1
|
Xu X, Su X, Xu L, Chen X, Li D, Liu J, Dai K, Liu J, Jiang Y, Peng F. Impaired glymphatic function in autoimmune glial fibrillary acidic protein astrocytopathy: a prospective analysis. Mult Scler Relat Disord 2025; 99:106447. [PMID: 40267745 DOI: 10.1016/j.msard.2025.106447] [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: 01/28/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND To evaluate the glymphatic dysfunction and its association with disease severity in autoimmune glial fibrillary acidic protein astrocytopathy (A-GFAP-A) patients, and to determine its clinical predictors. METHODS A total of 20 A-GFAP-A patients and 20 healthy controls (HC) were included. All participants underwent magnetic resonance imaging, and glymphatic function was assessed using the diffusion tensor imaging along the perivascular space (DTI-ALPS) index. Modified Rankin Scale (mRS) scores were recorded at baseline and 4 weeks post-immunotherapy. Multiple linear regression analysis was conducted to identify independent predictors of short-term prognosis. RESULTS The baseline DTI-ALPS index was significantly lower in A-GFAP-A patients compared to HC (mean ± SD: 1.50 ± 0.06 vs. 1.62 ± 0.04 [CI -0.16, -0.08], p = 0.003), after adjusting for confounding factors. Four weeks after immunotherapy, the DTI-ALPS index significantly increased (mean ± SD: 1.52 ± 0.14 vs. 1.59 ± 0.17 [CI 0.01, 0.14], p = 0.037). A significant negative correlation was observed between the residuals of the baseline DTI-ALPS index and the baseline mRS scores (r = -0.50 [CI -0.77, -0.07], p = 0.025). The baseline DTI-ALPS index was identified as an independent predictor of short-term prognosis (coefficient = -3.43 [CI -6.68, -0.04], p = 0.048). CONCLUSIONS This study indicates that A-GFAP-A patients exhibit significant glymphatic dysfunction, as detected by the DTI-ALPS index, which is related to the severity of the disease. The DTI-ALPS index may serve as a biomarker for monitoring disease progression and as a predictor of short-term prognosis in A-GFAP-A patients.
Collapse
Affiliation(s)
- Xiaofeng Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Xiaohong Su
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Li Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Xiaodong Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China; Department of Infectious Diseases, The Fifth People's Hospital of Ganzhou, Ganzhou, Jiangxi, China
| | - Dongcheng Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Junyu Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Kai Dai
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Jia Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China.
| | - Ying Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China.
| | - Fuhua Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Itkin M, Horak J, Pascual JL, Chang CWJ, Lile D, Tomita B, Bass GA, Kovach SJ, Kaplan LJ. Disorders of Lymphatic Architecture and Flow in Critical Illness. Crit Care Med 2025; 53:e665-e682. [PMID: 39791972 DOI: 10.1097/ccm.0000000000006561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
OBJECTIVES To provide a narrative review of disordered lymphatic dynamics and its impact on critical care relevant condition management. DATA SOURCES Detailed search strategy using PubMed and Ovid Medline for English language articles (2013-2023) describing congenital or acquired lymphatic abnormalities including lymphatic duct absence, injury, leak, or obstruction and their associated clinical conditions that might be managed by a critical care medicine practitioner. STUDY SELECTION Studies that specifically addressed abnormalities of lymphatic flow and their management were selected. The search strategy time frame was limited to the last 10 years to enhance relevance to current practice. DATA EXTRACTION Relevant descriptions or studies were reviewed, and abstracted data were parsed into structural or functional etiologies, congenital or acquired conditions, and their management within critical care spaces in an acute care facility. DATA SYNTHESIS Abnormal lymph flow may be identified stemming from congenital lymphatic anomalies including lymphatic structure absence as well as acquired obstruction or increased flow from clinical entities or acute therapy. Macro- and microsurgical as well as interventional radiological techniques may address excess, inadequate, or obstructed lymph flow. Patients with deranged lymph flow often require critical care, and those who require critical care may concomitantly demonstrate deranged lymph flow that adversely impacts care. CONCLUSIONS Critical care clinicians ideally demonstrate functional knowledge of conditions that are directly related to, or are accompanied by, deranged lymphatic dynamics to direct timely diagnostic and therapeutic interventions during a patient's ICU care episode.
Collapse
Affiliation(s)
- Maxim Itkin
- Division of Interventional Radiology, Department of Radiology, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Jiri Horak
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jose L Pascual
- Division of Trauma, Surgical Critical Care and Emergency Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Cherylee W J Chang
- Department of Neurology, Division of Neurocritical Care, Duke University, Durham, NC
| | - Deacon Lile
- Division of Trauma, Surgical Critical Care and Emergency Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Beverly Tomita
- Carle Illinois College of Medicine, University of Illinois Urbana-Champlain, Urbana, IL
| | - Gary Alan Bass
- Division of Trauma, Surgical Critical Care and Emergency Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephen J Kovach
- Division of Plastic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Lewis J Kaplan
- Division of Trauma, Surgical Critical Care and Emergency Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
5
|
Lüthi A, Nedergaard M. Anything but small: Microarousals stand at the crossroad between noradrenaline signaling and key sleep functions. Neuron 2025; 113:509-523. [PMID: 39809276 DOI: 10.1016/j.neuron.2024.12.009] [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: 01/29/2024] [Revised: 09/16/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Continuous sleep restores the brain and body, whereas fragmented sleep harms cognition and health. Microarousals (MAs), brief (3- to 15-s-long) wake intrusions into sleep, are clinical markers for various sleep disorders. Recent rodent studies show that MAs during healthy non-rapid eye movement (NREM) sleep are driven by infraslow fluctuations of noradrenaline (NA) in coordination with electrophysiological rhythms, vasomotor activity, cerebral blood volume, and glymphatic flow. MAs are hence part of healthy sleep dynamics, raising questions about their biological roles. We propose that MAs bolster NREM sleep's benefits associated with NA fluctuations, according to an inverted U-shaped curve. Weakened noradrenergic fluctuations, as may occur in neurodegenerative diseases or with sleep aids, reduce MAs, whereas exacerbated fluctuations caused by stress fragment NREM sleep and collapse NA signaling. We suggest that MAs are crucial for the restorative and plasticity-promoting functions of sleep and advance our insight into normal and pathological arousal dynamics from sleep.
Collapse
Affiliation(s)
- Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
6
|
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
|
7
|
Oldham MA, Spira AP, Yurcheshen M, Pigeon WR, Palanca BJA, Lee HB. Novel applications of sleep pharmacology as delirium therapeutics. Sleep Med Rev 2025; 79:102016. [PMID: 39541802 PMCID: PMC11750618 DOI: 10.1016/j.smrv.2024.102016] [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: 07/22/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Sleep-wake and circadian disruption (SCD) is a core feature of delirium. It has been hypothesized that SCD contributes to delirium pathogenesis; therefore, interventions that prevent or reverse SCD represent an array of promising opportunities in relation to delirium. This review explores the relationship between sleep-wake/circadian physiology and delirium pathophysiology with a focus on neurotransmitter systems. Across potential targets aimed at preventing or treating delirium, three broad approaches are considered: 1. Pharmacological mechanisms that contribute to physiological sleep may preserve or restore next-day cognition in patients with or at risk for delirium (e.g., alpha 2 agonists, dopamine 2 antagonists, serotonin 2 A antagonists, dual orexin receptor antagonists, or GHB agonists); 2. Pharmacological mechanisms that promote wakefulness during the day may combat hypoactive delirium (e.g., adenosine 2 A antagonists, dopamine transporter antagonists, orexin agonists, histamine 3 antagonists); and 3. Melatonergic and other circadian interventions could strengthen the phase or amplitude of circadian rhythms and ensure appropriately entrained timing in patients with or at risk for delirium (e.g., as informed by a person's preexisting circadian phase).
Collapse
Affiliation(s)
- Mark A Oldham
- University of Rochester Medical Center, Rochester, NY, USA.
| | | | | | - Wilfred R Pigeon
- University of Rochester Medical Center, Rochester, NY, USA; Center of Excellence for Suicide Prevention, U.S. Department of Veterans Affairs, USA
| | | | - Hochang B Lee
- University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
8
|
Chen Y, Guo H, Sun X, Wang S, Zhao M, Gong J, He A, Li J, Liu Y, Wang Z. Melatonin Regulates Glymphatic Function to Affect Cognitive Deficits, Behavioral Issues, and Blood-Brain Barrier Damage in Mice After Intracerebral Hemorrhage: Potential Links to Circadian Rhythms. CNS Neurosci Ther 2025; 31:e70289. [PMID: 39981743 PMCID: PMC11843476 DOI: 10.1111/cns.70289] [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: 08/27/2024] [Revised: 01/13/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a life-threatening cerebrovascular disorder with no specific pharmacological treatment. ICH causes significant behavioral deficits and cognitive impairments. Recent research suggests that circadian rhythm regulation could be a promising therapeutic strategy for ICH. Melatonin has been shown to alleviate glymphatic system (GS) dysfunction by regulating circadian rhythms, thereby improving depressive-like behaviors and postoperative sleep disorders in mice. However, its application in ICH treatment and specific mechanisms are not well understood. METHODS ICH models were created in 8-to-10-week-old mice using collagenase injection. Circadian rhythm modulation was tested with melatonin and luzindole. Behavioral and cognitive impairments were assessed with the modified neurological severity score, corner test, and novel object recognition test. Brain water content was measured by the dry/wet weight method, and cerebral perfusion was assessed by cerebral blood flow measurements. GS function was evaluated using RITC-dextran and Evans blue assays. Immunofluorescence and western blotting were used to analyze GS function and BBB permeability. RESULTS Melatonin restored GS transport after ICH, promoting hematoma and edema absorption, reducing BBB damage, and improving cognitive and behavioral outcomes. However, luzindole partially blocked these benefits and reversed the neuroprotective effects. CONCLUSION Melatonin and luzindole treatment affect GS function, BBB permeability, and cognitive-behavioral outcomes in mice with ICH. The underlying mechanism may involve the regulation of circadian rhythms.
Collapse
Affiliation(s)
- Yunzhao Chen
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
- Department of NeurosurgeryInner Mongolia Autonomous Region People's HospitalHohhotChina
| | - Hexi Guo
- Department of NeurosurgeryOrdos Central HospitalOrdosChina
| | - Xinguo Sun
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
- Department of NeurosurgeryBinzhou People's HospitalBinzhouChina
| | - Shanjun Wang
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
- Department of NeurosurgeryYidu Central Hospital of WeifangQingzhouChina
| | - Mingyu Zhao
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
| | - Junjie Gong
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
| | - Anqi He
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
| | - Jing Li
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
| | - Yuheng Liu
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Neurological InstituteTianjin Medical University General HospitalTianjinChina
| |
Collapse
|
9
|
Hou Y, Ye W, Tang Z, Li F. Anesthetics in pathological cerebrovascular conditions. J Cereb Blood Flow Metab 2025; 45:32-47. [PMID: 39450477 PMCID: PMC11563546 DOI: 10.1177/0271678x241295857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/21/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
The increasing prevalence of pathological cerebrovascular conditions, including stroke, hypertensive encephalopathy, and chronic disorders, underscores the importance of anesthetic considerations for affected patients. Preserving cerebral oxygenation and blood flow during anesthesia is paramount to prevent neurological deterioration. Furthermore, protecting vulnerable neurons from damage is crucial for optimal outcomes. Recent research suggests that anesthetic agents may provide a potentially therapeutic approach for managing pathological cerebrovascular conditions. Anesthetics target neural mechanisms underlying cerebrovascular dysfunction, thereby modulating neuroinflammation, protecting neurons against ischemic injury, and improving cerebral hemodynamics. However, optimal strategies regarding mechanisms, dosage, and indications remain uncertain. This review aims to clarify the physiological effects, mechanisms of action, and reported neuroprotective benefits of anesthetics in patients with various pathological cerebrovascular conditions. Investigating anesthetic effects in cerebrovascular disease holds promise for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuhui Hou
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Wei Ye
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Ziyuan Tang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Fengxian Li
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Chou CC, Lu YA, Weng CH, Lin HJ, Wang IJ, Jou TS, Wang CY, Tsai FJ, Cheng YD, Hsu TJ, Hung YT, Huang YH, Tien PT. The association between antiglaucomatous agents and Alzheimer's disease. Eye (Lond) 2024; 38:3511-3518. [PMID: 39341977 PMCID: PMC11621119 DOI: 10.1038/s41433-024-03348-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 08/03/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
OBJECTIVES To estimate the risk of Alzheimer's disease (AD) associated with long-term use of topical glaucoma medications among middle-aged and older glaucoma patients, and compare the AD risk among various glaucoma subtypes. METHODS This nationwide population-based cohort study utilized insurance claims data from Taiwan's National Health Insurance Research Database between 2008 and 2019. Participants were adults aged 45 years or older either with a diagnosis of glaucoma or without. Those with glaucoma must have received single antiglaucomatous medication (including α2-adrenergic agonists, cholinergic agonists, beta-blockers, prostaglandin analogs, and pilocarpine) for over 90 days. Those with pre-existing AD diagnoses prior to the index date were excluded. RESULTS A total of 202,000 participants were included in the study, with 101,000 in each group (glaucoma and control groups). Glaucoma patients on topical alpha-2 adrenergic agonist monotherapy exhibited a significantly higher AD risk (aHR 1.15, 95% CI = 1.01-1.31) compared to those on beta-blockers. Glaucoma was further categorized into primary open-angle glaucoma (POAG), normal-tension glaucoma (NTG), primary angle-closure glaucoma (PACG), and unspecified glaucoma. Irrespective of the type of glaucoma, individuals with glaucoma had a significantly higher risk of AD compared to those without glaucoma (POAG: aHR 1.23, 95% CI = 1.08-1.40; NTG: aHR 1.49, 95% CI = 1.19-1.85; PACG: aHR 1.35, 95% CI = 1.19-1.52; unspecified glaucoma: aHR 1.36, 95% CI = 1.23-1.50). CONCLUSIONS Topical alpha-2 adrenergic agonists might pose increased AD risk in individuals with glaucoma compared to beta-blockers. Accordingly, their utilization should be undertaken judiciously, especially in middle-aged and older populations. Our findings also indicate glaucoma may increase the risk of AD regardless of glaucoma subtype.
Collapse
Affiliation(s)
- Chien-Chih Chou
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taichung Veterans General Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-An Lu
- Department of Ophthalmology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chien-Hsiang Weng
- Department of Family Medicine, Brown University Warren Alpert Medical School, Providence, RI, USA
- Coastal Medical Lifespan, Providence, RI, USA
| | - Hui-Ju Lin
- Eye Center, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - I-Jong Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tzuu-Shuh Jou
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Yuan Wang
- Department of Ophthalmology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Division of Medical Genetics, China Medical University Children's Hospital, Taichung, Taiwan
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Yih-Dih Cheng
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Tzu-Ju Hsu
- Management Office for Health Data, Clinical Trial Research Center, China Medical University Hospital, Taichung, Taiwan
- Master's Program in Statistics and Actuarial Science, Department of Statistics, Feng Chia University, Taichung, Taiwan
| | - Yu-Tung Hung
- Management Office for Health Data, Clinical Trial Research Center, China Medical University Hospital, Taichung, Taiwan
- Institute of Public Health, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Han Huang
- Management Office for Health Data, Clinical Trial Research Center, China Medical University Hospital, Taichung, Taiwan
- Institute of Public Health, National Cheng Kung University, Tainan, Taiwan
| | - Peng-Tai Tien
- Eye Center, China Medical University Hospital, Taichung, Taiwan.
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
11
|
Fotiadis P, McKinstry-Wu AR, Weinstein SM, Cook PA, Elliott M, Cieslak M, Duda JT, Satterthwaite TD, Shinohara RT, Proekt A, Kelz MB, Detre JA, Bassett DS. Changes in brain connectivity and neurovascular dynamics during dexmedetomidine-induced loss of consciousness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616650. [PMID: 39416182 PMCID: PMC11482825 DOI: 10.1101/2024.10.04.616650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Understanding the neurophysiological changes that occur during loss and recovery of consciousness is a fundamental aim in neuroscience and has marked clinical relevance. Here, we utilize multimodal magnetic resonance neuroimaging to investigate changes in regional network connectivity and neurovascular dynamics as the brain transitions from wakefulness to dexmedetomidine-induced unconsciousness, and finally into early-stage recovery of consciousness. We observed widespread decreases in functional connectivity strength across the whole brain, and targeted increases in structure-function coupling (SFC) across select networks-especially the cerebellum-as individuals transitioned from wakefulness to hypnosis. We also observed robust decreases in cerebral blood flow (CBF) across the whole brain-especially within the brainstem, thalamus, and cerebellum. Moreover, hypnosis was characterized by significant increases in the amplitude of low-frequency fluctuations (ALFF) of the resting-state blood oxygen level-dependent signal, localized within visual and somatomotor regions. Critically, when transitioning from hypnosis to the early stages of recovery, functional connectivity strength and SFC-but not CBF-started reverting towards their awake levels, even before behavioral arousal. By further testing for a relationship between connectivity and neurovascular alterations, we observed that during wakefulness, brain regions with higher ALFF displayed lower functional connectivity with the rest of the brain. During hypnosis, brain regions with higher ALFF displayed weaker coupling between structural and functional connectivity. Correspondingly, brain regions with stronger functional connectivity strength during wakefulness showed greater reductions in CBF with the onset of hypnosis. Earlier recovery of consciousness was associated with higher baseline (awake) levels of functional connectivity strength, CBF, and ALFF, as well as female sex. Across our findings, we also highlight the role of the cerebellum as a recurrent marker of connectivity and neurovascular changes between states of consciousness. Collectively, these results demonstrate that induction of, and emergence from dexmedetomidine-induced unconsciousness are characterized by widespread changes in connectivity and neurovascular dynamics.
Collapse
Affiliation(s)
- Panagiotis Fotiadis
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Andrew R. McKinstry-Wu
- Department of Anesthesiology & Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah M. Weinstein
- Department of Epidemiology and Biostatistics, Temple University College of Public Health, Philadelphia, PA, USA
| | - Philip A. Cook
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Elliott
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Cieslak
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey T. Duda
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Theodore D. Satterthwaite
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing & Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T. Shinohara
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing & Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander Proekt
- Department of Anesthesiology & Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Max B. Kelz
- Department of Anesthesiology & Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John A. Detre
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dani S. Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Electrical & Systems Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
12
|
Xu Y, Teng X, Wei M, Liu Y. TREK-1 channel as a therapeutic target for dexmedetomidine-mediated neuroprotection in cerebral ischemia. Neurogenetics 2024; 25:367-375. [PMID: 38976083 DOI: 10.1007/s10048-024-00772-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Our objective is to explore the protective effect of Dexmedetomidine on brain apoptosis and its mechanism through TREK-1 pathway. Forty male Sprague-Dawley rats were allocated into four groups: Sham, Cerebral Ischemia/Reperfusion Injury (CIRI), 50 µg/kg Dex, and 100 µg/kg Dex. A rat model of middle cerebral artery occlusion (MCAO) was employed to simulate cerebral embolism. Primary cortical neurons were exposed to Dex for 48 h, with some receiving additional treatment with 100 µM yohimbine hydrochloride (YOH) or TREK-1 small interfering RNA (siRNA). Neuronal damage was assessed using hematoxylin and eosin (HE) staining. Cell viability and apoptosis were measured by Cell Counting Kit-8 (CCK8) and flow cytometry, respectively. Protein and gene expression levels of Bcl-2, Bax, and TREK-1 were determined by Western blot and real-time polymerase chain reaction (PCR). Histopathological changes revealed that Dex treatment at both 50 µg/kg and 100 µg/kg significantly mitigated neuronal damage compared to the CIRI group. YOH treatment and Trek1 siRNA significantly reduced cell viability (p < 0.05). The mRNA expression and protein levels of TREK-1 and Bax were remarkably increased, while mRNA expression and protein levels of Bcl-2 was seriously decreased after CIRI modeling. In contrast, Dex treatment at both concentrations led to decreased TREK-1 and Bax expression and increased Bcl-2 expression in primary cortical neurons. Addition of 100 µM YOH and Trek1 siRNA reversed the effects of Dex on apoptosis-related genes (p < 0.05). Dex exerts neuroprotective effects through the TREK-1 pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Yang Xu
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road (Street), NanGang District, Harbin, 150081, Heilongjiang Province, PR China
| | - XiaoDan Teng
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road (Street), NanGang District, Harbin, 150081, Heilongjiang Province, PR China
| | - Ming Wei
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road (Street), NanGang District, Harbin, 150081, Heilongjiang Province, PR China
| | - Yang Liu
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road (Street), NanGang District, Harbin, 150081, Heilongjiang Province, PR China.
| |
Collapse
|
13
|
Xie Z, He Z, Yuan Z, Wang M, Zhou F. The Regulation of Cerebral Lymphatic Drainage in the Transverse Sinus Region of the Mouse Brain. JOURNAL OF BIOPHOTONICS 2024:e202400250. [PMID: 39289863 DOI: 10.1002/jbio.202400250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/13/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
Cerebral lymphatic drainage is an important pathway for metabolic waste clearance in the brain, which plays a crucial role in the progression of central nervous system diseases. Recent studies have shown that norepinephrine (NE) is involved in the regulation of cerebral lymphatic drainage function, but the modulation mechanism remains unknown. In this study, we confirmed that NE rapidly reduced glymphatic influx and enhanced meningeal lymphatic clearance. Moreover, the transverse sinus (TS) was the vital region of cerebral lymphatic drainage regulation by NE. Further analysis revealed that NE inhibition could simultaneously enhance glymphatic drainage and dorsal meningeal lymphatic drainage, mainly acting on the TS region. This study demonstrated that the cerebral lymphatic drainage system can be regulated by NE, with the TS region serving as the primary modulating site. The findings provide a potential regulatory target for the amelioration of neurological diseases associated with cerebral lymphatic drainage function.
Collapse
Affiliation(s)
- Zengjun Xie
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, China
| | - Zhihui He
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, China
| | - Zhen Yuan
- Faculty of Health Sciences, Center for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR, China
| | - Miao Wang
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, China
| | - Feifan Zhou
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, China
| |
Collapse
|
14
|
Zhang R, Li J, Li X, Zhang S. Therapeutic approaches to CNS diseases via the meningeal lymphatic and glymphatic system: prospects and challenges. Front Cell Dev Biol 2024; 12:1467085. [PMID: 39310229 PMCID: PMC11413538 DOI: 10.3389/fcell.2024.1467085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
The brain has traditionally been considered an "immune-privileged" organ lacking a lymphatic system. However, recent studies have challenged this view by identifying the presence of the glymphatic system and meningeal lymphatic vessels (MLVs). These discoveries offer new opportunities for waste clearance and treatment of central nervous system (CNS) diseases. Various strategies have been developed based on these pathways, including modulation of glymphatic system function, enhancement of meningeal lymphatic drainage, and utilization of these routes for drug delivery. Consequently, this review explores the developmental features and physiological roles of the cerebral lymphatic system as well as its significance in various CNS disorders. Notably, strategies for ameliorating CNS diseases have been discussed with a focus on enhancing glymphatic system and MLVs functionality through modulation of physiological factors along with implementing pharmacological and physical treatments. Additionally, emphasis is placed on the potential use of the CNS lymphatic system in drug delivery while envisioning future directions in terms of mechanisms, applications, and translational research.
Collapse
Affiliation(s)
| | | | | | - Si Zhang
- Department of Neurosurgery, Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Persson NDÅ, Lohela TJ, Mortensen KN, Rosenholm M, Li Q, Weikop P, Nedergaard M, Lilius TO. Anesthesia Blunts Carbon Dioxide Effects on Glymphatic Cerebrospinal Fluid Dynamics in Mechanically Ventilated Rats. Anesthesiology 2024; 141:338-352. [PMID: 38787687 DOI: 10.1097/aln.0000000000005039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
BACKGROUND Impaired glymphatic clearance of cerebral metabolic products and fluids contribute to traumatic and ischemic brain edema and neurodegeneration in preclinical models. Glymphatic perivascular cerebrospinal fluid flow varies between anesthetics possibly due to changes in vasomotor tone and thereby in the dynamics of the periarterial cerebrospinal fluid (CSF)-containing space. To better understand the influence of anesthetics and carbon dioxide levels on CSF dynamics, this study examined the effect of periarterial size modulation on CSF distribution by changing blood carbon dioxide levels and anesthetic regimens with opposing vasomotor influences: vasoconstrictive ketamine-dexmedetomidine (K/DEX) and vasodilatory isoflurane. METHODS End-tidal carbon dioxide (ETco2) was modulated with either supplemental inhaled carbon dioxide to reach hypercapnia (Etco2, 80 mmHg) or hyperventilation (Etco2, 20 mmHg) in tracheostomized and anesthetized female rats. Distribution of intracisternally infused radiolabeled CSF tracer 111In-diethylamine pentaacetate was assessed for 86 min in (1) normoventilated (Etco2, 40 mmHg) K/DEX; (2) normoventilated isoflurane; (3) hypercapnic K/DEX; and (4) hyperventilated isoflurane groups using dynamic whole-body single-photon emission tomography. CSF volume changes were assessed with magnetic resonance imaging. RESULTS Under normoventilation, cortical CSF tracer perfusion, perivascular space size around middle cerebral arteries, and intracranial CSF volume were higher under K/DEX compared with isoflurane (cortical maximum percentage of injected dose ratio, 2.33 [95% CI, 1.35 to 4.04]; perivascular size ratio 2.20 [95% CI, 1.09 to 4.45]; and intracranial CSF volume ratio, 1.90 [95% CI, 1.33 to 2.71]). Under isoflurane, tracer was directed to systemic circulation. Under K/DEX, the intracranial tracer distribution and CSF volume were uninfluenced by hypercapnia compared with normoventilation. Intracranial CSF tracer distribution was unaffected by hyperventilation under isoflurane despite a 28% increase in CSF volume around middle cerebral arteries. CONCLUSIONS K/DEX and isoflurane overrode carbon dioxide as a regulator of CSF flow. K/DEX could be used to preserve CSF space and dynamics in hypercapnia, whereas hyperventilation was insufficient to increase cerebral CSF perfusion under isoflurane. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Niklas Daniel Åke Persson
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; 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
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Anaesthesiology, Intensive Care and Pain Medicine, HUS Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Kristian Nygaard Mortensen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marko Rosenholm
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Qianliang Li
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| | - Tuomas O Lilius
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Emergency Medicine and Services, HUS Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Li W, Sun B, Zhang X, Liu T, Zhu W, Liu X, Qu D, Hu C, Zhu S, Wang H. Near-Infrared-II Imaging Revealed Hypothermia Regulates Neuroinflammation Following Brain Injury by Increasing the Glymphatic Influx. ACS NANO 2024; 18:13836-13848. [PMID: 38753820 DOI: 10.1021/acsnano.4c02652] [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: 05/18/2024]
Abstract
Advanced in vivo imaging techniques have facilitated the comprehensive visual exploration of animal biological processes, leading to groundbreaking discoveries such as the glymphatic system. However, current limitations of macroscopic imaging techniques impede the precise investigation of physiological parameters regulating this specialized lymphatic transport system. While NIR-II fluorescence imaging has demonstrated advantages in peripheral lymphatic imaging, there are few reports regarding its utilization in the glymphatic system. To address this, a noninvasive transcranial macroscopic NIR-II fluorescence imaging model is developed using a cyanine dye-protein coupled nanoprobe. NIR-II imaging with high temporal and spatial resolution reveals that hypothermia can increase the glymphatic influx by reducing the flow rate of cerebrospinal fluid. In addition, respiratory rate, respiratory amplitude, and heart rate all play a role in regulating the glymphatic influx. Thus, targeting the glymphatic influx may alter the trajectory of immune inflammation following brain injury, providing therapeutic prospects for treating brain injury with mild hypothermia.
Collapse
Affiliation(s)
- Wenzhong Li
- Department of Neurosurgery, Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan 030001, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Bin Sun
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun 130012, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Xiaoyu Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130012, P.R. China
| | - Tianyi Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130012, P.R. China
| | - Wenhao Zhu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130012, P.R. China
| | - Xiaolong Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130012, P.R. China
| | - Donghao Qu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130012, P.R. China
| | - Changchen Hu
- Department of Neurosurgery, Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan 030001, P.R. China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun 130012, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Haifeng Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130012, P.R. China
| |
Collapse
|
17
|
Sun B, Fang D, Li W, Li M, Zhu S. NIR-II nanoprobes for investigating the glymphatic system function under anesthesia and stroke injury. J Nanobiotechnology 2024; 22:200. [PMID: 38654299 DOI: 10.1186/s12951-024-02481-w] [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: 01/14/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024] Open
Abstract
The glymphatic system plays an important role in the transportation of cerebrospinal fluid (CSF) and the clearance of metabolite waste in brain. However, current imaging modalities for studying the glymphatic system are limited. Herein, we apply NIR-II nanoprobes with non-invasive and high-contrast advantages to comprehensively explore the function of glymphatic system in mice under anesthesia and cerebral ischemia-reperfusion injury conditions. Our results show that the supplement drug dexmedetomidine (Dex) enhances CSF influx in the brain, decreases its outflow to mandibular lymph nodes, and leads to significant differences in CSF accumulation pattern in the spine compared to isoflurane (ISO) alone, while both ISO and Dex do not affect the clearance of tracer-filled CSF into blood circulation. Notably, we confirm the compromised glymphatic function after cerebral ischemia-reperfusion injury, leading to impaired glymphatic influx and reduced glymphatic efflux. This technique has great potential to elucidate the underlying mechanisms between the glymphatic system and central nervous system diseases.
Collapse
Affiliation(s)
- Bin Sun
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Danlan Fang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Wenzhong Li
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, China
| | - Mengfei Li
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, China.
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, China.
| |
Collapse
|
18
|
Campbell E, Figueiro MG. Postoperative cognitive dysfunction: spotlight on light, circadian rhythms, and sleep. Front Neurosci 2024; 18:1390216. [PMID: 38699675 PMCID: PMC11064652 DOI: 10.3389/fnins.2024.1390216] [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: 02/22/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a neurological disorder characterized by the emergence of cognitive impairment after surgery. A growing body of literature suggests that the onset of POCD is closely tied to circadian rhythm disruption (CRD). Circadian rhythms are patterns of behavioral and physiological change that repeat themselves at approximately, but not exactly, every 24 h. They are entrained to the 24 h day by the daily light-dark cycle. Postoperative CRD affects cognitive function likely by disrupting sleep architecture, which in turn provokes a host of pathological processes including neuroinflammation, blood-brain barrier disturbances, and glymphatic pathway dysfunction. Therefore, to address the pathogenesis of POCD it is first necessary to correct the dysregulated circadian rhythms that often occur in surgical patients. This narrative review summarizes the evidence for CRD as a key contributor to POCD and concludes with a brief discussion of how circadian-effective hospital lighting can be employed to re-entrain stable and robust circadian rhythms in surgical patients.
Collapse
Affiliation(s)
| | - Mariana G. Figueiro
- Light and Health Research Center, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
19
|
Antila H, Lilius TO, Palada V, Lohela T, Bell RF, Porkka-Heiskanen T, Kalso E. Effects of commonly used analgesics on sleep architecture-A topical review. Pain 2024; 165:00006396-990000000-00539. [PMID: 38442410 PMCID: PMC11247456 DOI: 10.1097/j.pain.0000000000003201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 03/07/2024]
Affiliation(s)
- Hanna Antila
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Finland
- SleepWell Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Individualized Drug Therapy Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
| | - Tuomas O. Lilius
- Individualized Drug Therapy Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland
- Department of Emergency Medicine and Services, Helsinki University Hospital and University of Helsinki, Finland
| | - Vinko Palada
- SleepWell Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Finland
| | - Terhi Lohela
- Individualized Drug Therapy Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital and University of Helsinki, Finland
| | - Rae F. Bell
- Regional Centre of Excellence in Palliative Care, Haukeland University Hospital, Bergen, Norway
| | | | - Eija Kalso
- SleepWell Research Program Unit, Faculty of Medicine, University of Helsinki, Finland
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Finland
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital and University of Helsinki, Finland
| |
Collapse
|
20
|
Eisen A, Nedergaard M, Gray E, Kiernan MC. The glymphatic system and Amyotrophic lateral sclerosis. Prog Neurobiol 2024; 234:102571. [PMID: 38266701 DOI: 10.1016/j.pneurobio.2024.102571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/18/2023] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
The glymphatic system and the meningeal lymphatic vessels provide a pathway for transport of solutes and clearance of toxic material from the brain. Of specific relevance to ALS, this is applicable for TDP-43 and glutamate, both major elements in disease pathogenesis. Flow is propelled by arterial pulsation, respiration, posture, as well as the positioning and proportion of aquaporin-4 channels (AQP4). Non-REM slow wave sleep is the is key to glymphatic drainage which discontinues during wakefulness. In Parkinson's disease and Alzheimer's disease, sleep impairment is known to predate the development of characteristic clinical features by several years and is associated with progressive accumulation of toxic proteinaceous products. While sleep issues are well described in ALS, consideration of preclinical sleep impairment or the potential of a failing glymphatic system in ALS has rarely been considered. Here we review how the glymphatic system may impact ALS. Preclinical sleep impairment as an unrecognized major risk factor for ALS is considered, while potential therapeutic options to improve glymphatic flow are explored.
Collapse
Affiliation(s)
- Andrew Eisen
- Department of Neurology, University of British Columbia, Vancouver, Canada.
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical School and Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Emma Gray
- Department of Neurology, Royal Prince Alfred Hospital and University of Sydney, NSW 2050, Australia
| | | |
Collapse
|
21
|
Chae J, Choi M, Choi J, Yoo SJ. The nasal lymphatic route of CSF outflow: implications for neurodegenerative disease diagnosis and monitoring. Anim Cells Syst (Seoul) 2024; 28:45-54. [PMID: 38292931 PMCID: PMC10826790 DOI: 10.1080/19768354.2024.2307559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/15/2024] [Indexed: 02/01/2024] Open
Abstract
Cerebrospinal fluid (CSF) plays a crucial role in the brain's lymphatics as it traverses the central nervous system (CNS). Its primary function is to facilitate the outward transport of waste. Among the various CSF outflow pathways, the route through the cribriform plate along the olfactory nerves stands out as the most predominant. This review describes the outflow pathway of CSF into the nasal lymphatics. Additionally, we examine existing studies to describe mutual influences observed between the brain and extracranial regions due to this outflow pathway. Notably, pathological conditions in the CNS often influence CSF outflow, leading to observable changes in extracranial regions. The established connection between the brain and the nose is significant, and our review underscores its potential relevance in monitoring CNS ailments, including neurodegenerative diseases. Considering that aging - the most significant risk factor for the onset of neurodegeneration - is also a principal factor in CSF turnover alterations, we suggest a novel approach to studying neurodegenerative diseases in therapeutic terms.
Collapse
Affiliation(s)
- Jiwon Chae
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Mina Choi
- Keybasic Co., ltd, Seoul, Republic of Korea
| | | | - Seung-Jun Yoo
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Huo QF, Zhu LJ, Guo JW, Jiang YA, Zhao J. Effects of ulinastatin combined with dexmedetomidine on cognitive dysfunction and emergence agitation in elderly patients who underwent total hip arthroplasty. World J Psychiatry 2024; 14:26-35. [PMID: 38327895 PMCID: PMC10845226 DOI: 10.5498/wjp.v14.i1.26] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/11/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND With the continuous growth of the modern elderly population, the risk of fracture increases. Hip fracture is a common type of fracture in older people. Total hip arthroplasty (THA) has significant advantages in relieving chronic pain and promoting the recovery of hip joint function. AIM To investigate the effect of ulinastatin combined with dexmedetomidine (Dex) on the incidences of postoperative cognitive dysfunction (POCD) and emergence agitation in elderly patients who underwent THA. METHODS A total of 397 patients who underwent THA from February 2019 to August 2022. We conducted a three-year retrospective cohort study in Shaanxi Provincial People's Hospital. Comprehensive demographic data were obtained from the electronic medical record system. We collected preoperative, intraoperative, and postoperative data. One hundred twenty-nine patients who were administered Dex during the operation were included in the Dex group. One hundred fifty patients who were intravenously injected with ulinastatin 15 min before anesthesia induction were included in the ulinastatin group. One hundred eighteen patients who were administered ulinastatin combined with Dex during the operation were included in the Dex + ulinastatin group. The patients' perioperative conditions, hemodynamic indexes, postoperative Mini-Mental State Examination (MMSE) scores, Ramsay score, incidence of POCD, and serum inflammatory cytokines were evaluated. RESULTS There was a significant difference in the 24 h visual analogue scale score among the three groups, and the score in the Dex + ulinastatin group was the lowest (P < 0.05). Compared with the Dex and ulinastatin group, the MMSE scores of the Dex + ulinastatin group were significantly increased at 1 and 7 d after the operation (all P < 0.05). Compared with those in the Dex and ulinastatin groups, incidence of POCD, levels of serum inflammatory cytokines in the Dex + ulinastatin group were significantly decreased at 1 and 7 d after the operation (all P < 0.05). The observer's assessment of the alertness/sedation score and Ramsay score of the Dex + ulinastatin group were significantly different from those of the Dex and ulinastatin groups on the first day after the operation (all P < 0.05). CONCLUSION Ulinastatin combined with Dex can prevent the occurrence of POCD and emergence agitation in elderly patients undergoing THA.
Collapse
Affiliation(s)
- Qi-Fan Huo
- Department of Anesthesiology, Shaanxi Provincial People’s Hospital, Xi’an 710068, Shaanxi Province, China
| | - Li-Juan Zhu
- Department of Anesthesiology, Shaanxi Provincial People’s Hospital, Xi’an 710068, Shaanxi Province, China
| | - Jian-Wei Guo
- Department of Anesthesiology, Shaanxi Provincial People’s Hospital, Xi’an 710068, Shaanxi Province, China
| | - Yan-An Jiang
- Department of Anesthesiology, Shaanxi Provincial People’s Hospital, Xi’an 710068, Shaanxi Province, China
| | - Jing Zhao
- Department of Anesthesiology, Shaanxi Provincial People’s Hospital, Xi’an 710068, Shaanxi Province, China
| |
Collapse
|
23
|
Verghese JP, Terry A, de Natale ER, Politis M. Research Evidence of the Role of the Glymphatic System and Its Potential Pharmacological Modulation in Neurodegenerative Diseases. J Clin Med 2022; 11:jcm11236964. [PMID: 36498538 PMCID: PMC9735716 DOI: 10.3390/jcm11236964] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
The glymphatic system is a unique pathway that utilises end-feet Aquaporin 4 (AQP4) channels within perivascular astrocytes, which is believed to cause cerebrospinal fluid (CSF) inflow into perivascular space (PVS), providing nutrients and waste disposal of the brain parenchyma. It is theorised that the bulk flow of CSF within the PVS removes waste products, soluble proteins, and products of metabolic activity, such as amyloid-β (Aβ). In the experimental model, the glymphatic system is selectively active during slow-wave sleep, and its activity is affected by both sleep dysfunction and deprivation. Dysfunction of the glymphatic system has been proposed as a potential key driver of neurodegeneration. This hypothesis is indirectly supported by the close relationship between neurodegenerative diseases and sleep alterations, frequently occurring years before the clinical diagnosis. Therefore, a detailed characterisation of the function of the glymphatic system in human physiology and disease would shed light on its early stage pathophysiology. The study of the glymphatic system is also critical to identifying means for its pharmacological modulation, which may have the potential for disease modification. This review will critically outline the primary evidence from literature about the dysfunction of the glymphatic system in neurodegeneration and discuss the rationale and current knowledge about pharmacological modulation of the glymphatic system in the animal model and its potential clinical applications in human clinical trials.
Collapse
|