu
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiang-fan Chen
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhihui Li
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
42
|
Ghanizada H, Nedergaard M. The glymphatic system. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:161-170. [PMID: 40122623 DOI: 10.1016/b978-0-443-19104-6.00006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The glymphatic system, a brain-wide network-supporting cerebrospinal fluid (CSF) and interstitial fluid (ISF) exchange, is essential for removing metabolic waste from the brain. This system's proper functioning is crucial for maintaining neural health and preventing the accumulation of harmful substances that can lead to neurodegenerative diseases. This chapter explores the glymphatic system's mechanisms, its dysfunction in various neurologic disorders, and potential therapeutic strategies. Recent discoveries reveal the glymphatic system's involvement in aging, sleep, cerebral edema, and conditions, such as Alzheimer, Parkinson, Huntington diseases, amyotrophic lateral sclerosis, small vessel disease, hydrocephalus, migraine, stroke, traumatic brain injury, and psychiatric disorders, where impaired waste clearance contributes to disease pathogenesis. Moreover, therapeutic interventions targeting glymphatic dysfunction present promising avenues for mitigating the effects of neurodegenerative diseases. The chapter underscores the potential of integrating glymphatic research into broader clinical practices, offering new strategies for disease management and prevention.
Collapse
Affiliation(s)
- Hashmat Ghanizada
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
43
|
Peng W, Yuan Y, Lei J, Zhao Y, Li Y, Qu Q, Wang J. Long-Term High-Fat Diet Impairs AQP4-Mediated Glymphatic Clearance of Amyloid Beta. Mol Neurobiol 2025; 62:1079-1093. [PMID: 38958889 DOI: 10.1007/s12035-024-04320-3] [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: 03/04/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
As a risk factor for Alzheimer's disease (AD), studies have demonstrated that long-term high-fat diet (HFD) could accelerate the deposition of amyloid beta (Aβ) in the brain. The glymphatic system plays a critical role in Aβ clearance from the brain. However, studies investigating the effects of long-term HFD on glymphatic function have reported paradoxical outcomes, and whether glymphatic dysfunction is involved in the disturbance of Aβ clearance in long-term HFD-fed mice has not been determined. In the present study, we injected fluorescently labeled Aβ into the hippocampus and found that Aβ clearance was decreased in HFD-fed mice. We found that long-term HFD-fed mice had decreased glymphatic function by injecting fluorescent tracers into the cisterna magna and corpus striatum. In long-term HFD-fed mice, aquaporin-4 (AQP4) polarization in the cortex was disrupted, and glymphatic clearance activity was positively correlated with the AQP4 polarization index. In HFD-fed mice, the disturbance of Aβ clearance from the hippocampus was exacerbated by TGN-020, a specific inhibitor of AQP4, whereas TGN-073, an enhancer of AQP4, ameliorated it. These findings suggest that long-term HFD disrupts Aβ clearance by inhibiting AQP4-mediated glymphatic function. The underlying mechanism may involve the disruption of AQP4 polarization.
Collapse
Affiliation(s)
- Wei Peng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Ye Yuan
- 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
| | - Yan Li
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 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.
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China.
| |
Collapse
|
44
|
Schulz LN, Varghese A, Michenkova M, Wedemeyer M, Pindrik JA, Leonard JR, Garcia-Bonilla M, McAllister JP, Cassady K, Wilson RK, Mardis ER, Limbrick DD, Isaacs AM. Neuroinflammatory pathways and potential therapeutic targets in neonatal post-hemorrhagic hydrocephalus. Pediatr Res 2024:10.1038/s41390-024-03733-z. [PMID: 39725707 DOI: 10.1038/s41390-024-03733-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Post-hemorrhagic hydrocephalus (PHH) is a severe complication in premature infants following intraventricular hemorrhage (IVH). It is characterized by abnormal cerebrospinal fluid (CSF) accumulation, disrupted CSF dynamics, and elevated intracranial pressure (ICP), leading to significant neurological impairments. OBJECTIVE This review provides an overview of recent molecular insights into the pathophysiology of PHH and evaluates emerging therapeutic approaches aimed at addressing its underlying mechanisms. METHODS Recent studies were reviewed, focusing on molecular and cellular mechanisms implicated in PHH, including neuroinflammatory pathways, immune mediators, and regulatory genes. The potential of advanced technologies such as whole genome/exome sequencing, proteomics, epigenetics, and single-cell transcriptomics to identify key molecular targets was also analyzed. RESULTS PHH has been strongly linked to neuroinflammatory processes triggered by the degradation of blood byproducts. These processes involve cytokines, chemokines, the complement system, and other immune mediators, as well as regulatory genes and epigenetic mechanisms. Current treatments, primarily surgical CSF diversion, do not address the underlying molecular pathology. Emerging therapies, such as mesenchymal stem cell-based interventions, show promise in modulating immune responses and mitigating neurological damage. However, concerns about the safety of these novel approaches in neonatal populations and their potential effects on brain development remain unresolved. CONCLUSIONS Advanced molecular tools and emerging therapies have the potential to transform the treatment of PHH by targeting its underlying pathophysiology. Further research is needed to validate these approaches, enhance their safety profiles, and improve outcomes for infants with PHH. IMPACT STATEMENT 1. This review elucidates the molecular complexities of post-hemorrhagic hydrocephalus (PHH) by examining specific immune pathways and their impact on disease pathogenesis and progression. 2. It outlines the application of genomic, epigenomic, and proteomic technologies to identify critical molecular targets in PHH, setting the stage for innovative, targeted therapeutic approaches that could improve the outcomes of neonates affected by PHH. 3. It discusses the potential of gene and stem cell therapies in treating PHH, offering non-surgical alternatives and focusing on the underlying neuroinflammatory mechanisms.
Collapse
Affiliation(s)
- Lauren N Schulz
- Department of Neurological Surgery, Ohio State University Medical Center, Columbus, OH, USA
| | - Aaron Varghese
- Department of Undergraduate Studies, Miami University, Oxford, OH, USA
| | - Marie Michenkova
- Medical Scientist Training Program, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Michelle Wedemeyer
- Department of Neurological Surgery, Ohio State University Medical Center, Columbus, OH, USA
- Division of Neurological Surgery, Nationwide Children's Hospital, Columbus, OH, USA
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Jonathan A Pindrik
- Department of Neurological Surgery, Ohio State University Medical Center, Columbus, OH, USA
- Division of Neurological Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Jeffrey R Leonard
- Department of Neurological Surgery, Ohio State University Medical Center, Columbus, OH, USA
- Division of Neurological Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Maria Garcia-Bonilla
- Department of Neurosurgery, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - James Pat McAllister
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin Cassady
- Division of Infectious Disease, Nationwide Children's Hospital, Columbus, OH, USA
- Center for Childhood Cancer Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Richard K Wilson
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Elaine R Mardis
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - David D Limbrick
- Medical Scientist Training Program, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Neurosurgery, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Albert M Isaacs
- Department of Neurological Surgery, Ohio State University Medical Center, Columbus, OH, USA.
- Division of Neurological Surgery, Nationwide Children's Hospital, Columbus, OH, USA.
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
45
|
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
|
46
|
Inami S, Koh K. Sleep induced by mechanosensory stimulation provides cognitive and health benefits in Drosophila. Sleep 2024; 47:zsae226. [PMID: 39331490 DOI: 10.1093/sleep/zsae226] [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: 07/10/2024] [Revised: 09/08/2024] [Indexed: 09/29/2024] Open
Abstract
STUDY OBJECTIVES Sleep is a complex phenomenon regulated by various factors, including sensory input. Anecdotal observations have suggested that gentle rocking helps babies fall asleep, and experimental studies have verified that rocking promotes sleep in both humans and mice. Recent studies have expanded this understanding, demonstrating that gentle vibration also induces sleep in Drosophila. Natural sleep serves multiple functions, including learning and memory, synaptic downscaling, and reduction of harmful substances associated with neurodegenerative diseases. Here, we investigated whether vibration-induced sleep (VIS) provides similar cognitive and health benefits in Drosophila. METHODS We administered gentle vibration to flies that slept very little due to a forced activation of wake-promoting neurons and investigated how the vibration influenced learning and memory in the courtship conditioning paradigm. Additionally, we examined the effects of VIS on synaptic downscaling by counting synaptic varicosities of select neurons. Finally, we determined whether vibration could induce sleep in Drosophila models of Alzheimer's disease (AD) and suppress the accumulation of Amyloid β (Aβ) and Tubulin Associated Unit (TAU). RESULTS VIS enhanced performance in a courtship conditioning paradigm and reduced the number of synaptic varicosities in select neurons. Moreover, vibration improved sleep in Drosophila models of AD, reducing Aβ and TAU levels. CONCLUSIONS Mechanosensory stimulation offers a promising noninvasive avenue for enhancing sleep, potentially providing associated cognitive and health benefits.
Collapse
Affiliation(s)
- Sho Inami
- Department of Neuroscience and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, USA
| | - Kyunghee Koh
- Department of Neuroscience and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
47
|
Toscano JD, Wu C, Ladrón-de-Guevara A, Du T, Nedergaard M, Kelley DH, Karniadakis GE, Boster KAS. Inferring in vivo murine cerebrospinal fluid flow using artificial intelligence velocimetry with moving boundaries and uncertainty quantification. Interface Focus 2024; 14:20240030. [PMID: 39649446 PMCID: PMC11621842 DOI: 10.1098/rsfs.2024.0030] [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: 08/27/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 12/10/2024] Open
Abstract
Cerebrospinal fluid (CSF) flow is crucial for clearing metabolic waste from the brain, a process whose dysregulation is linked to neurodegenerative diseases like Alzheimer's. Traditional approaches like particle tracking velocimetry (PTV) are limited by their reliance on single-plane two-dimensional measurements, which fail to capture the complex dynamics of CSF flow fully. To overcome these limitations, we employ artificial intelligence velocimetry (AIV) to reconstruct three-dimensional velocities, infer pressure and wall shear stress and quantify flow rates. Given the experimental nature of the data and inherent variability in biological systems, robust uncertainty quantification (UQ) is essential. Towards this end, we have modified the baseline AIV architecture to address aleatoric uncertainty caused by noisy experimental data, enhancing our measurement refinement capabilities. We also implement UQ for the model and epistemic uncertainties arising from the governing equations and network representation. Towards this end, we test multiple governing laws, representation models and initializations. Our approach not only advances the accuracy of CSF flow quantification but also can be adapted to other applications that use physics-informed machine learning to reconstruct fields from experimental data, providing a versatile tool for inverse problems.
Collapse
Affiliation(s)
- Juan Diego Toscano
- Division of Applied Mathematics, Brown University, Providence, RI02912, USA
| | - Chenxi Wu
- School of Engineering, Brown University, Providence, RI02912, USA
| | - Antonio Ladrón-de-Guevara
- Center for Translational Neuromedicine and Department of Neuroscience, University of Rochester Medical Center, Rochester, NY14627, USA
| | - Ting Du
- Center for Translational Neuromedicine and Department of Neuroscience, University of Rochester Medical Center, Rochester, NY14627, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine and Department of Neuroscience, University of Rochester Medical Center, Rochester, NY14627, USA
| | - Douglas H. Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, NY14627, USA
| | - George Em Karniadakis
- Division of Applied Mathematics, Brown University, Providence, RI02912, USA
- School of Engineering, Brown University, Providence, RI02912, USA
| | | |
Collapse
|
48
|
Kameya N, Sakai I, Saito K, Hamabe-Horiike T, Shinmyo Y, Nakada M, Okuda S, Kawasaki H. Evolutionary changes leading to efficient glymphatic circulation in the mammalian brain. Nat Commun 2024; 15:10048. [PMID: 39632840 PMCID: PMC11618516 DOI: 10.1038/s41467-024-54372-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
The functional significance of the morphological and genetic changes that occurred in the brain during evolution is not fully understood. Here we show the relationships between evolutionary changes of the brain and glymphatic circulation. We establish a mathematical model to simulate glymphatic circulation in the cerebral hemispheres, and our results show that cortical neurons accumulate in areas of the cerebral hemispheres where glymphatic circulation is highly efficient. We also find that cortical folds markedly enhance the efficiency of glymphatic circulation in the cerebral hemispheres. Furthermore, our in vivo study using ferrets reveals sulcus-dominant cerebrospinal fluid (CSF) influx, which enhances the efficiency of glymphatic circulation in the enlarged cerebral hemispheres of gyrencephalic brains. Sulcus-dominant CSF influx is mediated by preferential expression of aquaporin-4 in sulcal regions, and similar expression patterns of aquaporin-4 are also found in human cerebral hemispheres. These results indicate that evolutionary changes in the cerebral hemispheres are related to improved efficiency of glymphatic circulation. It seems plausible that the efficiency of glymphatic circulation is an important factor determining the evolutionary trajectory of the cerebral hemispheres.
Collapse
Affiliation(s)
- Narufumi Kameya
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Itsuki Sakai
- Nano Life Science Institute, Kanazawa University, Ishikawa, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Mitsutoshi Nakada
- Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Ishikawa, Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Satoru Okuda
- Nano Life Science Institute, Kanazawa University, Ishikawa, Japan.
- Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Ishikawa, Japan.
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.
- Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
49
|
Lin R, Luo R, Yu X, Zou J, Huang X, Guo Y. Depleting parenchymal border macrophages alleviates cerebral edema and neuroinflammation following status epilepticus. J Transl Med 2024; 22:1094. [PMID: 39623451 PMCID: PMC11613707 DOI: 10.1186/s12967-024-05912-2] [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: 08/12/2024] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Status epilepticus (SE) is a common severe neurological emergency. Cerebral edema caused by SE is unavoidable and may exacerbate epilepsy. Recent studies have identified cerebrospinal fluid (CSF) as a crucial fluid source of initial cerebral edema following ischemic stroke and cardiac arrest. Moreover, synchronized neuronal firings drive CSF influx into interstitial fluid (ISF). Parenchymal border macrophages (PBMs) have been found to play a role in regulating CSF flow dynamics. However, the involvement of CSF and PBMs in cerebral edema during SE remains unclear. Here, we investigated the fluid source of cerebral edema in the initial phase of SE with the role of PBMs involved. METHODS Lithium chloride-pilocarpine was used to induce SE in C57BL/6 J mice. Electroencephalogram (EEG) was acquired to assess changes in relative EEG power pre- and post-seizure onset. Apparent diffusion coefficient (ADC) maps reconstructed from diffusion-weighted imaging (DWI) were utilized to evaluate cytotoxic edema. Blood-brain barrier (BBB) permeability was examined using sodium fluorescein (NaFl). CSF tracer influx into the brain was assessed by transcranial imaging and brain slices. PBMs were depleted using clodronate liposomes. Immunohistochemistry was used to evaluate PBM depletion, severity of vasogenic edema, inflammation, and neuronal damage. RESULTS During the initial stage of SE, relative EEG power sharply increased and ADC values significantly decreased. Concurrently, CSF tracer influx into the cortex significantly elevated, though NaFl leakage from blood to brain parenchyma did not evidently alter. Following depletion of PBM, CSF influx declined but AQP4 expression and polarization remained unaffected. Post-PBM depletion, there was no significant alteration in relative EEG power, yet CSF influx decreased substantially during the initial stage of SE. The degree of ADC decline lessened, IgG extravasation after SE decreased, activated microglia and proliferating astrocytes count fell, and neuronal damage post-SE alleviated. CONCLUSIONS CSF appeared to contribute to cerebral edema in SE. Depletion of PBM alleviated cytotoxic edema in the initial phase of SE, and subsequent vasogenic edema, inflammatory response and neurological damage were reduced. These findings may provide potential novel strategies for treating cerebral edema following SE.
Collapse
Affiliation(s)
- Renbao Lin
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Rui Luo
- Dermatology Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Xinyue Yu
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Junjie Zou
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China.
| | - Xiaowei Huang
- Dongguan University of Technology, Dongguan, 510282, China.
| | - Yanwu Guo
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
50
|
Hu P, Yuan Y, Zou Y, Xiong R, Dai J, Zhao X, Xie L, Tang X. Alterations in the DTI-ALPS index and choroid plexus volume are associated with clinical symptoms in participants with narcolepsy type 1. Sleep Med 2024; 124:471-478. [PMID: 39427503 DOI: 10.1016/j.sleep.2024.10.019] [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: 06/03/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Narcolepsy type 1 (NT1) is a sleep disorder characterized by excessive daytime sleepiness accompanied by cataplexy. Sleep disorders have been shown to affect the glymphatic system. This study aimed to evaluate changes in the diffusion tensor imaging along the perivascular space (DTI-ALPS) index and choroid plexus (CP) volume in NT1 participants, and to further explore their clinical significance. METHODS We prospectively enrolled participants diagnosed with NT1 based on cerebrospinal fluid hypocretin-1 concentration and multiple sleep latency tests at our hospital. All participants underwent MRI to allow analysis of the DTI-ALPS index and CP volume. We subsequently performed correlation analyses between the DTI-ALPS index, CP volume, and important clinical parameters, including the Epworth Sleepiness Scale (ESS) score, Narcolepsy Severity Scale (NSS) score, stage rapid eye movement sleep (REM) ratio, stage 1 non-REM (N1) ratio, stage 2 non-REM (N2) ratio, and stage 3 non-REM (N3) ratio, among the NT1 participants. Inter-group and correlation analyses of DTI-ALPS index and CP volume were performed using age, sex, body mass index, and lateral ventricle volume as covariates. RESULTS This study enrolled 41 NT1 participants and 42 healthy controls (HC). The DTI-ALPS index of NT1 participants was significantly lower than HC (1.444 ± 0.119 vs.1.661 ± 0.135, P < 0.001), while the CP volume of NT1 participants was significantly larger than those of HC (0.831 ± 0.146 vs. 0.645 ± 0.137, P < 0.001). The DTI-ALPS index was negatively correlated with both the ESS (PFDR-corrected<0.001) and NSS scores (PFDR-corrected = 0.010), but positively correlated with the Stage N3 ratio (PFDR-corrected = 0.033). The CP volume of NT1 participants was positively correlated with ESS (PFDR-corrected = 0.047) and NSS scores (PFDR-corrected = 0.047), but negatively correlated with the stage N3 ratio (PFDR-corrected = 0.047). CONCLUSION Our study suggests that the DTI-ALPS index was lower and CP volume was larger in NT1 participants. The DTI-ALPS index and CP volume in the NT1 participants were related to disease severity and sleep structure. These findings may provide new insights into the mechanisms underlying NT1.
Collapse
Affiliation(s)
- Pengxin Hu
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; Intelligent Medical Imaging of Jiangxi Key Laboratory, Nanchang, 330006, China
| | - Yuqing Yuan
- Department of Neurology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yu Zou
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; Intelligent Medical Imaging of Jiangxi Key Laboratory, Nanchang, 330006, China
| | - Ruifang Xiong
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; Intelligent Medical Imaging of Jiangxi Key Laboratory, Nanchang, 330006, China
| | - Jiankun Dai
- MR Research, GE Healthcare, Beijing, 100000, China
| | - Xihai Zhao
- Biomedical Imaging Research Center, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Liang Xie
- Department of Neurology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xiaoping Tang
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China; Intelligent Medical Imaging of Jiangxi Key Laboratory, Nanchang, 330006, China; Biomedical Imaging Research Center, School of Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|